Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
INDOLINONES COMPOUNDS AND THEIR USE IN THE TREATMENT OF FIBROTIC DISEASES
Document Type and Number:
WIPO Patent Application WO/2017/109513
Kind Code:
A1
Abstract:
The present invention relates inter alia to a compound of formula (I) Wherein R1, R2, R3 and Z are as defined in the specification and to compositions comprising the same and to the use of the compounds and to compositions of the compounds in treatment, for example in the treatment of fibrotic diseases or interstitial lung diseases, in particular idiopathic pulmonary fibrosis.

Inventors:
WALTERS IAIN (GB)
BIRCH LOUISE (GB)
HILL-COUSINS JOSEPH (GB)
COLLINGWOOD STEPHEN PAUL (GB)
STEVENSON CHRISTOPHER SCOTT (GB)
Application Number:
PCT/GB2016/054069
Publication Date:
June 29, 2017
Filing Date:
December 23, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
RESPIVERT LTD (GB)
International Classes:
C07D403/12; A61K31/404; A61K31/497; A61P11/00; C07C205/57; C07D209/34; C07D413/12
Domestic Patent References:
WO2006067168A12006-06-29
Other References:
GERALD J. ROTH ET AL: "Design, Synthesis, and Evaluation of Indolinones as Inhibitors of the Transforming Growth Factor [beta] Receptor I (TGF[beta]RI)", JOURNAL OF MEDICINAL CHEMISTRY, vol. 53, no. 20, 28 October 2010 (2010-10-28), pages 7287 - 7295, XP055079035, ISSN: 0022-2623, DOI: 10.1021/jm100812a
ROTH GERALD J ET AL: "Design, Synthesis, and Evaluation of Indolinones as Triple Angiokinase Inhibitors and the Discovery of a Highly Specific 6-Methoxycarbonyl-Substituted Indolinone (BIBF 1120)", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 52, no. 14, 1 July 2009 (2009-07-01), pages 4466 - 4480, XP002582395, ISSN: 0022-2623, [retrieved on 20090612], DOI: 10.1021/JM900431G
Attorney, Agent or Firm:
TEUTEN, Andrew et al. (GB)
Download PDF:
Claims:
Claims

A compound of formula (I):

wherein

Ri represents H, Me, Et, CH=CH2, C≡C-H or C≡C-Me;

R2 represents H, d-Cealkyl, d-Cealkoxy, d-Cscycloalkyl, -CH2-(C3-C8cycloalkyl), halogen or cyano;

R3 represents (i)

or (ii)

O

,N— Y2

or (iii)

O-Y3

/ \

, Q

Q represents a heteroatom selected from O, N and S and if N may optionally be substituted with Ci-4alkyl;

Z represents CO or SO2; Yi represents (CH2)n and, except when n represents 0, may optionally be substituted by Me;

Xi represents (CH2)m and, except when m represents 0, may optionally be substituted by Me;

n and m independently represent 0, 1 , 2, 3, 4 or 5;

Y2 represents (CH2)S and may optionally be substituted by Me;

s represents 2, 3, 4, 5 or 6;

Y3 represents (ChbX and may optionally be substituted by Me;

X2 represents (CH2)V and may optionally be substituted by Me;

t and v independently represent 2 or 3 save that t + v = 4 or 5;

R4 represents H, OH, NR6R7 or an aliphatic 4-8 membered heterocycle containing one or more heteroatoms selected from O, N and S, save that when R4 is OH or NR6R7, m is 2, 3, 4 or 5;

R5 represents H, OH, NRsR9 or aliphatic 4-8 membered heterocycle containing one or more heteroatoms selected from O, N and S save that when R5 is OH or NRsR9, n is 2, 3, 4 or 5;

in which the aliphatic heterocycle groups that R4 and R5 may represent may optionally contain a carbonyl or sulphone group and may optionally be substituted by one or more groups selected from -Ci-C4alkyl, Ci-C4hydroxyalkyl-, Ci-C4alkoxy(Ci-C4)alkyl-, -Ci- C alkyleneCONRi0Ri i , CN, OH and N R12R13;

R6, R7, Re, R9 independently represent H, or C1-C4 alkyl optionally substituted by OH, oxo, NRi4Ri5 or -Ci-C4alkoxy; and

R10, R11 , R12 , i3 , i4 and R15 independently represent H or Ci-C4alkyl;

or a pharmaceutically acceptable salt thereof.

2. A compound of formula (I) according to claim 1 wherein Ri represents Me or H.

3. A compound of formula (I) according to claim 2 wherein Ri represents Me. 4. A compound of formula (I) according to any one of claims 1 to 3 wherein R2 represents H, Me or F, especially H.

5. A compound of formula (I) according to any one of claims 1 to 4 wherein Z represents CO.

6. A compound of formula (I) according to any one of claims 1 to 4 wherein Z represents S02.

7. A compound of formula (I) according to any one of claims 1 to 6 wherein R3 is represented by formula (i).

8. A compound of formula (I) according to claim 7 wherein Xi represents (CH2)o, CH2, CH(CH3)CH2 or (CH2)2, especially (CH2)o or CH2, preferably (CH2)0.

9. A compound of formula (I) according to claims 7 or 8 wherein R4 represents H, N- methyl-piperidine or dimethylamine, especially H.

10. A compound of formula (I) according to any one of claims 7 to 9 wherein moiety -Xr R4 represents H, Me, CH2CH3, isopropyl, 1-methyl-piperidin-4-yl or N,N-dimethylamine, especially H or Me, preferably H.

1 1. A compound of formula (I) according to any one of claims 7 to 10 wherein Yi represents (CH2)0, CH2, (CH2)2 or (CH2)3, especially (CH2)2.

12. A compound of formula (I) according to any one of claims 7 to 1 1 wherein R5 represents H, dimethylamine, N-methylethanolamine, N-methyl-piperazine, N-methyl- piperidine, 1 ,2,6-trimethylpiperazine, N-ethyl-piperazine, 3-(N,N-dimethylamine)-pyrrolidine, N-(CH2CH2OH)-piperazine, piperidine, morpholine, 4-hydroxy-piperidine, 4-cyano-piperidine, 2,6-dimethyl-piperidine, N-methoxyethyl-piperazine, 2-methyl-piperazine, N-methyl-2-(N- piperazinyl)acetamide, 4-(N,N-dimethylamine)-piperidine, 4-methoxy-piperidine, S-dioxy- thiomorpholine, N-piperazin-3-one, 2,5-diazabicyclo[2.2.1]heptane, or 3,8- diazabicyclo[3.2.1]octane, 3,6-diazabicyclo[3.1.1]heptanyl, especially dimethylamine or N- methyl-piperazine, preferably dimethylamine.

13. A compound of formula (I) according to any one of claims 7 to 12 wherein moiety -Yi- R5 represents Me, -(CH2)2-dimethylamino, -(CH2)3-dimethylamino, -(CH2)2-(N-methyl)- ethanolamino, -(CH2)2-piperazin-1-yl, -(CH2)2-(4-methyl)-piperazin-1-yl, -(CH2)2-(3-methyl)- piperazin-1-yl, -(CH2)3-(4-methyl)-piperazin-1-yl, -(CH2)2-(1-methyl)-piperidin-4-yl, N-methyl- piperidin-4-yl, -(CH2)2-(4-ethyl)-piperazin-1-yl, -(CH2)2-3-(N,N-dimethylamino)-pyrrolidin-1-yl, -(CH2)2-(4-(CH2CH2OH)-piperazin-1-yl), -(CH2)2-3,4,5-trimethylpiperazin-1-yl, -(CH2)2- piperidin-1-yl, -(CH2)3-piperidin-1-yl, -(CH2)2- morpholin-4-yl,-(CH2)2-(4-hydroxy- piperidin-1- yl), -(CH2)2-(4-cyano-piperidin-1-yl), -(CH2)2-(2,6-dimethyl-piperidin-1-yl), -(CH2)2-(4- methoxyethyl-piperazin-1-yl), -(CH2)2-4-(N,N-dimethylamino-piperidin-1-yl), -(CH2)2-(4- methoxy-piperidin-1-yl), -(CH2)3-(4-methoxy- piperidin-1-yl), -(CH2)2-4-(N-methylacetamido)- piperazin-1-yl)), -(CH2)2-(4-dioxy-thiomorpholin-1-yl), -(CH2)2-(3-oxopiperazin-1-yl), -(CH2)2- (2,5-diazabicyclo[2.2.1]heptan-2-yl), -(CH2)2-(3,8-diazabicyclo[3.2.1]octan-8-yl), (CH2)2-(3,8- diazabicyclo[3.2.1]octan-3-yl), (CH2)2-3,6-diazabicyclo[3.1.1]heptan-3-yl especially -(CH2)2- dimethylamino or -(CH2)2-(4-methyl-piperazin-1-yl), more preferably -(CH2)2-dimethylamino.

14. A compound of formula (I) according to any one of claims 7 to 13 wherein formula (i) represents a moiety in which: (a) -X1-R4 represents H and -Y1-R5 represents -(CH2)2- dimethylamino, -(CH2)3-dimethylamineo, -(CH2)2-(N-methyl)-ethanolamino, -(CH2)2-4-methyl- piperazin-1-yl, -(CH2)3-(4-methyl)-piperazin-1-yl, N-methyl-piperidin-4-yl, -(CH2)2-3,4,5- trimethylpiperazin-1-yl, -(CH2)2-4-ethyl-piperazin-N-yl, -(CH2)2-3-(N,N-dimethylamino)- pyrrolidin-1-yl, -(CH2)2-(4- (CH2CH2OI-l)-piperazin-1-yl), -(CH2)2-piperidin-1-yl, -(CH2)3- piperidin-1-yl, -(CH2)2-morpholin-4-yl, -(CH2)2-(4-hydroxy-piperidin-1-yl), -(CH2)2-4-cyano- piperidin-1-yl, -(CH2)2-(2,6-dimethyl-piperidin-1-yl), -(CH2)2-4-methoxyethyl-piperazin-1-yl, - (CH2)2-4-(N,N-dimethylamino)-piperidin-1-yl, -(CH2)2-4-methoxy-piperidin-1-yl, -(CH2)3-4- methoxy-piperidin-1-yl, -(CH2)2-4-dioxy-thiomorpholin-1-yl, -(CH2)2-(3-oxopiperazin-1-yl), especially -X1-R4 represents H and -Y1-R5 represents -(CH2)2-dimethylamino or -(CH2)2-4- methyl-piperazin-1-yl, more preferably -X1-R4 represents H and -Y1-R5 represents -(CH2)2- dimethylamino; or (b) -X1-R4 represents Me and -Y1-R5 represents Me, -(CH2)2- dimethylamino, -(CH2)2-(N-methyl)-ethanolamino, -(CH2)2-piperazin-1-yl, -(CH2)2-4-methyl- piperazin-1-yl, -(CH2)2-(3-methyl)- piperazin-1-yl, -(CH2)2-(4-(CH2CH2OH)- piperazin-1-yl), - (CH2)2-(4-(N-methylacetamido)-piperazin-1-yl)), -(CH2)2-(2,5-diazabicyclo[2.2.1]heptan-2-yl), -(CH2)2- (3,8-diazabicyclo[3.2.1]octan-8-yl), (CH2)2- (3,8-diazabicyclo[3.2.1]octan-3-yl), (CH2)2-3,6-diazabicyclo[3.1.1]heptan-3-yl, especially -X1-R4 represents Me and -Y1-R5 represents (CH2)2-dimethylamino or -(CH2)2-4-methyl-piperazin-1-yl, more preferably -X1-R4 represents Me and -Y1-R5 represents -(CH2)2-4-methyl-piperazin-1-yl; or (c) -X1-R4 represents 1-methyl-piperidin-4-yl and -Y1-R5 represents Me; or (d) -X1-R4 represents N,N- dimethylamino and -Y1-R5 represents Me; or (e) -X1-R4 represents CH2CH3 and -Y1-R5 represents (CH2)2-piperazin-1-yl or (f) -X1-R4 represents isopropyl and -Y1-R5 represents (CH2)2-piperazin-1-yl, especially moiety (a).

15. A compound of formula (I) according to any one of claims 1 to 6 wherein R3 is represented by formula (ii).

16. A compound of formula (I) according to claim 15 wherein s represents 2.

17. A compound of formula (I) according to any one of claims 1 to 6 wherein R3 is represented by formula (iii).

18. A compound of formula (I) according to claim 17 wherein t represents 2 and v represents 2.

19. A compound of formula (I) according to any one of claims 16 to 18 wherein Q is N or O, especially N.

20. A compound of formula (I) according to claim 1 which is selected from:

(Z)-Methyl 3-(((4-(methoxy(methyl)carbamoyl)phenyl)amino)(phenyl)methylene)-2- oxoindoline-6-carboxylate;

(Z)-Methyl 5-methyl-3-(((4-(/V-(2-(4-methylpiperazin-1- yl)ethoxy)sulfamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate;

(Z)-Methyl 3-(((4-((2-(dimethylamino)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2- oxoindoline-6-carboxylate;

(Z)-Methyl 3-(((4-((2-(4-methylpiperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate;

(Z)-Methyl 3-(((4-((2-(dimethylamino)ethoxy)carbamoyl)-3- methylphenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline-6-carboxylate;

(Z)-Methyl 5-methyl-3-(((4-((2-(4-methylpiperazin-1 - yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate;

(Z)-Methyl 3-(((4-((2-(4-(2-hydroxyethyl)piperazin-1 - yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline-6- carboxylate;

(Z)-Methyl 2-oxo-3-(phenyl((4-((2-(piperidin-1- yl)ethoxy)carbamoyl)phenyl)amino)methylene)indoline-6-carboxylate;

(Z)-Methyl 5-methyl-3-(((4-(methyl(2-(4-(2-(methylamino)-2-oxoethyl)piperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate;

(Z)-Methyl 5-methyl-3-(((4-(methyl(2-(4-(2-(methylamino)-2-oxoethyl)piperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate;

(Z)-Methyl 3-(((4-(/V-methoxy-/\/-methylsulfamoyl)phenyl)amino)(phenyl)methylene)-2- oxoindoline-6-carboxylate;

(Z)-Methyl 3-(((4-((2-morpholinoethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2- oxoindoline-6-carboxylate;

(Z)-Methyl 3-(((4-((2-(dimethylamino)ethoxy)carbamoyl)-3- methylphenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate;

(Z)-Methyl 3-(((4-(methyl(2-(4-methylpiperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate; (Z)-Methyl 3-(((4-(A/-(2-(dimethylamino)ethoxy)sulfamoyl)phenyl)amino)(phenyl)methylene)-

2-oxoindoline-6-carboxylate;

(Z)-Methyl 3-(((3-methyl-4-((2- morpholinoethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate; (Z)-Methyl 3-(((3-methyl-4-((2-(4-methylpiperazin-1 - yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylen)-2-oxoindoline-6-carboxylate;

(Z)-Methyl 3-(((3-methyl-4-(methyl(2-(4-methylpiperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate;

(Z)-Methyl 3-(((4-((3-(dimethylamino)propoxy)carbamoyl)phenyl)amino)(phenyl)methylene)- 2-oxoindoline-6-carboxylate;

(Z)-Methyl 3-(((4-((2-(dimethylamino)ethoxy)carbamoyl)-3- fluorophenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate;

(Z)-Methyl 3-(((4-(methoxy(1 -methylpiperidin-4- yl)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate;

(Z)-Methyl 3-(((4-((2-(dimethylamino)ethoxy)(methyl)carbamoyl)-3- methylphenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate;

(Z)-Methyl 3-(((4-((2-

(dimethylamino)ethyl)(methoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindolin 6-carboxylate;

(Z)-Methyl 3-(((4-(5-methyl-1 ,2,5-oxadiazepane-2- carbonyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate;

(Z)-Methyl 3-(((4-(/V-methyl-/\/-(2-(4-methylpiperazin-1- yl)ethoxy)sulfamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate;

(Z)-Methyl 3-(((4-((2-(dimethylamino)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-5- methyl-2-oxoindoline-6-carboxylate;

(Z)-Methyl 3-(((4-((3-(dimethylamino)propoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-

5-methyl-2-oxoindoline-6-carboxylate;

(Z)-Methyl 5-methyl-3-(((3-methyl-4-((2-(4-methylpiperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate;

(Z)-Methyl 3-(((4-((3-(dimethylamino)propoxy)carbamoyl)-3- methylphenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline-6-carboxylate;

(Z)-Methyl 5-methyl-3-(((4-(((1 -methylpiperidin-4- yl)oxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate;

(Z)-Methyl 5-methyl-3-(((3-methyl-4-(((1 -methylpiperidin-4- yl)oxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate; (Z)-Methyl 3-(((3-fluoro-4-((2-(4-methylpiperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline-6- carboxylate;

(Z)-Methyl 3-(((3-fluoro-4-((2-(4-methylpiperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate; (Z)-Methyl 3-(((4-(methoxy(1 -methylpiperidin-4-yl)carbamoyl)-3- methylphenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline-6-carboxylate;

(Z)-Methyl 3-(((4-(methoxy(1 -methylpiperidin-4- yl)carbamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline-6-carboxylate; (Z)-Methyl 3-(((4-((2-(4-ethylpiperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline-6- carboxylate;

(Z)-Methyl 3-(((4-(1 ,2-oxazetidine-2-carbonyl)phenyl)amino)(phenyl)methylene)-2- oxoindoline-6-carboxylate;

(S,Z)-Methyl 3-(((4-((2-(3-(dimethylamino)pyrrolidin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate; (Z)-Methyl 3-(((4-((2-(4-(2-hydroxyethyl)piperazin-1 - yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate; (Z)-Methyl 3-(((4-((2-(4-hydroxypiperidin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate; (Z)-Methyl 3-(((4-((2-(1 , 1- dioxidothiomorpholino)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindolin carboxylate;

(Z)- Methyl 2-oxo-3-(((4-((2-(3-oxopiperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)indoline-6-carboxylate;

(Z)- Methyl 3-(((4-((2-(4-cyanopiperidin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate; (Z)-Methyl 3-(((4-((2-((2S,6 )-2,6-dimethylpiperidin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate; (Z)-Methyl-3-(((4-((2-((2- hydroxyethyl)(methyl)amino)ethoxy)carbamoyl)phenyl)arnino)(phenyl)methylene)-2- oxoindoline-6-carboxylate;

(Z)-Methyl 3-(((4-((2-(4-(2-methoxyethyl)piperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate; (Z)-Methyl 3-(((4-((2-(4-(dimethylamino)piperidin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate; (Z)-Methyl 3-(((4-(A/-(2-(dimethylamino)ethoxy)sulfamoyl)phenyl)amino)(phenyl)methylene)-

5-methyl-2-oxoindoline-6-carboxylate;

(Z)-Methyl 5-methyl-2-oxo-3-(phenyl((4-((2-(piperidin-1- yl)ethoxy)carbamoyl)phenyl)amino)methylene)indoline-6-carboxylate;

(Z)-Methyl 3-(((4-((2-(4-(dimethylamino)piperidin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline-6- carboxylate;

(S,Z)-Methyl 3-(((4-((2-(3-(dimethylamino)pyrrolidin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline-6- carboxylate;

(Z)-Methyl 3-(((4-((2-(4-methoxypiperidin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline-6- carboxylate;

(Z)-Methyl 5-methyl-2-oxo-3-(phenyl((4-((3-(piperidin-1- yl)propoxy)carbamoyl)phenyl)amino)methylene)indoline-6-carboxylate;

(Z)-Methyl 3-(((4-((2-(4-hydroxypiperidin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline-6- carboxylate;

(Z)-Methyl 3-(((4-((3-(4-methoxypiperidin-1- yl)propoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline-6- carboxylate;

(Z)-Methyl 3-(((4-((2-((2- hydroxyethyl)(methyl)amino)ethoxy)(methyl)carbamoyl)phenyl)amino)(phenyl)methylene)-5- methyl-2-oxoindoline-6-carboxylate;

(Z)-Methyl 5-methyl-3-(((4-(methyl(2-(piperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate;

(Z)-Methyl 3-(((4-((2-(4-(2-hydroxyethyl)piperazin-1 - yl)ethoxy)(methyl)carbamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline-6- carboxylate;

(Z)-Methyl 3-(((4-((2-((1 S,4S)-2,5-diazabicyclo[2.2.1]heptan-2- yl)ethoxy)(methyl)carbamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline-6- carboxylate;

(Z)-Methyl 3-(((4-((2-(3,8-diazabicyclo[3.2.1]octan-8- yl)ethoxy)(methyl)carbamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline-6- carboxylate; (Z)-Methyl 3-(((4-((2-(3,8-diazabicyclo[3.2.1]octan-3- yl)ethoxy)(methyl)carbamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline-6- carboxylate;

(S,Z)-Methyl 5-methyl-3-(((4-(methyl(2-(3-methylpiperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate; (Z)-Methyl 3-(((4-((2-(4-cyanopiperidin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline-6- carboxylate;

(Z)-Methyl 5-methyl-3-(((4-((3-(4-methylpiperazin-1 - yl)propoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate; {R,Z}- Methyl 5-methyl-3-(((4-(methyl(2-(3-methylpiperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate; (Z)-Methyl 3-(((4-((2-(3,6-diazabicyclo[3.1.1 ]heptan-3- yl)ethoxy)(methyl)carbamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline-6- carboxylate;

(Z)-Methyl 5-methyl-2-oxo-3-(phenyl((4-((2-((3 ,5S)-3,4,5-trimethylpiperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)methylene)indoline-6-carboxylate;

(Z)-Methyl 3-(((4-(ethyl(2-(piperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline-6- carboxylate; and

(Z)-Methyl 3-(((4-(isopropyl(2-(piperazin-1 - yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline-6- carboxylate;

{R,Z}- Methyl 5-methyl-3-(((4-(methyl(2-(3-methylpiperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate; (Z)-Methyl 3-(((4-((2-(3,6-diazabicyclo[3.1.1 ]heptan-3- yl)ethoxy)(methyl)carbamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline-6- carboxylate;

(Z)-Methyl 5-methyl-2-oxo-3-(phenyl((4-((2-((3 ,5S)-3,4,5-trimethylpiperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)methylene)indoline-6-carboxylate;

(Z)-Methyl 3-(((4-(ethyl(2-(piperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline-6- carboxylate;

(Z)-Methyl 3-(((4-(isopropyl(2-(piperazin-1 - yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline-6- carboxylate;

and pharmaceutically acceptable salts thereof.

21. A compound of formula (I) according to claim 1 which is selected from: (Z)-Methyl 5-methyl-2-oxo-3-(((4-((2-(3-oxopiperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)indoline-6-carboxylate;

(Z)-Methyl 3-(((4-(methyl(2-(4-(2-(methylamino)-2-oxoethyl)piperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate;

(Z)-Methyl 3-(((4-((2-(4-(2-(methylamino)-2-oxoethyl)piperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate;

(Z)-Methyl 5-methyl-3-(((4-((2-(4-(2-(methylamino)-2-oxoethyl)piperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline-6-carboxylate;

and pharmaceutically acceptable salts thereof.

22. A pharmaceutical composition comprising a compound according to any one of claims 1 to 21 , in combination with one or more pharmaceutically acceptable diluents or carriers.

23. A compound of formula (I) according to any one of claims 1 to 21 for use as a medicament. 24. A compound of formula (I) according to any one of claims 1 to 21 or a composition according to claim 22 for use in the treatment of fibrotic diseases or interstitial lung diseases e.g. selected from IPF, giant cell interstitial pneumonia, sarcoidosis, cystic fibrosis, respiratory distress syndrome, drug-induced lung fibrosis, granulomatosis, silicosis, asbestosis, systemic scleroderma, the virally induced hepatic cirrhosis selected from hepatitis C induced hepatic cirrhosis, or diseases of the skin with a fibrotic component e.g. selected from scleroderma, sarcoidosis and systemic lupus erythematosus, and especially IPF, or for use in the treatment of diseases characterized by hyperproliferation of cells, for example, cancer, particularly lung cancer or for use in the treatment of respiratory disorders including COPD (including chronic bronchitis and emphysema), asthma, paediatric asthma, allergic rhinitis, rhinitis, sinusitis, especially asthma, chronic bronchitis and COPD.

25. A compound of formula (I) according to any one of claims 1 to 21 or a composition according to claim 22 for use in the treatment of fibrotic diseases such as lung fibrosis associated with rheumatoid arthritis, respiratory distress syndrome including acute respiratory distress syndrome, acute lung injury, radiation induced lung fibrosis or pneumonitis, chronic hypersensitivity pneumonitis, systemic sclerosis, Sjogren's syndrome, interstitial lung diseases, pulmonary arterial hypertension (PAH), including the vascular component of PAH, or diseases of the skin with a fibrotic component e.g. selected from hypertrophic scarring and keloids, or eye diseases where fibrosis is a component including glaucoma, age related macular degeneration, diabetic macular edema, dry eye disease and diabetic retinopathy, or fibrosis in the gut e.g. associated with inflammatory bowel disease.

26. Use of a compound of formula (I) according to any one of claims 1 to 21 or a composition according to claim 22 for the manufacture of a medicament for the treatment of fibrotic diseases or interstitial lung diseases selected from IPF, giant cell interstitial pneumonia, sarcoidosis, cystic fibrosis, respiratory distress syndrome, drug-induced lung fibrosis, granulomatosis, silicosis, asbestosis, systemic scleroderma, the virally induced hepatic cirrhosis selected from hepatitis C induced hepatic cirrhosis, or diseases of the skin with a fibrotic component e.g. selected from scleroderma, sarcoidosis and systemic lupus erythematosus, and especially IPF, or for the treatment of diseases characterized by hyperproliferation of cells, for example, cancer, particularly lung cancer or for the treatment of respiratory disorders including COPD (including chronic bronchitis and emphysema), asthma, paediatric asthma, allergic rhinitis, rhinitis, sinusitis, especially asthma, chronic bronchitis and COPD.

27. Use of a compound of formula (I) according to any one of claims 1 to 21 or a composition according to claim 22 for the manufacture of a medicament for the treatment of fibrotic diseases such as lung fibrosis associated with rheumatoid arthritis, respiratory distress syndrome including acute respiratory distress syndrome, acute lung injury, radiation induced lung fibrosis or pneumonitis, chronic hypersensitivity pneumonitis, systemic sclerosis, Sjogren's syndrome, interstitial lung diseases, pulmonary arterial hypertension (PAH), including the vascular component of PAH, or diseases of the skin with a fibrotic component e.g. selected from hypertrophic scarring and keloids, or eye diseases where fibrosis is a component including glaucoma, age related macular degeneration, diabetic macular edema, dry eye disease and diabetic retinopathy, or fibrosis in the gut e.g.

associated with inflammatory bowel disease.

28. A compound of formula (I) according to any one of claims 1 to 21 or a composition according to claim 22 in combination with nintedanib or pirfenidone for use in the treatment of fibrotic diseases or interstitial lung diseases selected from IPF, giant cell interstitial pneumonia, sarcoidosis, cystic fibrosis, respiratory distress syndrome, drug-induced lung fibrosis, granulomatosis, silicosis, asbestosis, systemic scleroderma, the virally induced hepatic cirrhosis selected from hepatitis C induced hepatic cirrhosis, or diseases of the skin with a fibrotic component e.g. selected from scleroderma, sarcoidosis and systemic lupus erythematosus, and especially IPF, or for use in the treatment of diseases characterized by hyperproliferation of cells, for example, cancer, particularly lung cancer or for use in the treatment of respiratory disorders including COPD (including chronic bronchitis and emphysema), asthma, paediatric asthma, allergic rhinitis, rhinitis, sinusitis, especially asthma, chronic bronchitis and COPD.

29. A compound of formula (I) according to any one of claims 1 to 21 or a composition according to claim 22 in combination with nintedanib or pirfenidone for use in the treatment of fibrotic diseases such as lung fibrosis associated with rheumatoid arthritis, respiratory distress syndrome including acute respiratory distress syndrome, acute lung injury, radiation induced lung fibrosis or pneumonitis, chronic hypersensitivity pneumonitis, systemic sclerosis, Sjogren's syndrome, interstitial lung diseases, pulmonary arterial hypertension (PAH), including the vascular component of PAH, or diseases of the skin with a fibrotic component e.g. selected from hypertrophic scarring and keloids, or eye diseases where fibrosis is a component including glaucoma, age related macular degeneration, diabetic macular edema, dry eye disease and diabetic retinopathy, or fibrosis in the gut e.g.

associated with inflammatory bowel disease.

30. A method of treatment of a fibrotic disease or interstitial lung disease e.g. selected from IPF, giant cell interstitial pneumonia, sarcoidosis, cystic fibrosis, respiratory distress syndrome, drug-induced lung fibrosis, granulomatosis, silicosis, asbestosis, systemic scleroderma, the virally induced hepatic cirrhosis selected from hepatitis C induced hepatic cirrhosis, or diseases of the skin with a fibrotic component e.g. selected from scleroderma, sarcoidosis and systemic lupus erythematosus and especially IPF, or a method of treatment of diseases characterized by hyperproliferation of cells, for example, cancer, particularly lung cancer or a method of treatment of respiratory disorders including COPD (including chronic bronchitis and emphysema), asthma, paediatric asthma, allergic rhinitis, rhinitis, sinusitis, especially asthma, chronic bronchitis and COPD, which comprises administering to a subject an effective amount of a compound of formula (I) according to any one of claims 1 to 21 or a pharmaceutical composition according to claim 22.

31. A method of treatment of a fibrotic disease such as lung fibrosis associated with rheumatoid arthritis, respiratory distress syndrome including acute respiratory distress syndrome, acute lung injury, radiation induced lung fibrosis or pneumonitis, chronic hypersensitivity pneumonitis, systemic sclerosis, Sjogren's syndrome, interstitial lung diseases, pulmonary arterial hypertension (PAH), including the vascular component of PAH, or diseases of the skin with a fibrotic component e.g. selected from hypertrophic scarring and keloids, or eye diseases where fibrosis is a component including glaucoma, age related macular degeneration, diabetic macular edema, dry eye disease and diabetic retinopathy, or fibrosis in the gut e.g. associated with inflammatory bowel disease which comprises administering to a subject an effective amount of a compound of formula (I) according to any one of claims 1 to 21 or a pharmaceutical composition according to claim 22.

32. A compound of formula (II):

wherein

Ri represents Me, Et, CH=CH2, C≡C-H or C≡C-Me, especially Me

or a protected derivative thereof or a salt thereof.

33. A compound of formula (VI):

wherein

Ri represents Et, CH=CH2, C≡C-H or C≡C-Me,

or a protected derivative thereof or a salt thereof.

34. A compound of formula (VII):

wherein

Ri represents Me, Et, CH=CH2, C≡C-H or C≡C-Me, especially Me

or a protected derivative thereof or a salt thereof.

35. A compound of formula (IX):

wherein

Ri represents Me, Et, CH=CH2, C≡C-H or C≡C-Me, especially Me; and

R2 represents H, Ci-CealkyI, Ci-Cealkoxy, Cs-CscycloalkyI, -CH2-(C3-C8cycloalkyl), halogen or cyano;

or a protected derivative thereof or a salt thereof.

36. A compound of formula (XI):

wherein

Ri represents Me, Et, CH=CH2, C≡C-H or C≡C-Me, especially Me; and

R2 represents H, Ci-CealkyI, Ci-Cealkoxy, Cs-CscycloalkyI, -CH2-(C3-C8cycloalkyl), halogen or cyano;

or a protected derivative thereof or a salt thereof.

37. A compound of formula (XIII):

wherein

Ri represents Me, Et, CH=CH2, C≡C-H or C≡C-Me, especially Me; and

R2 represents H, Ci-CealkyI, Ci-Cealkoxy, Cs-CscycloalkyI, -CH2-(C3-C8cycloalkyl), halogen or cyano;

or a protected derivative thereof or a salt thereof.

38. A compound of formula (XVI):

wherein

Z represents CO or SO2; and

Ri represents Me, Et, CH=CH2, C≡C-H or C≡C-Me; and

R2 represents H, Ci-CealkyI, Ci-Cealkoxy, Cs-CscycloalkyI, -CH2-(C3-C8cycloalkyl), halogen or cyano;

or a protected derivative thereof or a salt thereof.

39. A compound of formula (XVII): OH

wherein

Z represents CO or SO2; and

Ri represents Me, Et, CH=CH2, C≡C-H or C≡C-Me; and

R2 represents H, d-Cealkyl, d-Cealkoxy, d-dcycloalkyl, -CH2-(C3-C8cycloalkyl), halogen or cyano;

or a protected derivative thereof or a salt thereof.

40. A process for preparing a compound of formula (I) or a pharmaceutically acceptable salt thereof according to any one of claims 1 to 21 which comprises reacting a compound of formula

(II):

wherein

Ri is as defined in any one of claims 1 to 21 ;

or a salt thereof;

with a compound of formula (III):

wherein

R2, R3 and Z are as defined in any one of claims 1 to 21 ;

or a salt thereof.

42. A process for preparing a compound of formula (I), wherein Z is CO and R3 is formula (i), or a pharmaceutically acceptable salt thereof according to any one of claims 1 to 21 which comprises reacting a compound of formula (IX):

wherein

Ri and R2 are as defined in any one of claims 1 to 21 ;

or a protected derivative thereof or a salt thereof; with a compound of formula (X):

(X) o-R'

H N wherein

R' represents Y1-R5 as defined in any one of claims 1 to 21 ;

R" represents X1-R4 as defined in any one of claims 1 to 21 ;

or a protected derivative thereof or a salt thereof.

43. A process for preparing a compound of formula (I), wherein R3 is formula (i) and X1-R4 is Me, or a pharmaceutically acceptable salt thereof according to any one of claims 1 to 21 which comprises reacting a

wherein

Ri and R2 are as defined in any one of claims 1 to 21 ;

or a protected derivative thereof or a salt thereof; with a compound of formula (XIV):

wherein

Rs and Rg are as defined in any one of claims 1 to 21 or NRsRg represents an aliphatic 4-8 membered heterocycle containing one or more heteroatoms selected from O, N and S, as defined in any one of claims 1 to 21 ;

or a protected derivative thereof or a salt thereof.

Description:
INDOLINONES COMPOUNDS AND THEIR USE IN THE TREATMENT OF FIBROTIC DISEASES

Field of the invention

The invention relates inter alia to novel compounds which inhibit protein kinases and to their use in therapy, particularly for the treatment of fibrotic diseases or interstitial lung diseases, especially Idiopathic Pulmonary Fibrosis and respiratory diseases. The invention also extends to pharmaceutical compositions comprising the compounds.

Background of the invention

Interstitial lung diseases (ILDs) are characterized by scarring of the lung that leads to lung dysfunction, which can eventually lead to respiratory failure. There are many ILDs with no known cause, which are termed idiopathic. Idiopathic Pulmonary Fibrosis (IPF) is the most common type of ILD. IPF affects about 170,000 people in Europe and 130,000 people in the United States, with approximately 48,000 new cases diagnosed each year in the US alone and 40,000 people dying in the US annually. IPF mortality rates are very high with median survival of 3-5 years from diagnosis and reported 5-year survival rates of less than 30%, on a par with the most lethal cancers. Until recently few treatment options other than lung transplantation have been shown to be effective and treatment for most patients has been symptom control and palliative care.

IPF is a chronic and fatal disease primarily characterised by a progressive decline in lung function caused by the scarring of lung tissue which results in worsening dyspnea. Vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF) and platelet derived growth factor (PDGF) are known potent mitogens for fibroblast cells, which then replace the normal tissue in lungs when fibrosis occurs. In ILDs, the evidence for a pathogenic role for PDGF, VEGF, and FGF has been demonstrated clinically. The primary site affected is the interstitium, the tissue between the air sacs in the lung, but it does also affect the airspaces, peripheral airways and vessels. The disease process is believed to be initiated by a series of microinjuries to the alveolar epithelium in the lung. After the injury, increased vascular permeability leads to clot formation and resident epithelial cells proliferate in an attempt to replace those cells that died as a result of the injury. This process triggers the release of a variety of growth factors (eg, PDGF, VEGF, FGF, and transforming growth factor β (TGFp)), leading to the aberrant activation of the epithelial cells, abnormal vascular remodelling, and most notably, the proliferation and migration of fibroblasts into the lung. Growth factors also induce resident cells to transform into myofibroblasts, which together with fibroblasts organize into foci (King TE Jr, et al., Lancet, 2011 , 3;378(9807): 1949-61 ; Selman M, et al., Ann Intern Med., 2001 , 16; 134(2): 136-51). These cellular changes result in the disruption of the basement membrane and excessive accumulation of extracellular matrix proteins in the interstitial space. The result is the eventual destruction of the normal architecture of the alveolar capillary unit and lung scarring. The pathologies that define the usual interstitial pattern (UIP) of fibrosis characteristic of IPF are a heterogeneous pattern of alternating areas of normal lung, interstitial inflammation, dense fibrosis, fibroblastic foci, and honeycombing, especially in the subpleural area of the lung (Du Bois RM., Nat Rev Drug Discov., 2010, 9(2): 129-40; Selman M, et al., Ann Intern Med., 2001 , 16; 134(2): 136-51 ; King TE Jr, et al., Lancet, 2011 , 3;378(9807):1949-61). The loss of normal architecture and scarring of the interstitial space leads to a significant decline in gas exchange capacity leading to development of the classical symptoms of the disease namely dyspnea, chronic cough, inspiratory crackles on auscultation, and abnormal spirometry (Castriotta RJ, et al., Chest, 2010, 138(3):693-703). While the disease course is heterogeneous, the median survival is approximately 3-5 years and the most common cause of death is respiratory failure due to the progressive pathologies that disrupt normal lung functioning and gas-exchange.

To achieve better tolerability and also better efficacy in treatment of lung disorders it can be advantageous to deliver a drug directly to the site of action in the lung. This enables higher concentrations of drug to be achieved at the site of action resulting in a lower overall dose consequently lowering systemic side effects.

Nintedanib, a protein kinase inhibitor, was approved by the FDA in 2014 for treatment of IPF by oral administration. However, it is associated with significant systemic adverse events, including abdominal pain, vomiting and diarrhoea. WO2006/067165 teaches that inhibitors of VEGFR, FGFR and PDGFR, such as nintedanib, may be expected to be useful in treatment of fibrotic diseases, such as IPF. Fehrenbach. H., et al., Virchows Arch., 1999, 435(1):20-31 discloses that VEGFR is linked to the cause of pulmonary fibrosis. Lindroos. P., Am J Physio Lung Cell Mol Physiol., 2001 , 280:L354-L362 teaches that the upregulation of PDGF receptor is a mechanism of myofibroblast hyperplasia during pulmonary fibrosis. WO01/27081 shows that compounds that have inhibiting effects on kinases, including VEGFR, PDGFR and FGFR, are suitable for treating fibrotic diseases and discloses a series of 6-position substituted indolinones. Similarly, WO2006/067165 and WO2006/067168 also disclose 6-position substituted indolinones for use as medicaments for the treatment or prevention of fibrotic diseases. There remains a need in the art to develop further compounds, especially compounds that are better tolerated than nintedanib, for treating fibrotic diseases and interstitial lung diseases, such as IPF. Desirably such compounds would have low dose, long duration of action suitable for once, twice or three time daily dosing and good efficacy and tolerability when delivered topically to the lung. The compounds of formula (I) described herein address this issue.

Summary of the invention

According to the invention, there is provided a compound of formula (I):

wherein

Ri represents H, Me, Et, CH=CH 2 , C≡C-H or C≡C-Me;

R2 represents H, C1-C6 alkyl, C1-C6 alkoxy, C3-Cscycloalkyl, -CH2-(C3-C8cycloalkyl), halogen or cyano;

R3 represents (i)

or (ii)

o

N— Y 2

or (iii) Q represents a heteroatom selected from O, N and S and if N may optionally be substituted with Ci-4alkyl;

Z represents CO or SO2;

Yi represents (CH2)n and, except when n represents 0, may optionally be substituted by Me;

Xi represents (CH2) m and, except when m represents 0, may optionally be substituted by Me;

n and m independently represent 0, 1 , 2, 3, 4 or 5;

Y2 represents (CH2) S and may optionally be substituted by Me;

s represents 2, 3, 4, 5 or 6;

Y3 represents (CH2X and may optionally be substituted by Me;

X2 represents (CH2) V and may optionally be substituted by Me;

t and v independently represent 2 or 3 save that t + v = 4 or 5;

R 4 represents H, OH, NR6R7 or an aliphatic 4-8 membered heterocycle containing one or more heteroatoms selected from O, N and S, save that when R 4 is OH or NR6R7, m is 2, 3, 4 or 5;

R5 represents H, OH, NRsR9 or aliphatic 4-8 membered heterocycle containing one or more heteroatoms selected from O, N and S save that when R5 is OH or NRsR9, n is 2, 3, 4 or 5;

in which the aliphatic heterocycle groups that R 4 and R5 may represent may optionally contain a carbonyl or sulphone group and may optionally be substituted by one or more groups selected from -Ci-C 4 alkyl, Ci-C 4 hydroxyalkyl-, Ci-C 4 alkoxy(Ci-C 4 )alkyl-, -Ci- C 4 alkyleneCONRi 0 Ri i , CN, OH and N R12R13;

R6, R7, Re, R9 independently represent H, or C1-C4 alkyl optionally substituted by OH, oxo, NRi 4 Ri5 or -Ci-C 4 alkoxy; and

R10, R11 , R12 , i3 , i4 and R15 independently represent H or Ci-C 4 alkyl;

or a pharmaceutically acceptable salt thereof (hereinafter "compounds of the invention" or "a compound of the invention"). Brief Description of the Figures

Figure 1 : shows the artificial membrane permeability of representative examples of the invention and nintedanib (see results of PAMPA permeability assay and Table 8: average values were used for compounds where the experiment was repeated)

Figure 2: shows the total lung exposure following intravenous and intra-tracheal administration of examples of the invention or nintedanib in rats (see results of pharmacokinetic measurements in rodents) Figure 3: shows the total lung exposure following intravenous and intra-tracheal administration of examples of the invention or nintedanib in rats (see results of pharmacokinetic measurements in rodents)

Figure 4: shows the total lung exposure following intravenous and intra-nasal administration of examples of the invention or nintedanib in mice (see results of pharmacokinetic measurements in rodents)

Detailed description of the invention Alkyl groups may be branched or straight chain. Ci-salkyl groups may for example represent Ci-6alkyl, or Ci-3alkyl. Exemplary alkyl groups include methyl, ethyl, n-propyl, i-propyl, n-butyl, t-butyl and ChbCHMea. In one embodiment alkyl refers to straight chain alkyl. Alkylene is to be construed in the same way as alkyl except that it is a divalent group. Alkoxy as used herein means -Oalkyl and includes straight or branched chain alkoxy, for example methoxy, ethoxy, propoxy, butoxy.

Hydroxyalkyl means alkyl with a hydroxyl substituent in any position. Examples include hydroxymethyl, 2-hydroxyethyl, 3-hydroxy-n-propyl and 4-hydroxy-n-butyl.

Halogens may suitably be Br, CI or F, especially CI or F, particularly F.

Examples of aliphatic 4-8 membered heterocyclic rings containing one or more heteroatoms selected from O, S and N include azetidine, pyrrolidine, piperidine, piperazine, morpholine, dioxane, tetrahydrofuran and thiomorpholine. Suitably the heterocyclic ring incudes 1 or 2, especially 1 heteroatom. Such rings may contain a carbonyl or sulphone group and examples include pyrrolidinone or piperidinone.

C3-Cscycloalkyl refers to an aliphatic carbocyclic ring containing typically 3 to 8 ring members with optional branching and containing 3 to 8 carbon atoms in total. Examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, methylcyclohexyl, cycloheptyl and cyclooctyl.

Aliphatic 4-8 membered heterocyclic rings may optionally be substituted. In one embodiment the rings are not substituted. In another embodiment the ring bears one substituent. A substituent may be on a carbon or a nitrogen atom. Examples of substituted heterocyclic rings include N-methyl-piperazine, N-methyl-piperidine, N-ethyl-piperazine, 3-(N,N-dimethylamine)- pyrrolidine, N-(CH2CH20H)-piperazine, 4-hydroxy-piperidine, 4-cyano-piperidine, 2,6- dimethyl-piperidine, N-methoxyethyl-piperazine, 3-(N,N-dimethylamine)-piperidine and 4- methoxy-piperidine.

In one embodiment there is provided a pharmaceutically acceptable salt of the compound of the invention.

The compounds of the disclosure include those where the atom specified is a naturally occurring or non-naturally occurring isotope. In one embodiment the isotope is a stable isotope. Thus the compounds of the disclosure include, for example those containing one or more deuterium atoms in place of hydrogen atoms and the like.

The disclosure also extends to all polymorphic forms of the compounds herein defined including salts thereof. The disclosure also extends to all solvates of the compounds herein defined. Examples of solvates include hydrates.

Suitably Ri represents Me or H, especially Me. Suitably R2 represents H, d-Cealkyl, d-Cealkoxy, C3-C6cycloalkyl, halogen or cyano, more suitably H, Ci-C4alkyl or halogen,, yet more suitably H, Me or halogen, most suitably H, Me or F, especially H.

In a preferred embodiment Z represents CO. In an alternative embodiment Z is SO2.

In one preferred embodiment R3 is formula (i).

Suitably either R 4 represents NR6R7 or an aliphatic 4-8 membered heterocycle containing one or more heteroatoms selected from O, N and S, save that when R 4 is NR6R7, m is 2, 3, 4 or 5 and R5 represents H or OH save that when R5 is OH, n is 2, 3, 4 or 5; or

R5 represents NRsR9 or aliphatic 4-8 membered heterocycle containing one or more heteroatoms selected from O, N and S save that when R5 is NRsR9, n is 2, 3, 4 or 5 and R 4 represents H or OH save that when R 4 is OH, m is 2, 3, 4 or 5. Suitably Xi represents (CH2) m .

Suitably Xi represents (CH 2 )o, CH 2 , CH(CH 3 )CH 2 or (CH 2 ) 2 , especially (CH 2 ) 0 or CH 2 , preferably (CH 2 )o. Suitably R 4 represents H, N-methyl-piperidine or dimethylamine, especially H.

Suitably moiety -Xi-R 4 represents H, Me, CH2CH3, isopropyl, 1-methyl-piperidin-4-yl or N,N- dimethylamine, especially H or Me, preferably H.

Suitably Yi represents (CH2)n.

Suitably Y^ represents (CH 2 )o, CH 2 , (CH 2 ) 2 or (CH 2 )3, especially (CH 2 ) 2 .

Suitably R5 represents H, dimethylamine, N-methylethanolamine, N-methyl-piperazine, N- methyl-piperidine, 1 ,2,6-trimethylpiperazine, N-ethyl-piperazine, 3-(N,N-dimethylamine)- pyrrolidine, N-(CH2CH20H)-piperazine, piperidine, morpholine, 4-hydroxy-piperidine, 4-cyano- piperidine, 2,6-dimethyl-piperidine, N-methoxyethyl-piperazine, 2-methyl-piperazine, N- methyl-2-(N-piperazinyl)acetamide, 4-(N,N-dimethylamine)-piperidine, 4-methoxy-piperidine, S-dioxy-thiomorpholine, N-piperazin-3-one, 2,5-diazabicyclo[2.2.1]heptane, or 3,8- diazabicyclo[3.2.1]octane, 3,6-diazabicyclo[3.1.1]heptanyl especially dimethylamine or N- methyl-piperazine, preferably dimethylamine.

Suitably R5 does not represent H when n represents 0.

Suitably moiety -Y1-R5 represents Me, -(CH2)2-dimethylamino, -(CH2)3-dimethylamino, -(CH2)2- (N-methyl)-ethanolamino, -(CH2)2-piperazin-1-yl, -(CH2)2-(4-methyl)-piperazin-1-yl, -(CH2)2-(3- methyl)-piperazin-1-yl, -(CH2)3-(4-methyl)-piperazin-1-yl, -(CH2)2-(1-methyl)-piperidin-4-yl, N- methyl-piperidin-4-yl, -(CH2)2-(4-ethyl)-piperazin-1-yl, -(CH2)2-3-(N,N-dimethylamino)- pyrrolidin-1-yl, -(CH2)2-(4-(CH2CH 2 OH)-piperazin-1-yl), -(CH 2 )2-3,4,5-trimethylpiperazin-1-yl, - (CH2)2- piperidin-1-yl, -(CH2)3-piperidin-1-yl, -(CH2)2- morpholin-4-yl,-(CH2)2-(4-hydroxy- piperidin-1-yl), -(CH2)2-(4-cyano-piperidin-1-yl), -(CH2)2-(2,6-dimethyl-piperidin-1-yl), -(CH2)2- (4-methoxyethyl-piperazin-1-yl), -(CH2)2-4-(N,N-dimethylamino-piperidin-1-yl), -(CH2)2-(4- methoxy-piperidin-1-yl), -(CH2)3-(4-methoxy- piperidin-1-yl), -(CH2)2-4-(N-methylacetamido)- piperazin-1-yl)), -(CH2)2-(4-dioxy-thiomorpholin-1-yl), -(CH2)2-(3-oxopiperazin-1-yl), -(CH2)2- (2,5-diazabicyclo[2.2.1]heptan-2-yl), -(CH 2 ) 2 -(3,8-diazabicyclo[3.2.1]octan-8-yl), (CH 2 ) 2 -(3,8- diazabicyclo[3.2.1]octan-3-yl), (CH2)2-3,6-diazabicyclo[3.1.1]heptan-3-yl especially -(CH2)2- dimethylamine or -(CH2)2-(4-methyl-piperazin-1-yl), more preferably -(CH2)2-dimethylamino.

Suitably formula (i) represents a moiety in which: (a) -Xi-R 4 represents H and -Y1-R5 represents -(CH2)2-dimethylamino, -(CH2)3-dimethylamineo, -(CH2)2-(N-methyl)-ethanolamino, -(CH2)2-4-methyl-piperazin-1-yl, -(CH2)3-(4-methyl)-piperazin-1-yl, N-methyl-piperidin-4-yl, - (CH 2 )2-3,4,5-trimethylpiperazin-1-yl, -(CH 2 ) 2 -4-ethyl-piperazin-N-yl, -(CH 2 ) 2 -3-(N,N- dimethylamino)-pyrrolidin-1-yl, -(CH2)2-(4- (CH2CH20H)-piperazin-1-yl), -(CH2)2-piperidin-1-yl, -(CH2)3-piperidin-1-yl, -(CH2)2-morpholin-4-yl, -(CH2)2-(4-hydroxy-piperidin-1-yl), -(CH2)2-4- cyano-piperidin-1-yl, -(CH2)2-(2,6-dimethyl-piperidin-1-yl), -(CH2)2-4-methoxyethyl-piperazin- 1-yl, -(CH2)2-4-(N,N-dimethylamino)-piperidin-1-yl, -(CH2)2-4-methoxy-piperidin-1-yl, -(CH2)3- 4-methoxy-piperidin-1-yl, -(CH2)2-4-dioxy-thiomorpholin-1-yl, -(CH2)2-(3-oxopiperazin-1-yl), especially -X1-R4 represents H and -Y1-R5 represents -(CH2)2-dimethylamino or -(CH2)2-4- methyl-piperazin-1-yl, more preferably -X1-R4 represents H and -Y1-R5 represents -(CH2)2- dimethylamino; or (b) -X1-R4 represents Me and -Y1-R5 represents Me, -(CH2)2-dimethylamino, -(CH2)2-(N-methyl)-ethanolamino, -(CH2)2-piperazin-1-yl, -(CH2)2-4-methyl-piperazin-1-yl, - (CH 2 )2-(3-methyl)- piperazin-1-yl, -(CH 2 )2-(4-(CH 2 CH 2 OH)- piperazin-1-yl), -(CH 2 ) 2 -(4-(N- methylacetamido)-piperazin-1-yl)), -(CH 2 )2-(2,5-diazabicyclo[2.2.1]heptan-2-yl), -(CH 2 )2- (3,8- diazabicyclo[3.2.1]octan-8-yl), (CH 2 )2- (3,8-diazabicyclo[3.2.1]octan-3-yl), (CH 2 )2-3,6- diazabicyclo[3.1.1]heptan-3-yl, especially -X1-R4 represents Me and -Y1-R5 represents (CH2)2- dimethylamino or -(CH2)2-4-methyl-piperazin-1-yl, more preferably -X1-R4 represents Me and - Y1-R5 represents -(CH2)2-4-methyl-piperazin-1-yl; or (c) -X1-R4 represents 1-methyl-piperidin- 4-yl and -Y1-R5 represents Me; or (d) -X1-R4 represents Ν,Ν-dimethylamino and -Y1-R5 represents Me; or (e) -X1-R4 represents CH2CH3 and -Y1-R5 represents (CH2)2-piperazin-1-yl or (f) -X1-R4 represents isopropyl and -Y1-R5 represents (CH2)2-piperazin-1-yl. The moiety of formula (i) is preferably represented by (a).

In one embodiment R3 is formula (ii).

Suitably Y2 represents (Chbjs.

Suitably s is 2, 3, or 4, more suitably 2 or 3 especially 2.

Suitably formula (ii) represents 1 ,2-oxazetidine.

In one embodiment R3 is formula (iii).

Suitably Y3 represents (CH2)t.

Suitably X2 represents (CH2) V .

Suitably t is 2 and v is 2.

Suitably Q is N or O, especially N . When Q represents N it may suitably be substituted with methyl.

In an embodiment formula (iii) represents 5-methyl-[1 ,2,5]-oxadiazepane.

Suitably Re, R7, Re, R9 independently represent H, or C1-C4 alkyl optionally substituted by OH,

Suitably R6 and R 7 independently represent Me.

Suitably Rs represents Me and Rg represents Me or CH2CH2OH , especially Me. Suitably R10 represents H and Rn represents Me. Suitably R12 and R13 independently represent Me.

Suitably R14 and R15 are independently selected from H and Me.

The compounds of formula (I) may conveniently be prepared by a process comprising reacting a compound of formula (II), in whi ch as -OC1-C4 alkyl, e.g. Oethyl:

or a protected derivative thereof with a compound of formula (III):

Typically, compounds of formulae (II) and (III) may be reacted in the presence of a solvent, such as DMF, and heated to about 80 °C for approximately 18 hours. Following this step, a deprotection step is performed to remove the protecting group, acetyl. To achieve this, the reaction mixture may be cooled to room temperature and a nucleophile, such as piperidine, added and stirred for 1 to 24 hours.

Compounds of formula (II) in which L represents -Oethyl may be prepared by reacting a compound of formula (IV):

or a protected derivative thereof with a compound of formula (V):

Typically compounds of formulae (IV) and (V) may be reacted in the presence of acetic anhydride at a temperature of about 110 °C for approximately 4 hours. Other compounds of formula (II) may be prepared in an analogous manner. Compounds of formula (IV) may be prepared by reacting a compound of formula (VI):

with acetic anhydride. Typically the reaction is performed at about 1 10 °C. Alternatively compounds of formula (II) in which L represents -Oethyl may be prepared directly from compounds of formula (VI) by treatment with a compound of formula (V) in the presence of acetic anhydride at a temperature of about 110 °C for approximately 4 hours.

Compounds of formula (VI) may be prepared by reducing the -NO2 group of a compound of formula (VII) to an -NH2 group:

followed by an amide forming cyclisation, which is a well-known procedure in the field.

Reduction conditions may typically include use of hb-Pd/C at room temperature, 5 bar pressure for about 36 hours in a solvent, such as acetic acid, which is a well-known procedure in the art. Compounds of formula (VII) may be prepared by reacting a compound of formula (VIII):

with methylchloroacetate. Typically the reaction occurs in the presence of a polar organic solvent, such a DMF, and a base, such as KO'Bu, under a nitrogen atmosphere between approximately -20 to -10 °C.

Alternatively compounds of formula (la), which are compounds of formula (I) in which Z is CO and R3 is formula (i) or (iii), may be prepared by reacting a compound of formula (IX):

or a protected derivative thereof with a compound of formula (X):

(X) - R '

in which R' may represent Y1-R5 and R" may represent X1-R4, or a protected derivative thereof, or R' and R" are joined together with the N and O atoms to make the heterocyclic ring of choice from the examples listed. The compounds may typically be reacted for about 2 to 18 hours at room temperature in the presence of a coupling agent, such as HATU and a base, such as Hunig's base (DIPEA), in a polar organic solvent such as DMF, although other polar organic solvents can be used. This process may be followed, where appropriate, by deprotection.

Compounds of formula (IX) may be prepared by deprotection of a compound of formula (XI):

Deprotection may be achieved using standard reagents in the art, such as TFA, and the compounds are typically stirred at room temperature for about 16 hours in a solvent, such as DCM.

Compounds of formula (XI) may be prepared by reacting a compound of formula (II) with a compound of formula (XII):

The compounds may be reacted in the presence of DMF for about 18 hours at 100 °C.

Alternatively, compounds of formula (lb) which are compounds of formula (I) in which R3 is formula (i) and X1-R4 represents H. NRsRg represents NRsR9 as previously defined, or an aliphatic 4-8 membered heterocycle containing one or more heteroatoms selected from O, N and S, as previously defined:

may be prepared by reacting a compound of formula (XIII):

with a compound of formula (

or NRsR9 represents an aliphatic 4-8 membered heterocycle containing one or more heteroatoms selected from O, N and S, as previously defined. Typical reaction conditions may be stirring the mixture at room temperature for about 16 hours. This process may be followed, where appropriate, by deprotection. Compounds of formula (XIII) may be prepared by reacting a compound of formula (IX) with a compound of formula (XV):

(XV) 2 )^^ Br

The compounds may typically be reacted in the presence of HATU, Hunig's base (DIPEA) and DMF, or another polar organic solvent.

Alternatively compounds of formula (Ic) which are compounds of formula (I) in which Z represents CO or SO2 and R3 represents formula (i), X1-R4 represents Me and NRsR9 represents NRsR9 as previously defined, or an aliphatic 4-8 membered heterocycle containing one or more heteroatoms selected from O, N and S, as previously defined:

with a compound of formula (XIV). Typical reaction conditions may be stirring the mixture in a solvent such as DMF at approximately 60 °C for about 5 hours optionally in the presence of a base such as Hunig's base (DIPEA), followed by a deprotection step, such as by treatment with a nucleophile such as piperidine in a solvent such as DMF at approximately 60 °C for about 18 hours. Compounds of formula (XVI) may be prepared by reacting a compound of formula (XVII): OH

with CBr 4 and PP under mild conditions, a common reaction known in the field as the Appel Reaction. Typically, the reaction may be stirred for between 2 and 16 hours at room temperature in a solvent such as DCM.

Compounds of formula (XVII), wherein Z represents CO, may be prepared by reacting a compound of formula (IX) with a compound of formula (XVIII):

(XVIII)

H

The compounds may typically be reacted in the presence of HATU, Hunig's base (DIPEA) and DMF, or another polar organic solvent. Typically, the reaction may be stirred for about 2 hours at room temperature.

Compounds of formula (XVII), wherein Z represents SO2, may be prepared by reacting a compound of formula (II) with a compound of formula (III) in which Z represents SO2, and R3 is formula (i) wherein Xi-R 4 represents Me and Y1-R5 represents CH2CH2OH. The

compounds may typically be reacted in the presence of Hunig's base (DIPEA) and DMF, or another polar organic solvent at a temperature such as 80 °C for about 18 hours.

Synthesis of compounds of formula (III) may be prepared by reacting a compound of formula (X) with a compound of formula (XIX):

The compounds may typically be reacted in the presence of Hunig's base (DIPEA) and DMF for about 16 hours at room temperature, followed by a deprotection step using standard reagents in the art, such as TFA. Compounds of formula (III), wherein R3 is formula (ii) and (iii), may be prepared by reacting a compound of formula (X), wherein R' and R" are joined together with the N and O atoms to make the heterocyclic ring of choice from the examples listed, with a compound of formula (XIX). These heterocyclic rings and their preparation methods are common knowledge in the field.

Compounds of formula (X), wherein R" is H, may be prepared by reacting a compound of formula (XX):

with, typically, hydrazine hydrate in the presence of solvents, typically methanol and DCM for about 18 hours at room temperature.

Compounds of formula (XX) may be prepared by reacting a compound of HOR' with a compound of 2-hydroxyisoindoline-1 ,3-dione. The compounds may typically be reacted in the presence of triphenylphosphine, an azodicarboxylate, such as DIAD, and a polar solvent, such as THF. Typically, the reaction is stirred for 1 hour at 0 °C and then warmed to room temperature and stirred for about 16 hours.

Compounds of formula (X), wherein R" is Me, may be prepared by reducing a compound of formula (XXI):

using standard methods in the art, for example, by the use of a reducing agent, such as a compound of formula (XXII):

The reaction may typically take place in the presence of a polar solvent, such as THF, an acid, such as hydrochloric acid and an aprotic solvent, such as dioxane.

Compounds of formula (XXI) may be prepared by the condensation a compound of formula

(XXIII):

H 2 N— O

(XXIII)

R' with, for example, formaldehyde. The condensation may typically occur by use of paraformaldehyde as a source of formaldehyde in the presence of a solvent, such as ethanol and heated under reflux for about 18 hours. Novel intermediates including compounds of formula (II), (VII), (IX), (XI) (XIII), (XVI) and (XVII) wherein Ri is Me, Et, CH=CH 2 , C≡C-H or C≡C-Me, and formula (VI) wherein Ri is Et, CH=CH 2 , C≡C-H or C≡C-Me, and salts thereof are claimed as an aspect of the invention.

Compounds of formulae (V), (VIII), (X), (XII), (XIV), (XV), (XVIII), (XIX), (XXII) and (XXIII) may be prepared by known methods, or methods analogous to those described herein.

Compounds of formula (I) may be prepared or employed in the form of a pharmaceutically acceptable salt, including the therapeutically active non-toxic acid addition salts that the compounds of formula (I) are able to form. These pharmaceutically acceptable acid addition salts can conveniently be obtained by treating the free base form with such appropriate acids in a suitable solvent or mixture of solvents. Appropriate acids comprise, for example, ethanesulfonic, maleic, malonic, L-tartaric, fumaric, citric, succinic, acetic, triphenyl acetic, hydrochloric, sulfuric, phosphoric, 1-hydroxy-2-naphthoic, hydrobromic, methanesulfonic, tartaric, palmitic, isethionic, pamoic, formic, cinnamic benzoic, ascorbic , galactaric , lactic, malic , oxalic, para- toluenesulfonic, benzenesulphonic, propionic, furoic, phosphonic and glutaric. Conversely said salt forms can be converted by treatment with an appropriate base into the free base form.

The invention provides a compound of the invention for use as a pharmaceutical.

Further, the present invention provides a pharmaceutical composition comprising a compound according to the invention optionally in combination with one or more pharmaceutically acceptable diluents or carriers. Diluents and carriers may include those suitable for parenteral, oral, topical including by inhalation via the mouth into the lungs or inhalation via the nose, mucosal and rectal administration, and may be different depending on the route of administration.

In one embodiment compositions may be prepared e.g. for parenteral administration e.g., subcutaneous, intramuscular, intravenous, intra-dermal, intra-articular or peri-articular administration, particularly in the form of liquid solutions or suspensions; for oral administration, particularly in the form of tablets, capsules, powder, granules, solid dispersions or in the form of liquid solutions or suspensions including nanosuspensions; for inhalation to the lungs or nose e.g. pulmonary or intranasal administration, particularly in the form of dry powders, solutions, suspensions including nanosuspensions for nebulisation, nasal sprays or drops comprising solutions or suspensions or suspension or solution pressurised or non-pressurised aerosols; for topical or transdermal administration e.g. as creams, sprays, foams, gels, ointments, liquids, patches; for mucosal administration e.g. to buccal, sublingual or vaginal mucosa, and for rectal administration e.g. in the form of a foam or suppository.

The compositions may conveniently be administered in unit dosage form and may be prepared by any of the methods well-known in the pharmaceutical art, for example as described in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, PA., (1985). The compositions may also conveniently be administered in multiple unit dosage form.

Formulations for parenteral administration may contain as excipients sterile water or saline, buffers, tonicity-adjusting agents, preservatives, anti-oxidants, viscosity adjusting agents, alkylene glycols such as propylene glycol, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin, hydrogenated naphthalenes and the like.

Compositions suitable for oral administration may comprise one or more physiologically compatible carriers and/or excipients and may be in solid or liquid form. Tablets and capsules may be prepared with binding agents, for example, syrup, acacia, gelatin, sorbitol, tragacanth, celluloses or polyvinylpyrrolidone; fillers, such as lactose, sucrose, corn starch, calcium phosphate, sorbitol, or glycine; lubricants, such as magnesium stearate, talc, polyethylene glycol, or silica; and surfactants, such as sodium lauryl sulfate. Liquid compositions may contain conventional additives such as suspending agents, for example sorbitol syrup, methyl cellulose, sugar syrup, gelatin, carboxymethyl-cellulose, or edible fats; emulsifying agents and surfactants such as lecithin, or acacia; vegetable oils such as almond oil, coconut oil, cod liver oil, or peanut oil; preservatives such as butylated hydroxyanisole (BHA) and butylated hydroxytoluene (BHT). Liquid compositions may be encapsulated in, for example, gelatin to provide a unit dosage form.

Solid oral dosage forms include tablets, two-piece hard shell capsules and soft elastic gelatin (SEG) capsules. Such two-piece hard shell capsules may be made from, for example, gelatin or hydroxypropyl methylcellulose (HPMC).

A dry shell formulation typically comprises of about 40%-60% concentration of gelatin, about a 20%-30% concentration of plasticizer (such as glycerin, sorbitol or propylene glycol) and about a 30%-40% concentration of water. Other materials such as preservatives, dyes, opacifiers and flavours also may be present. The liquid fill material comprises a solid drug that has been dissolved, solubilized or dispersed (with suspending agents such as beeswax, hydrogenated castor oil or polyethylene glycol 4000) or a liquid drug in vehicles or combinations of vehicles such as mineral oil, vegetable oils, triglycerides, glycols, polyols and surface-active agents.

Formulations for nasal administration may be powders and may contain excipients, for example, lactose or dextran, or may be aqueous or oily solutions for use in the form of nasal drops or metered spray. Formulations for nasal administration may also be in the form of aqueous suspensions or pressurised non-aqueous solutions or suspensions. For buccal administration typical excipients include sugars, calcium stearate, magnesium stearate, pregelatinated starch, and the like. Suitably the compound of formula (I) is administered topically to the lung. Hence in one embodiment there is provided a pharmaceutical composition comprising a compound of the disclosure optionally in combination with one or more topically acceptable diluents or carriers.

Topical administration to the lung may be achieved by use of a non-pressurised formulation such as an aqueous solution or suspension. These formulations may be administered by means of a nebuliser e.g. one that can be hand-held and portable or for home or hospital use (ie non-portable). The formulation may comprise excipients such as water, buffers, tonicity adjusting agents, pH adjusting agents, surfactants, preservatives, suspending agents, bulking agents and co-solvents. Suspension liquid and aerosol formulations (whether pressurised or unpressurised) will typically contain the compound of the invention in finely divided form suitable for deposition into the lung, for example with a D50 of 0.5-10 μηι e.g. around 1-5 μηι. Powders in finely divided form may be prepared by a micronization or milling process, by spray drying, by spray freezing, or by wet milling followed by spray drying. Micronization may be performed using a jet mill such as those manufactured by Hosokawa Alpine. The resultant particle size distribution may be measured using laser diffraction (e.g. with a Malvern Mastersizer 2000S or a Mastersizer 3000 instrument). Particle size distributions may be represented using D10, D50 and D90 values. The D50 median value of particle size distributions is defined as the particle size that divides the distribution in half. The measurement derived from laser diffraction is more accurately described as a volume distribution, and consequently the D50 value obtained using this procedure is more meaningfully referred to as a Dvso value (median for a volume distribution). As used herein Dv values refer to particle size distributions measured using laser diffraction. Similarly, D10 and D90 values, used in the context of laser diffraction, are taken to mean Dvio and Dvgo values and refer to the particle size whereby 10% of the distribution lies below the D10 value, and 90% of the distribution lies below the Dgo value, respectively. In another embodiment, composite particles of the compound of the disclosure and excipients for use in nebulisation of a suspension formulation may be formed by co-milling and / or co-spray drying the compound and excipients together, wherein the composite particles comprising both active and excipients have a D50 of 1-10 μηι. Aqueous suspension formulations for delivery to the lung could also comprise nanosuspensions or suspensions of composite particles containing nanoparticles. Topical administration to the lung may also be achieved by use of a pressurised aerosol formulation. Aerosol formulations typically comprise the active ingredient suspended or dissolved in a suitable aerosol propellant, such as a chlorofluorocarbon (CFC) or a hydrofluorocarbon (HFC). Suitable CFC propellants include trichloromonofluoromethane (propellant 1 1), dichlorotetrafluoromethane (propellant 1 14), and dichlorodifluoromethane (propellant 12). Suitable HFC propellants include tetrafluoroethane (HFC-134a) and heptafluoropropane (HFC-227). The propellant typically comprises 40%-99.5% e.g. 40%-90% by weight of the total inhalation composition. The formulation may comprise excipients including co-solvents (e.g. ethanol) and surfactants or stabilisers (e.g. lecithin, sorbitan trioleate and the like). Other possible excipients include polyethylene glycol, polyvinylpyrrolidone, glycerine and the like. Aerosol formulations are packaged in canisters and a suitable dose is delivered by means of a metering valve (e.g. as supplied by Bespak, Aptar or 3M or alternatively by Coster or Vari). Pressurised suspensions formulations for delivery to the lung could also comprise nanosuspensions or suspensions of composite particles containing nanoparticles.

Topical administration to the lung may also be achieved by use of a dry-powder formulation. A dry powder formulation will contain the compound of the disclosure in finely divided form, typically with a D50 of 0.5-10 μηι e.g. around 1-5 μηι. Powders in finely divided form may be prepared by a micronization or milling process, by spray drying, by spray freezing, or by wet milling followed by spray drying. Micronization may be performed using a jet mill such as those manufactured by Hosokawa Alpine. The resultant particle size distribution may be measured using laser diffraction (e.g. with a Malvern Mastersizer 2000S or a Mastersizer 3000 instrument). The formulation will typically contain one or more topically acceptable diluents such as lactose, glucose, trehalose or mannitol (preferably lactose), usually of comparatively large particle size e.g. a D50 of 15-250 μηι. In another embodiment, composite particles of the compound of the disclosure and excipients may also be formed by co-milling and / or co-spray drying the compound and excipients together, wherein the composite particles comprising both active and excipients have a D50 of 1-10 μηι. As used herein, the term "lactose" refers to a lactose-containing component, including a-lactose monohydrate, β-lactose monohydrate, a- lactose anhydrous, β-lactose anhydrous and amorphous lactose. Lactose components may be processed by micronization, sieving, milling, compression, agglomeration or spray drying. Commercially available forms of lactose in various forms are also encompassed, for example Lactohale ® (DFE Pharma), InhaLac ® (Meggle), Pharmatose ® (DFE Pharma) and Respitose ® (DFE Pharma) products. In one embodiment, the lactose component is selected from the group consisting of a-lactose monohydrate, a-lactose anhydrous and amorphous lactose. Preferably, the lactose is α-lactose monohydrate.

Dry powder formulations may also contain other excipients such as leucine, sodium stearate, calcium stearate or magnesium stearate. Dry powder particles could be composite particles and could be comprised of nanoparticles in a composite matrix. A dry powder formulation is typically delivered using a dry powder inhaler (DPI) device. This can either be a unit dose device where the formulation is presented in individual units, either as capsules or blisters, or in a multi-dose device where more than one dose of formulation is contained in a device, either in a bulk reservoir or as multiple containers within a device (eg multiple blisters or pockets). Example dry powder inhalers include SPINHALER, DISKHALER, TURBOHALER, DISKUS, ELLIPTA.CLICKHALER, ECLIPSE, ROTAHALER, HANDIHALER, AEROLISER, CYCLOHALER, MONODOSE, BREEZHALER/NEOHALER, FLOWCAPS, TWINCAPS, X-CAPS, TWISTER, TURBOSPIN, ELPENHALER, TURBUHALER, MIATHALER, NEXTHaler, TWISTHALER, NOVOLIZER, GENUAIR, SKYEHALER, ORIEL dry powder inhaler, MICRODOSE, ACCUHALER, PULVINAL, EASYHALER, ULTRAHALER, TAIFUN, PULMOJET, OMNIHALER, GYROHALER, TAPER, CONIX, XCELOVAIR and PROHALER.

The compounds of the invention are expected to be useful in the treatment of fibrotic diseases such as lung fibrosis and pulmonary diseases with a fibrotic component e.g. selected from IPF, giant cell interstitial pneumonia, sarcoidosis, cystic fibrosis, respiratory distress syndrome, drug-induced lung fibrosis, granulomatosis, silicosis, asbestosis, systemic scleroderma, the virally induced hepatic cirrhosis selected from hepatitis C induced hepatic cirrhosis, or diseases of the skin with a fibrotic component e.g. selected from scleroderma, sarcoidosis and systemic lupus erythematosus, and especially IPF. More generally, the compounds of the invention are expected to be useful in the treatment of interstitial lung diseases. In addition, the compounds of the invention are expected to be useful in the treatment of diseases characterized by hyperproliferation of cells, for example, cancer and wherein the compounds are delivered by inhalation, particularly lung cancer. Furthermore, the compounds of the invention may also be useful in the treatment of respiratory disorders including COPD (including chronic bronchitis and emphysema), asthma, paediatric asthma, allergic rhinitis, rhinitis, sinusitis, especially asthma, chronic bronchitis and COPD.

The compounds of the invention are also expected to be useful in the treatment of other fibrotic diseases such as lung fibrosis associated with rheumatoid arthritis, respiratory distress syndrome including acute respiratory distress syndrome, acute lung injury, radiation induced lung fibrosis or pneumonitis, chronic hypersensitivity pneumonitis, systemic sclerosis, Sjogren's syndrome, interstitial lung diseases, pulmonary arterial hypertension (PAH), including the vascular component of PAH, or diseases of the skin with a fibrotic component e.g. selected from hypertrophic scarring and keloids, or eye diseases where fibrosis is a component including glaucoma, age related macular degeneration, diabetic macular edema, dry eye disease and diabetic retinopathy, or fibrosis in the gut e.g. associated with inflammatory bowel disease.

In addition, the compounds of the invention are expected to be useful in the prevention of diseases characterized by hyperproliferation of cells, for example, cancer e.g. wherein the compounds are delivered by inhalation, particularly lung cancer.

The invention provides a compound of the invention for use in the treatment of one or more of the above mentioned diseases. The invention also provides use of a compound of the invention in the manufacture of a medicament for the treatment of one or more of the above mentioned diseases.

The invention also provides a method of treatment of one of the above mentioned diseases which comprises administering to a subject (especially a human subject) in need thereof a therapeutically effective amount of a compound of the invention. The word "treatment" is intended to embrace prophylaxis as well as therapeutic treatment.

Compounds of the invention may be administered once, twice or thrice per day, especially once or twice per day. A suitable dosage amount may be determined by reference to the severity of the disease and the size of the subject. Typical dosage amounts are in the range 0.01 mg to 100 mg, e.g. 0.1 mg to 10 mg e.g. 0.25 mg to 5 mg per human dose to be delivered once, twice or thrice per day, especially once or twice per day. The compound of the disclosure may also be administered in combination with one or more other active ingredients e.g. active ingredients suitable for treating the above mentioned conditions. For example possible active ingredients include nintedanib or pirfenidone (these being known for treatment of IPF). Other active ingredients to be used in combination include substances with a secretolytic, broncholytic and/or anti-inflammatory activity, such as anticholinergic agents, beta-2 mimetics, steroids, PDE-IV inhibitors, p38 MAP kinase inhibitors, MK2 inhibitors, galectin inhibitors, NKi antagonists, LTD4 antagonists, EGFR inhibitors, VEGF inhibitors, PDGF inhibitors, FGF inhibitors, TGFbeta inhibitors, LPA1 antagonists, LOXL2 inhibitors, CTGF inhibitors, pentoxyfylline, N-acetylcysteine, anti-IL13 agents, anti IL4 agents, ΑΙρϊ^νβθ integrin inhibitors, IGF inhibitors, PI3K inhibitors, mTOR inhibitors, JNK inhibitors, pentraxin2 and endothelin-antagonists.

Other active ingredients to be used in combination include substances with an antifibrotic activity, such as PDE-III inhibitors, combined anti-IL4/13 agents, combined PI3k/mTOR inhibitors, autotaxin inhibitors, P2X3 antagonists, CTGF antagonists, 5-LO antagonists, leukotriene antagonists and ROCK inhibitors.

In one embodiment the combination of active ingredients are co-formulated. In one embodiment the combination of active ingredients is co-administered sequentially or simultaneously.

In one embodiment the compounds of the invention are delivered by inhalation and the other possible active ingredients are delivered orally or parenterally.

In one embodiment there is provided a combination product comprising:

(A) a compound of the invention; and

(B) a further active ingredient (as mentioned above)

wherein each of components (A) and (B) is formulated in admixture with pharmaceutically- acceptable diluent(s) or carrier(s). The combination may optionally comprise additional relevant excipients.

In one embodiment there is provided a compound of the invention for use as a medicament to be administered in combination with one or more further active ingredients (as mentioned above). Compounds of the invention are expected to have one or more of the following advantageous properties:

- Good inhibitory activity of kinases selected from VEGFR (e.g. VEGFR1 and VEGFR2), FGFR and PDGFR;

- Good anti-fibrosis activity e.g. as determined in in vivo models (e.g. bleomycin fibrosis model) when delivered topically to the lung;

- Suitable physical and chemical properties and low dose for a medicinal product, particularly one intended to be delivered topically to the lung;

- Good residency time in the lung or duration of action when administered topically to the lung; - Low permeability in the PAMPA permeability assay;

-Good duration of action e.g. as measured by inhibition of PDGF-BB induced phosphorylation of BDGFRβin human fetal lung fibroblast cells;

- Good safety and tolerability when administered topically to the lung;

- Low oral bioavailability.

EXPERIMENTAL SECTION

Abbreviations used herein are defined below (Table 1). Any abbreviations not defined are intended to convey their generally accepted meaning.

Table 1 : Abbreviations

AcOH glacial acetic acid

aq aqueous

BAL bronchoalveolar lavage

br broad

BEH ethylene bridged hybrid

Boc te/f-butoxycarbonyl

CSH charged surface hybrid

cone concentrated

d doublet

δ chemical shift

DCM dichloromethane

DIAD diisopropyl azodicarboxylate

DMF A/,A/-dimethylformamide

DMSO dimethyl sulfoxide

(ES + ) electrospray ionization, positive mode electrospray ionization, negative mode

ethyl

ethyl acetate

ethanol

hour(s)

1-[bis(dimethylamino)methylene]-1 /-/-1 ,2,3-triazolo[4,5- 5]pyridinium 3- oxid hexafluorophosphate

base A/./V-diisopropylethylamine

molar

multiplet

protonated molecular ion

deprotonated molecular ion

methyl

acetonitrile

methanol

megahertz

minute(s)

mass-to-charge ratio

nuclear magnetic resonance (spectroscopy)

pentuplet

phenyl

pyridine

quartet

room temperature

high performance liquid chromatography

singlet

saturated

solid supported anion exchange (resin)

solid supported cation exchange (resin)

triplet

te/f-butyl

tetrahydrofuran

trifluoroacetic acid

ultra-violet

weight Chemistry Examples

General Procedures All starting materials and solvents were obtained either from commercial sources or prepared according to the literature citation. Unless otherwise stated all reactions were stirred. Organic solutions were routinely dried over anhydrous magnesium sulfate. Hydrogenations were performed on a Thales H-cube flow reactor under the conditions stated or under pressure in a gas autoclave (bomb).

Column chromatography was performed on pre-packed silica (230-400 mesh, 40-63 μηι) cartridges using the amount indicated. SCX was purchased from Supelco and treated with 1 M hydrochloric acid prior to use. Unless stated otherwise the reaction mixture to be purified was first diluted with MeOH and made acidic with a few drops of AcOH. This solution was loaded directly onto the SCX and washed with MeOH. The desired material was then eluted by washing with 1 % NH 3 in MeOH.

Preparative Reverse Phase High Performance Liquid Chromatography

Performed using UV detection at 215 and 254 nm with either a Waters X-Select Prep-C18, 5 μηι, 19x50 mm column eluting with a H20-MeCN gradient containing 0.1 % v/v formic acid over 10 min (Method A), or a Waters X-Bridge Prep-C18, 5 μηι, 19x50 mm column eluting with a H20-MeCN gradient containing 0.1 % ammonium bicarbonate over 10 min (Method B).

Analytical Methods

Reverse Phase High Performance Liquid Chromatography

Method 1 : Waters XSelect CSH C18 2.5 μπι (4.6 x 30 mm) at 40°C; flow rate 2.5-4.5 mL min -1 eluted with a H20-MeCN gradient containing 0.1 % v/v formic acid over 4 min employing UV detection at 254 and 215 nm. Gradient information: 0-3.00 min, ramped from 95% H2O- 5% MeCN to 5% H 2 0-95% MeCN; 3.00-3.01 min, held at 5% H 2 0-95% MeCN, flow rate increased to 4.5 mL min "1 ; 3.01-3.50 min, held at 5% H 2 0-95% MeCN; 3.50-3.60 min, returned to 95% H 2 0-5% MeCN, flow rate reduced to 3.50 mL min "1 ; 3.60-3.90 min, held at 95% H 2 0-5% MeCN; 3.90-4.00 min, held at 95% H 2 0-5% MeCN, flow rate reduced to 2.5 mL min -1 . Method 2: Waters XBridge BEH C18, 2.5 μΓΠ (4.6 x 30 mm) at 40°C; flow rate 2.5-4.5 mL min -1 eluted with a H 2 0-MeCN gradient containing 10 mM ammonium bicarbonate over 4 min employing UV detection at 254 nm. Gradient information: 0-3.00 min, ramped from 95% H 2 0- 5% MeCN to 5% H 2 0-95% MeCN; 3.00-3.01 min, held at 5% H 2 0-95% MeCN, flow rate increased to 4.5 mL min "1 ; 3.01- 3.50 min, held at 5% H 2 0-95% MeCN; 3.50-3.60 min, returned to 95% H 2 0-5% MeCN, flow rate reduced to 3.50 mL min "1 ; 3.60-3.90 min, held at 95% H 2 0-5% MeCN; 3.90-4.00 min, held at 95% H 2 0-5% MeCN, flow rate reduced to 2.5 mL min -1 . 1 H NMR Spectroscopy

1 H NMR spectra were acquired on a Bruker Avance III spectrometer at 400 MHz using residual undeuterated solvent as reference and unless specified otherwise were run in DMSO-de. All chemical names have been generated using CambridgeSoft ENotebook 12.0.

Example 1 : (Z)-Methyl 3-(((4-(methoxy(methyl)carbamoyl)phenyl)amino)

(phenyl)methylene)-2-oxoindoline-6-carboxylate

Intermediate A: (£)-Methyl 1 -acetyl-3-(ethoxy(phenyl)methylene)-2-oxoindoline-6- carboxylate

To a stirred solution of methyl 2-oxoindoline-6-carboxylate (1.00 g, 5.23 mmol) in acetic anhydride (10.9 mL, 1 15 mmol) was added (triethoxymethyl)benzene (3.55 mL, 15.7 mmol) and the mixture was stirred at 1 10 °C for 4 h. The reaction mixture was cooled to rt and the resulting precipitate collected by filtration. The precipitate was washed with hexanes (100 mL) and dried in vacuo to afford the subtitle compound (£)-methyl 1-acetyl-3- (ethoxy(phenyl)methylene)-2-oxoindoline-6-carboxylate as a beige solid (1.66 g, 82%); R l 2.61 min (Method 1); m/z 366 (M+H) + (ES + ). (Z)-Methyl 3-(((4-(methoxy(methyl)carbamoyl)phenyl)amino)(phenyl)methyl ene)-2- oxoindoline-6-carbox late

A mixture of 4-amino-A/-methoxy-A/-methylbenzamide (32.6 mg, 0.181 mmol) and (£)-methyl 1-acetyl-3-(ethoxy(phenyl)methylene)-2-oxoindoline-6-carboxy late (Intermediate A) (60 mg, 0.16 mmol) in DMF (1 mL) was heated to 80 °C for 18 h then allowed to cool to rt. Piperidine (45.0 μί, 0.455 mmol) was added and the reaction mixture stirred at rt for 24 h. The reaction mixture was partitioned between DCM (25 mL) and saturated NaHCC>3 solution (25 mL) and the layers separated. The organic layer was washed with brine (25 mL) and then

concentrated under reduced pressure. The crude product so obtained was purified by preparative HPLC (Method A, 30-50% MeCN in water) to afford the title compound (Z)- methyl 3-(((4-(methoxy(methyl)carbamoyl)phenyl)amino)(phenyl)methyl ene)-2-oxoindoline-6- carboxylate as a yellow solid (21 mg, 28%); R* 2.14 min (Method 1); m/z 458 (M+H) + (ES + ); 1 H NMR δ: 3.19 (3H, s), 3.48 (3H, s), 3.77 (3H, s), 5.87 (1 H, d), 6.86 (2H, m), 7.21 (1 H, dd), 7.36-7.45 (3H, overlapping m), 7.53 (2H, m), 7.56-7.68 (3H, overlapping m), 1 1.01 (1 H, s), 12.29 (1 H, s).

Example 2: (Z)-Methyl 5-methyl-3-(((4-( V-(2-(4-methylpiperazin-1 - yl)ethoxy)sulfamoyl)phenyl)amino)(phenyl)methylene)-2-oxoind oline-6-carboxylate, formate

Intermediate B: Methyl 4-(2-methoxy-2-oxoethyl)-2-methyl-5-nitrobenzoate

To a stirred solution of potassium te/f-butoxide (35.9 g, 320 mmol) in DMF (350 mL), under a nitrogen atmosphere at -20 °C, was added a solution of methyl 2-methyl-5-nitrobenzoate (25.0 g, 128 mmol) and methyl 2-chloroacetate (16.9 mL, 192 mmol) in DMF (300 mL) dropwise over 40 minutes. The reaction mixture was warmed to -10 °C over 2 h and then poured onto an ice-HCI slurry (900 g ice, 500 mL 35 wt% HCI). The mixture was extracted with DCM (2 x 600 mL) and the combined organic layers washed with brine (2 x 400 mL) and then evaporated under reduced pressure. The crude product so obtained was purified by flash column chromatograhy (S1O2, 330 g, 0-10% EtOAc in DCM, gradient elution) to afford the subtitle compound methyl 4-(2-methoxy-2-oxoethyl)-2-methyl-5-nitrobenzoate as an orange syrup (31.0 g, 89%); R' 2.06 min (Method 1); m/z 266 (M-H) " (ES " ); 1 H NMR δ: 2.61 (3H, s), 3.62 (3H, s), 3.88 (3H, s), 4.12 (2H, s), 7.59 (1 H, s), 8.51 (1 H, s).

Intermediat -methyl-2-oxoindoline-6-carboxylate

To a solution of methyl 4-(2-methoxy-2-oxoethyl)-2-methyl-5-nitrobenzoate (Intermediate B)

(23.0 g, 86.0 mmol) in acetic acid (301 mL, 5.25 mol) was added palladium on carbon

[5 wt%, 58% water, type 87L] (3.30 g, 1.55 mmol). The mixture was hydrogenated at rt under an atmosphere of H2 (5 bar) for 36 h and then filtered through a pad of celite. The filter cake was washed with EtOAc (500 mL) and the filtrate concentrated under reduced pressure. The crude residue was dissolved in hot refluxing MeOH (200 mL) and the mixture cooled to rt. The resultant solid was filtered, rinsing with MeOH (200 mL) and dried in vacuo to afford the subtitle compound methyl 5-methyl-2-oxoindoline-6-carboxylate as a brown powder (7.00 g, 39%); R* 1.48 min (Method 1); m/z 206 (M+H) + (ES + ); 1 H NMR δ: 2.45 (3H, s), 3.32 (2H, s), 3.81 (3H, s), 7.17 (1 H, s), 7.22 (1 H, s), 10.43 (1 H, s).

Intermediate D: (£)-Methyl 1 -acetyl-3-(ethoxy(phenyl)methylene)-5-methyl-2- oxoindoli -6-carboxylate

To a stirred solution of methyl 5-methyl-2-oxoindoline-6-carboxylate (Intermediate C) (5.00 g, 24.4 mmol) in acetic anhydride (50.6 mL, 536 mmol) was added (triethoxymethyl)benzene (22.1 mL, 97.0 mmol) and the mixture was stirred at 110 °C for 3 h. Thereafter, stirring was continued at rt for 18 h. The reaction mixture was concentrated under reduced pressure and the residue was triturated with MeOH (50 mL). The resultant solid was filtered, rinsing with MeOH (50 ml_) and dried in vacuo to afford the subtitle compound (£)-methyl 1-acetyl-3- (ethoxy(phenyl)methylene)-5-methyl-2-oxoindoline-6-carboxyla te as a yellow powder (4.50 g, 48%); R' 2.70 min (Method 1); m/z 380 (M+H) + (ES + ); 1 H NMR δ: 1.35 (3H, t), 2.42 (3H, s), 2.58 (3H, s), 3.84 (3H, s), 4.01 (2H, q), 7.45-7.62 (5H, overlapping m), 7.90 (1 H, s), 8.64 (1 H, s).

Intermediate E: 2-(2-(4-Methylpiperazin-1 -yl)ethoxy)isoindoline-1 ,3-dione

To a solution of 2-hydroxyisoindoline-1 ,3-dione (2.04 g, 12.5 mmol) and triphenylphosphine (3.27 g, 12.5 mmol) in THF (40 ml_) at 0 °C was added 2-(4-methylpiperazin-1-yl)ethanol (1.50 ml_, 10.4 mmol) dropwise. The mixture was then stirred at 0 °C for 30 min before DIAD (2.43 ml_, 12.5 mmol) was added dropwise. The reaction was stirred for a further 30 min at 0 °C before being warmed to rt and stirred for 16 h. The solvent was removed under reduced pressure and the residue re-dissolved in EtOAc (75 ml_). The organic layer was washed with saturated NaHCC>3 solution (2 x 50 ml_) and then dried over Na2S0 4 , filtered and

concentrated under reduced pressure to approximately 30 ml_. The organic layer was cooled to 0 °C and cold 1 M aqueous HCI solution (30 ml_) was added. On complete addition, the mixture was warmed to rt and stirred for 20 min. The layers were separated and the aqueous layer washed with Et20 (2 x 30 ml_). After cooling to 0 °C, the aqueous layer was basified by slow addition of a saturated NaHCC>3 solution before being extracted with CHC (3 x 50 ml_). The organic extracts were combined and then dried over Na2S0 4 , filtered and concentrated under reduced pressure to afford the subtitle compound 2-(2-(4-methylpiperazin-1- yl)ethoxy)isoindoline-1 ,3-dione as a beige solid (1.96 g, 63%); 1 H NMR δ: 1.84-2.47 (8H, overlapping m), 1.98 (3H, s), 2.65 (2H, t), 4.24 (2H, t), 7.85-7.87 (4H, overlapping m).

Intermediate F: 0-(2-(4-Methylpiperazin-1 -yl)ethyl)hydroxylamine

To a solution of 2-(2-(4-methylpiperazin-1-yl)ethoxy)isoindoline-1 ,3-dione (Intermediate E) (1.88 g, 6.50 mmol) in MeOH/DCM (2:1 , 15 ml_) was added a 25% aqueous solution of hydrazine (2.45 mL, 19.5 mmol). The mixture was stirred at rt for 18 h, then was diluted with Et20 (100 mL). The precipitate was removed by filtration and the solvent was removed under reduced pressure. The residue was re-dissolved in DCM (30 mL) and was dried over

Na2S0 4 . The solvent was removed under reduced pressure to afford the subtitle compound 0-(2-(4-methylpiperazin-1-yl)ethyl)hydroxylamine as a pale yellow oil (0.902 g, 83%); 1 H NMR δ: 2.13 (3H, s), 2.18-2.47 (8H, overlapping m), 2.43 (2H, t), 3.60 (2H, t), 5.96 (2H, s).

Intermediate G: ferf-Butyl (4-(/V-(2-(4-methylpiperazin-1 - yl)ethoxy)sulfamoyl)phen

To a suspension of 0-(2-(4-methylpiperazin-1-yl)ethyl)hydroxylamine (Intermediate F) (464 mg, 2.91 mmol) in DMF (15 mL) was added Hunig's base (1.05 mL, 6.00 mmol) followed by te/f-butyl (4-(chlorosulfonyl)phenyl)carbamate (500 mg, 1.71 mmol). The mixture was stirred at rt overnight. The majority of the solvent was removed under reduced pressure. The residue was re-dissolved in MeOH (50 mL) and filtered through an SCX column (5 g), washing with MeOH (75 mL). The filtrate was discarded before the column was washed with 1 % NH3 in MeOH (75 mL). The solvent was removed under reduced pressure to afford the subtitle compound te/f-butyl (4-(/V-(2-(4-methylpiperazin-1- yl)ethoxy)sulfamoyl)phenyl)carbamate as as a pale colourless gum (552 mg, 70%); R' 1.77 min (Method 1); m/z 415 (M+H) + (ES + ); 1 H NMR δ: 1.49 (9H, s), 2.12 (3H, s), 2.16-2.46 (10H, overlapping m), 3.93 (2H, t), 7.64 (2H, m), 7.74 (2H, m), 9.87 (1 H, s), 10.19 (1 H, br s).

Intermediate H: 4-Amino- V-(2-(4-methylpiperazin-1 -yl)ethoxy)benzenesulfonamide, di- trifluoroacetate salt

To a solution of te/f-butyl (4-(/V-(2-(4-methylpiperazin-1- yl)ethoxy)sulfamoyl)phenyl)carbamate (Intermediate G) (334 mg, 0.806 mmol) in DCM (5 mL) was added TFA (1 mL). The mixture was stirred at rt for 4 h. The solvent was removed under reduced pressure to afford 4-amino-/V-(2-(4-methylpiperazin-1- yl)ethoxy)benzenesulfonamide, di-trifluoroacetate salt as a clear oil (334 mg, 67%); R l 1.01 min (Method 2); m/z 315 (M+H) + (ES + ).

(Z)-Methyl 5-methyl-3-(((4-( V-(2-(4-methylpiperazin-1 - yl)ethoxy)sulfamoyl)phenyl)amino)(phenyl)methylene)-2-oxoind oline-6-carboxylate, formate

(E)-Methyl 1-acetyl-3-(ethoxy(phenyl)methylene)-5-methyl-2-oxoindoline- 6-carboxylate (Intermediate D) (100 mg, 0.264 mmol) and 4-amino-/V-(2-(4-methylpiperazin-1- yl)ethoxy)benzenesulfonamide, di-trifluoroacetate salt (Intermediate H) (172 mg, 0.316 mmol) were combined in DMF (3 ml_). The mixture was heated at 80 °C for 24 h. More 4- amino-/V-(2-(4-methylpiperazin-1-yl)ethoxy)benzenesulfonamid e, di-trifluoroacetate salt (50 mg) was added and the reaction mixture was reheated to 80 °C for 24 h. Piperidine (261 μΙ, 2.64 mmol) was added and the mixture was stirred for 1.5 h. The solvent was removed under reduced pressure. The material was taken up in a 10% MeOH in DCM solution (20 ml_) and washed with water (20 ml_). The phases were separated using a phase separator cartridge. The crude product was purified by flash column chromatography (S1O2, 40 g, 0-10% MeOH in DCM, gradient elution). The columned product was purified by preparative HPLC (Method A, 20-50% MeCN in water) to afford the title compound (Z)-methyl 5-methyl-3-(((4-(/V-(2-(4- methylpiperazin-1-yl)ethoxy)sulfamoyl)phenyl)amino)(phenyl)m ethylene)-2-oxoindoline-6- carboxylate, formate as a light yellow solid (17.4 mg, 10%); R' 1.64 min (Method 1); m/z 606 (M+H) + (ES + ); 1 H NMR δ: 2.13 (3H, s), 2.15 (3H, s), 2.18-2.36 (8H, overlapping m), 2.42 (2H, t), 3.76 (3H, s), 3.91 (2H, t), 5.63 (1 H, s), 6.95 (2H, m), 7.36 (1 H, s), 7.53-7.60 (2H, overlapping m), 7.60-7.73 (5H, overlapping m), 8.20 (1 H, s), 10.27 (1 H, br s), 10.94 (1 H, s), 12.28 (1 H, s). Example 3: (Z)-Methyl 3-(((4-((2-

(dimethylamino)ethoxy)carbamoyl)phenyl)amino)(phenyl)meth ylene)-2-oxoindoline-6- carboxylate

Intermediate I: (Z)-Methyl 1 -acetyl-3-(((4-(iert- but

A mixture of (£)-methyl 1-acetyl-3-(ethoxy(phenyl)methylene)-2-oxoindoline-6-carboxy late (Intermediate A) (4.00 g, 1 1.0 mmol) and te/f-butyl 4-aminobenzoate (2.12 g, 1 1.0 mmol) in DMF (20 mL) was heated at 100 °C for 16 h. After cooling to rt, the precipitate was collected by filtration, washed with Et20 (100 mL) and dried in vacuo to afford the subtitle compound (Z)-methyl 1-acetyl-3-(((4-(fe/f-butoxycarbonyl)phenyl)amino)(phenyl)me thylene)-2- oxoindoline-6-carboxylate as a yellow solid (3.68 g, 59%); R' 3.17 min (Method 1); m/z 513 (M+H) + (ES + ). Intermediate J: (Z)-4-(((1 -Acetyl-6-(methoxycarbonyl)-2-oxoindolin-3- ylidene)(

To a solution of (Z)-methyl 1-acetyl-3-(((4-(te/f- butoxycarbonyl)phenyl)amino)(phenyl)methylene)-2-oxoindoline -6-carboxylate (Intermediate I) (3.67 g, 7.16 mmol) in DCM (75 mL) was added TFA (5.52 mL, 71.6 mmol) and the mixture was stirred at rt for 16 h. The solvent was removed under reduced pressure to afford the subtitle compound (Z)-4-(((1-acetyl-6-(methoxycarbonyl)-2-oxoindolin-3- ylidene)(phenyl)methyl)amino)benzoic acid, trifluoroacetate adduct as a yellow solid (3.63 g, 84%); R* 2.59 min (Method 1); m/z 457 (M+H) + (ES + ).

Intermediate K: 2-(2-(Dimethylamino)ethoxy)isoindoline-1 ,3-dione

To a solution of 2-hydroxyisoindoline-1 ,3-dione (2.34 g, 14.3 mmol) and triphenylphosphine (3.75 g, 14.3 mmol) in THF (40 mL) at O °C was added 2-(dimethylamino)ethanol (1.20 mL, 1 1.9 mmol) dropwise. The mixture was then stirred at 0 °C for 30 min before DIAD (2.78 mL, 14.3 mmol) was added dropwise. The reaction was stirred for a further 30 min at 0 °C before being warmed to rt and stirred for 16 h. The solvent was removed under reduced pressure and the residue re-dissolved in EtOAc (75 mL). The organic layer was washed with saturated NaHCC>3 solution (2 x 50 mL) and then dried over Na2S0 4 , filtered and concentrated under reduced pressure to approximately 30 mL. The organic layer was cooled to 0 °C and cold 1 M aqueous HCI solution (20 mL) was added. On complete addition, the mixture was warmed to rt and stirred for 20 min. The layers were separated and the aqueous washed with Et20 (2 x 30 mL). The aqueous layer was then cooled to 0 °C and basified by slow addition of saturated NaHC03 solution before being extracted with CHC (3 x 50 mL). The organic extracts were combined and then dried over Na2S0 4 , filtered and concentrated under reduced pressure to afford the subtitle compound 2-(2-(dimethylamino)ethoxy)isoindoline- 1 ,3-dione as a yellow oil (2.29 g, 78%); 1 H NMR (CDC ) δ: 2.34 (6H, s), 2.76 (2H, t), 4.31 (2H, t), 7.71-7.77 (2H, overlapping m), 7.80-7.86 (2H, overlapping m).

Intermediate L: 2-(Aminooxy)- V, V-dimethylethanamine, 2HCI

To a solution of 2-(2-(dimethylamino)ethoxy)isoindoline-1 ,3-dione (Intermediate K) (2.29 g, 9.78 mmol) in MeOH/DCM (2: 1 , 30 mL) was added a 25% aqueous solution of hydrazine (3.68 mL, 29.3 mmol). The mixture was stirred at rt for 18 h. The mixture was diluted with Et20 (100 mL) and filtered through a phase separator. The filtrate was de-gassed before 4M HCI in dioxane (10 mL) was added. The mixture was stirred at rt for 1 h. The solvent was removed under reduced pressure and the residue triturated with Et20 (2 x 50 mL) to afford the subtitle compound 2-(aminooxy)-A/,A/-dimethylethanamine, 2HCI as a white solid (891 mg, 50%); 1 H NMR δ: 2.79 (6H, s), 3.42 (2H, m), 4.45 (2H, m), 11.13 (4H, br s).

(Z)-Methyl 3-(((4-((2-

(dimethylamino)ethoxy)carbamoyl)phenyl)amino)(phenyl)meth ylene)-2-oxoindoline-6-

To a solution of (Z)-4-(((1-acetyl-6-(methoxycarbonyl)-2-oxoindolin-3- ylidene)(phenyl)methyl)amino)benzoic acid, trifluoroacetate adduct (Intermediate J) (0.050 g, 0.088 mmol), 2-(aminooxy)-A/,A/-dimethylethanamine, 2HCI (Intermediate L) (0.031 g, 0.18 mmol) and HATU (0.067 g, 0.18 mmol) in DMF (2 mL) was added Hunig's base (0.077 mL, 0.44 mmol) dropwise. The mixture was stirred at rt for 2 h before piperidine (0.087 mL, 0.876 mmol) was added. The mixture was stirred at rt for 2 h before the solvent was removed under reduced pressure. The residue was re-dissolved in MeOH (5 mL) and filtered through an SCX column, washing with MeOH (100 mL). The filtrate was discarded before the column was washed with 1 % NH3 in MeOH (100 mL). The solvent was removed under reduced pressure. The crude product was purified by preparative HPLC (Method B, 35-65% MeCN in water) to afford the title compound (Z)-methyl 3-(((4-((2-

(dimethylamino)ethoxy)carbamoyl)phenyl)amino)(phenyl)meth ylene)-2-oxoindoline-6- carboxylate as a yellow solid (13 mg, 28%); R' 1.80 min (Method 2); m/z 501 (M+H) + (ES + ); 1 H NMR δ: 2.20 (6H, s), 2.54 (2H, m), 3.79 (3H, s), 3.94 (2H, t), 5.90 (1 H, d), 6.86 (2H, m), 7.20 (1 H, dd), 7.45 (1 H, d), 7.48-7.68 (7H, overlapping m), 9.41 (1 H, s), 10.73 (1 H, s), 12.28 (1 H, s). Example 4: (Z)-Methyl 3-(((4-((2-(4-methylpiperazin-1 -

To a solution of (Z)-4-(((1-acetyl-6-(methoxycarbonyl)-2-oxoindolin-3- ylidene)(phenyl)methyl)amino)benzoic acid, trifluoroacetate adduct (Intermediate J) (100 mg, 0.18 mmol), 0-(2-(4-methylpiperazin-1-yl)ethyl)hydroxylamine (Intermediate F) (28 mg, 0.18 mmol) and HATU (73 mg, 0.19 mmol) in DMF (3 mL) was added Hunig's base (0.092 mL, 0.53 mmol) dropwise. The mixture was stirred at rt for 18 h. Piperidine (0.17 mL, 1.75 mmol) was added. The mixture was stirred at rt for 1.5 h before the majority of the solvent was removed under reduced pressure. The residue was partitioned between EtOAc (10 mL) and water/saturated NaHCC>3 solution (1 : 1 , 10 mL). The layers were separated and the aqueous layer extracted with EtOAc (3 x 10 mL). The organic extracts were combined and then dried over Na2S0 4 , filtered and concentrated under reduced pressure. The residue was re- dissolved in MeOH (10 mL) and filtered through an SCX column, washing with MeOH (100 mL). The filtrate was discarded before the column was washed with 1 % NH3 in MeOH (100 mL). The solvent was removed under reduced pressure. The crude product was purified by flash column chromatography (Si0 2 , 24 g, 10% MeOH in DCM then 1 % NH 3 in MeOH) to afford the title compound (Z)-methyl 3-(((4-((2-(4-methylpiperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoind oline-6-carboxylate as a yellow solid (64 mg, 64%); R< 1.45 min (Method 1); m/z 556 (M+H) + (ES + ); 1 H NMR δ: 2.13 (3H, s), 2.20-2.47 (8H, overlapping m), 2.54 (2H, t), 3.77 (3H, s), 3.92 (2H, t), 5.87 (1 H, d), 6.86 (2H, m), 7.21 (1 H, dd), 7.42 (1 H, d), 7.49-7.56 (4H, overlapping m), 7.57-7.68 (3H, overlapping m), 11.03 (1 H, s), 12.28 (1 H, s). Example 5: (Z)-Methyl 3-(((4-((2-(dimethylamino)ethoxy)carbamoyl)-3- methylphenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline -6-carboxylate

Intermediate M : (Z)-Methyl 1 -acetyl-3-(((4-(ierf-butoxycarbonyl)-3- met

A mixture of (£)-methyl 1-acetyl-3-(ethoxy(phenyl)methylene)-5-methyl-2-oxoindoline- 6- carboxylate (Intermediate D) (1.11 g, 2.91 mmol) and te/f-butyl 4-amino-2-methylbenzoate (604 mg, 2.91 mmol) in DMF (9 mL) was heated at 100 °C for 18 h. After cooling to rt, the precipitate was collected by filtration, washed with Et20 (10 mL) and dried in vacuo to afford the subtitle compound (Z)-methyl 1-acetyl-3-(((4-(te/f-butoxycarbonyl)-3- methylphenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline -6-carboxylate as a yellow solid (1.05 g, 65%); R' 3.28 min (Method 1); m/z 541 (M+H) + (ES + ); 1 H NMR δ: 1.49 (9H, s), 2.13 (3H, s), 2.33 (3H, s), 2.73 (3H, s), 3.77 (3H, s), 5.53 (1 H, s), 6.74 (1 H, dd), 6.92 (1 H, m), 7.43-7.56 (3H, overlapping m), 7.57-7.71 (3H, overlapping m), 8.67 (1 H, s), 11.87 (1 H, s).

Intermediate N: (Z)-4-(((1 -Acetyl-6-(methoxycarbonyl)-5-methyl-2-oxoindolin-3- ylidene)(

To a solution of (Z)-methyl 1-acetyl-3-(((4-(te/f-butoxycarbonyl)-3- methylphenyl)amino)(phenyl)methylene)-5-methyl-2-oxoindoline -6-carboxylate (Intermediate M) (1.0 g, 1.85 mmol) in DCM (14 mL) was added TFA (1.42 mL, 18.5 mmol) and the mixture was stirred at rt for 16 h. The solvent was removed under reduced pressure to afford the subtitle compound (Z)-4-(((1 -acetyl-6-(methoxycarbonyl)-5-methyl-2-oxoindolin-3- ylidene)(phenyl)methyl)amino)-2-methylbenzoic acid, trifluoroacetate adduct as a yellow solid (1.1 g, 97%); R' 2.85 min (Method 1); m/z 485 (M+H) + (ES + ); 1 H NMR δ: 2.13 (3H, s), 2.36 (3H, s), 2.73 (3H, s), 3.77 (3H, s), 5.53 (1 H, s), 6.74 (1 H, dd), 6.91 (1 H, m), 7.48-7.56 (2H, overlapping m), 7.55-7.73 (4H, overlapping m), 8.68 (1 H, s), 1 1.89 (1 H, s).

(Z)-Methyl 3-(((4-((2-(dimethylamino)ethoxy)carbamoyl)-3-

To a solution of (Z)-4-(((1-acetyl-6-(methoxycarbonyl)-5-methyl-2-oxoindolin- 3- ylidene)(phenyl)methyl)amino)-2-methylbenzoic acid, trifluoroacetate adduct (Intermediate N) (400 mg, 0.668 mmol), 2-(aminooxy)-A/,A/-dimethylethanamine, 2HCI (166 mg, 0.936 mmol) (Intermediate L) and HATU (305 mg, 0.802 mmol) in DMF (20 mL) was added Hunig's base (0.584 mL, 3.34 mmol) dropwise. The mixture was stirred at rt for 2 h. Piperidine (0.662 mL, 6.68 mmol) was added. The mixture was stirred at rt for 16 h before the majority of the solvent was removed under reduced pressure. The residue was partitioned between 10% MeOH in DCM solution (10 mL) and water/saturated NaHC0 3 solution (1 : 1 , 10 mL). The layers were separated and the aqueous extracted with 10% MeOH in DCM solution (3 x 10 mL). The organic extracts were combined and then filtered through an SCX column, washing with MeOH (50 mL). The filtrate was discarded before the column was washed with 1 % NH3 in MeOH (50 mL). The solvent was removed under reduced pressure. The crude product was purified by flash column chromatography (S1O2, 24 g, 0-10% MeOH in DCM then 1 % NH 3 in MeOH) to afford the title compound (Z)-methyl 3-(((4-((2- (dimethylamino)ethoxy)carbamoyl)-3-methylphenyl)amino)(pheny l)methylene)-5-methyl-2- oxoindoline-6-carboxylate as a yellow solid (100 mg, 28%); R l 1.66 min (Method 1); m/z 529 (M+H) + (ES + ); 1 H NMR δ: 2.13 (3H, s), 2.17 (9H, s), 2.44-2.48 (2H, overlapping m), 3.75 (3H, s), 3.91 (2H, t), 5.58 (1 H, s), 6.59 (1 H, dd), 6.78 (1 H, m), 7.08 (1 H, d), 7.36 (1 H, s), 7.53 (2H, m), 7.57-7.71 (3H, overlapping m), 10.86 (1 H, s), 12.18 (1 H, s). Example 6: (Z)-Methyl 5-methyl-3-(((4-((2-(4-methylpiperazin-1 - yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoind oline-6-carboxylate Intermediate O: (Z)-Methyl 1 -acetyl-3-(((4-(iert- butoxycarbonyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-ox oindoline-6- ca

A mixture of (£)-methyl 1-acetyl-3-(ethoxy(phenyl)methylene)-5-methyl-2-oxoindoline- 6- carboxylate (Intermediate D) (1.00 g, 2.64 mmol) and te/f-butyl 4-aminobenzoate (509 mg, 2.64 mmol) in DMF (9 mL) was heated at 100 °C for 18 h. After cooling to rt, the precipitate was collected by filtration, washed with Et20 (10 mL) and dried in vacuo to afford the subtitle compound (Z)-methyl 1-acetyl-3-(((4-(fe/f-butoxycarbonyl)phenyl)amino)(phenyl)me thylene)- 5-methyl-2-oxoindoline-6-carboxylate as a yellow solid (1.20 g, 86%); R l 3.23 min (Method 1); m/z 527 (M+H) + (ES + ). Intermediate P: (Z)-4-(((1 -Acetyl-6-(methoxycarbonyl)-5-methyl-2-oxoindolin-3- ylidene)(phenyl)methyl)amino)benzoic acid, trifluoroacetate adduct

To a solution of (Z)-methyl 1-acetyl-3-(((4-(te/f- butoxycarbonyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-ox oindoline-6-carboxylate

(Intermediate O) (1.20 g, 2.28 mmol) in DCM (14 mL) was added TFA (1.76 mL, 22.8 mmol) and the mixture was stirred at rt for 72 h. The solvent was removed under reduced pressure to afford the subtitle compound (Z)-4-(((1-acetyl-6-(methoxycarbonyl)-5-methyl-2-oxoindolin- 3-ylidene)(phenyl)methyl)amino)benzoic acid, trifluoroacetate adduct as a yellow solid (1.00 g, 72%); R< 2.72 min (Method 1); m/z 471 (M+H) + (ES + ); 1 H NMR δ: 2.13 (3H, s), 2.73 (3H, s), 3.78 (3H, s), 5.54 (1 H, s), 7.04 (2H, m), 7.50 (2H, m), 7.57-7.76 (5H, overlapping m), 8.68 (1 H, s), 1 1.89 (1 H, s), 12.89 (1 H, s).

(Z)-Methyl 5-methyl-3-(((4-((2-(4-methylpiperazin-1 -

To a solution of (Z)-4-(((1-acetyl-6-(methoxycarbonyl)-5-methyl-2-oxoindolin- 3- ylidene)(phenyl)methyl)amino)benzoic acid, trifluoroacetate adduct (Intermediate P) (57 mg, 0.098 mmol), 0-(2-(4-methylpiperazin-1-yl)ethyl)hydroxylamine (Intermediate F) (16 mg,

0.098 mmol) and HATU (41 mg, 0.107 mmol) in DMF (3 mL) was added Hunig's base (0.051 mL, 0.293 mmol) dropwise. The mixture was stirred at rt for 2 h. Piperidine (0.097 mL, 0.975 mmol) was added. The mixture was stirred at rt for 2 h before the majority of the solvent was removed under reduced pressure. The residue was partitioned between EtOAc (10 mL) and water/saturated NaHCC>3 solution (1 : 1 , 10 mL). The layers were separated and the aqueous extracted with EtOAc (3 x 10 mL). The organic extracts were combined and then filtered through an SCX column, washing with MeOH (50 mL). The filtrate was discarded before the column was washed with 1 % NH3 in MeOH (50 mL). The solvent was removed under reduced pressure. The crude product was purified by flash column chromatography (S1O2, 12 g, 0-10% MeOH in DCM then 1 % NH 3 in MeOH) to afford the title compound (Z)-methyl 5- methyl-3-(((4-((2-(4-methylpiperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoind oline-6-carboxylate as a yellow solid (36 mg, 64%); R' 1.49 min (Method 1); m/z 570 (M+H) + (ES + ); 1 H NMR δ: 2.13 (3H, s), 2.13 (3H, s), 2.17-2.35 (4H, overlapping m), 2.34-2.47 (4H, overlapping m), 2.54 (2H, t), 3.75 (3H, s), 3.92 (2H, t), 5.61 (1 H, s), 6.87 (2H, m), 7.36 (1 H, s), 7.47-7.56 (4H, overlapping m), 7.57-7.71 (3H, overlapping m), 10.88 (1 H, s), 1 1.52 (1 H, s), 12.22 (1 H, s). Example 7: (Z)-Methyl 3-(((4-((2-(4-(2-hydroxyethyl)piperazin-1 - yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-5-methyl -2-oxoindoline-6- carboxylate, formate

Intermediate Q: -(2-Bromoethoxy)isoindoline-1 ,3-dione

2-Hydroxyisoindoline-1 ,3-dione (5.00 g, 30.7 mmol), tnethylamine (9.40 mL, 67.4 mmol) and 1 ,2-dibromoethane (12.4 mL, 144 mmol) were combined in DMF (32 mL) and stirred at rt for 16 h. The precipitate was removed by filtration, washing with DMF (3 x 5m L) and the solid was discarded. The filtrate was diluted with water (250 mL) and the product was extracted with EtOAc (50 mL). The organic layer was washed with 1 M aqueous HCI solution (50 mL) and water (50mL), dried over MgSCU, filtered and concentrated under reduced pressure to afford the subtitle compound 2-(2-bromoethoxy)isoindoline-1 ,3-dione as a cream solid (6.28 g, 68%); 1 H NMR δ: 3.75 (2H, t), 4.45 (2H, t), 7.82-7.91 (4H, overlapping m). Intermediate R: 0-(2-Bromoethyl)hydroxylamine, HBr

2-(2-Bromoethoxy)isoindoline-1 ,3-dione (Intermediate Q) (1.00 g, 3.70 mmol) was combined with cone HBr (48 wt% in water, 4.5 mL) and acetic acid (3 mL, 52.4 mmol). The mixture was heated at 120 °C for 15 min. The reaction mixture was cooled, the precipitate was removed by filtration and the filtrate concentrated under reduced pressure to afford the subtitle compound 0-(2-Bromoethyl)hydroxylamine, HBr as a tan solid (550 mg, 74%); 1 H NMR δ: 3.72 (2H, m), 4.28 (2H, m), 10.70 (3H, br s).

Intermediate S: (Z)-Methyl 1 -acetyl-3-(((4-((2- bromoethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-5-meth yl-2-oxoindoline-6- carboxylate

To a solution of (Z)-4-(((1-acetyl-6-(methoxycarbonyl)-5-methyl-2-oxoindolin- 3- ylidene)(phenyl)methyl)amino)benzoic acid, trifluoroacetate adduct (Intermediate P) (300 mg, 0.638 mmol), HATU (291 mg, 0.765 mmol) and Hunig's base (334 μΙ_, 1.91 mmol) in DMF (2 mL) was added 0-(2-bromoethyl)hydroxylamine, HBr (Intermediate R) (211 mg, 0.956 mmol). The mixture was stirred at rt for 10 min. The reaction mixture was diluted with 10% MeOH in DCM and washed with a saturated aqueous ammonium chloride solution. The organic layer was dried over MgSCU and the solvent was evaporated under reduced pressure to afford (Z)-Methyl 1-acetyl-3-(((4-((2- bromoethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-5-meth yl-2-oxoindoline-6- carboxylate; R' 2.67 min (Method 1); m/z 592/594 (M+H) + (ES + ). The crude material was used directly in the next step assuming quantitative yield.

(Z)-Methyl 3-(((4-((2-(4-(2-hydroxyethyl)piperazin-1 - yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-5-methyl -2-oxoindoline-6-

(Z)-Methyl 1-acetyl-3-(((4-((2-bromoethoxy)carbamoyl)phenyl)amino)(phen yl)methylene)-5- methyl-2-oxoindoline-6-carboxylate (Intermediate S) (100 mg, 0.169 mmol) was combined with 2-(piperazin-1-yl)ethanol (659 mg, 5.06 mmol) and the mixture was stirred at rt for 16 h. The reaction mixture was diluted with 10% MeOH in DCM (10 mL) and washed with water (5 mL). The aqueous layer was further extracted with 10% MeOH in DCM (10 mL) and the combined organic layers were concentrated under reduced pressure. The crude product was purified by preparative HPLC (Method A, 20-50% MeCN in water) to afford the title compound (Z)-methyl 3-(((4-((2-(4-(2-hydroxyethyl)piperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-5-methyl -2-oxoindoline-6- carboxylate, formate as a yellow solid (25 mg, 23%); R' 1.86 min (Method 2); m/z 600 (M+H) + (ES + ); 1 H NMR δ: 2.13 (3H, s), 2.30-2.50 (8H, overlapping m), 2.36 (2H, t), 2.54 (2H, m), 3.47 (2H, t), 3.75 (3H, s), 3.92 (2H, t), 5.61 (1 H, s), 6.87 (2H, m), 7.36 (1 H, s), 7.50-7.56 (4H, overlapping m), 7.57-7.70 (4H, overlapping m), 8.17 (1 H, s), 10.89 (1 H, s), 12.21 (1 H, s). Example 8: (Z)-Methyl 2-oxo-3-(phenyl((4-((2-(piperidin-1 - y

To a solution of (Z)-4-(((1-acetyl-6-(methoxycarbonyl)-2-oxoindolin-3- ylidene)(phenyl)methyl)amino)benzoic acid, trifluoroacetate adduct (Intermediate J) (100 mg, 0.2 mmol), HATU (80 mg, 0.21 mmol) and Hunig's base (0.092 ml, 0.526 mmol) in DMF (3 ml_) was added 0-(2-bromoethyl)hydroxylamine, HBr (Intermediate R) (27 mg, 0.123 mmol). The mixture was stirred at rt for 1 h. More 0-(2-(4-methylpiperazin-1-yl)ethyl)hydroxylamine, HBr (20 mg) was added. The reaction mixture was stirred for a another hour. Half of the reaction mixture was taken out and added to 0.35 ml_ of piperidine and the reaction mixture was stirred for 16 h. The solvent was evaporated and the residue was purified by preparative HPLC (Method A, 15-70% MeCN in water) to afford the title compound (Z)-methyl 2-oxo-3- (phenyl((4-((2-(piperidin-1-yl)ethoxy)carbamoyl)phenyl)amino )methylene)indoline-6- carboxylate, 0.7 formic acid (1.6 mg, 2%). R< 1.66 min (Method 1); m/z 541 (M+H) + (ES + ); 1 H NMR δ 1.33-1.42 (2H, overlapping m), 1.45-1.52 (4H, overlapping m), 2.40-2.47 (4H, overlapping m), 2.57 (2H, t), 3.77 (3H, s), 3.94 (2H, t), 5.87 (1 H, d), 6.86 (2H, m), 7.21 (1 H, dd), 7.42 (1 H, d), 7.50-7.56 (4H, overlapping m), 7.57-7.69 (3H, overlapping m), 8.19 (0.7 H, s), 11.04 (1 H, s), 12.27 (1 H, s). Example 9: (Z)-Methyl 5-methyl-3-(((4-(methyl(2-(4-(2-(methylamino)-2- oxoethyl)piperazin-1 -yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2- oxoindoline-6-carboxylate

Intermediate T: Formaldehyde 0-(2-hydroxyethyl) oxime

Paraformaldehyde

.O^ /\ EtOH o

H 2 N OH " ^^OH

A mixture of 2-(aminooxy)ethanol (3.16 g, 41.0 mmol) and paraformaldehyde (1.23 g, 41.0 mmol) in EtOH (50 ml_) was heated under reflux for 18 h. The solvent was removed under reduced pressure to afford the subtitle compound formaldehyde 0-(2-hydroxyethyl) oxime as a colourless oil (3.56 g, 97%); 1 H NMR δ: 3.57 (2H, q), 4.05-3.96 (2H, m), 4.67 (1 H, t), 6.57 (1 H, d), 7.05 (1 H, d).

Intermediate U: 2-((Methylamino)oxy)ethanol

Py-BH 3 , THF

HCI in dioxane

To a solution of formaldehyde 0-(2-hydroxyethyl) oxime (Intermediate T) (3.56 g, 40.0 mmol) in THF (25 ml_) at 0 °C was added Py-BH 3 (6.05 ml, 59.9 mmol). 4M HCI in dioxane (15 ml_) was added over 1 h. The mixture was warmed to rt and stirred for 24 h, then was slowly diluted with MeOH (30 ml_) and filtered through an SCX column, washing with MeOH (250 ml_). The filtrate was discarded before the column was washed with 1 % NH3 in MeOH (250 ml_). The solvent was removed under reduced pressure to afford the subtitle compound 2- ((methylamino)oxy)ethanol as a colourless oil (0.833 g, 18%); 1 H NMR δ: 2.51 (3H, m), 3.50 (2H, m), 3.57 (2H, m), 4.53 (1 H, s), 6.45 (1 H, m).

Intermediate V: (Z)-Methyl 1 -acetyl-3-(((4-((2- hydroxyethoxy)(methyl)carbamoyl)phenyl)amino)(phenyl)methyle ne)-5-methyl-2-

To a solution of (Z)-4-(((1-acetyl-6-(methoxycarbonyl)-5-methyl-2-oxoindolin- 3- ylidene)(phenyl)methyl)amino)benzoic acid, trifluoroacetate adduct (Intermediate P) (0.528 g, 0.903 mmol), 2-((methylamino)oxy)ethanol (Intermediate U) (0.158 g, 1.36 mmol) and HATU (0.378 g, 0.99 mmol) in DMF (3 ml_) was added Hunig's base (0.47 ml, 2.71 mmol) dropwise. The mixture was stirred at rt for 2 h. The solvent was removed under reduced pressure and the residue was partitioned between 10% MeOH in EtOAc (10 ml_) and water (10 ml_). The layers were separated and the aqueous extracted with EtOAc (3 x 10 ml_). The organic extracts were combined and then dried over Na2S0 4 , filtered and concentrated under reduced pressure. The crude product was purified by flash column chromatography (S1O2, 24 g, 0-5% MeOH in DCM, gradient elution) to afford the subtitle compound (Z)-methyl 1-acetyl- 3-(((4-((2-hydroxyethoxy)(methyl)carbamoyl)phenyl)amino)(phe nyl)methylene)-5-methyl-2- oxoindoline-6-carboxylate as a yellow solid (0.482 g, 88%); R' 2.38 min (Method 1); m/z 544 (M+H) + (ES + ).

Intermediate W: (Z)-Methyl 1 -acetyl-3-(((4-((2- bromoethoxy)(methyl)carbamoyl)phenyl)amino)(phenyl)methylene )-5-methyl-2-

To a solution of (Z)-methyl 1-acetyl-3-(((4-((2- hydroxyethoxy)(methyl)carbamoyl)phenyl)amino)(phenyl)methyle ne)-5-methyl-2-oxoindoline- 6-carboxylate (Intermediate V) (0.482 g, 0.887 mmol) in DCM (10 ml_) at 0 °C were added triphenylphosphine (0.256 g, 0.975 mmol) and CBr 4 (0.323 g, 0.975 mmol). The mixture was warmed to rt and stirred for 2 h. The solvent was removed under reduced pressure and the crude product was purified by flash column chromatography (S1O2, 24 g, 0-2% MeOH in DCM, gradient elution) to afford the subtitle compound (Z)-methyl 1-acetyl-3-(((4-((2- bromoethoxy)(methyl)carbamoyl)phenyl)amino)(phenyl)methylene )-5-methyl-2-oxoindoline- 6-carboxylate as an orange solid (681 mg, quant.); R' 2.80 min (Method 2); m/z 606/608 (M+H) + (ES + ).

(Z)-Methyl 5-methyl-3-(((4-(methyl(2-(4-(2-(methylamino)-2-oxoethyl)pip erazin-1 -

To a mixture of (Z)-methyl 1-acetyl-3-(((4-((2- bromoethoxy)(methyl)carbamoyl)phenyl)amino)(phenyl)methylene )-5-methyl-2-oxoindoline- 6-carboxylate (Intermediate W) (75 mg, 0.124 mmol) and A/-methyl-2-(piperazin-1- yl)acetamide, 2HCI (142 mg, 0.618 mmol) in DMF (2 mL) was added Hunig's base (0.259 ml, 1.48 mmol). The mixture was heated at 60 °C for 5 h. After cooling to rt, piperidine (0.245 ml, 2.47 mmol) was added and the mixture was heated at 60 °C for 16 h. The reaction mixture was cooled to rt and the solvent was removed under reduced pressure. The residue was partitioned between EtOAc (10 mL) and water/saturated aqueous NaHCC>3 solution (1 :1 , 10 mL). The layers were separated and the aqueous phase was extracted with EtOAc (3 x 10 mL). The organic extracts were combined and filtered directly through an SCX column, washing with MeOH (50 mL). The filtrate was discarded before the column was washed with 1 % NH3 in MeOH (50 mL). The solvent was removed under reduced pressure and the crude product was purified by flash column chromatography (S1O2, 12 g, 0-10% 0.7 M ammonia in MeOH in DCM, gradient elution) to afford the title compound (Z)-methyl 5-methyl-3-(((4- (methyl(2-(4-(2-(methylamino)-2-oxoethyl)piperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2-oxoind oline-6-carboxylate as a yellow solid (11 mg, 13%); R' 1.58 min (Method 1); m/z 641 (M+H) + (ES + ); 1 H NMR δ: 2.14 (3H, s), 2.23-2.36 (8H, overlapping m), 2.59 (3H, d), 2.87 (2H, m), 3.21 (3H, s), 3.31 (2H, s), 3.75 (3H, s), 3.78 (2H, t), 5.62 (1 H, s), 6.86 (2H, m), 7.36 (1 H, s), 7.42 (2H, m), 7.53 (2H, m), 7.58-7.66 (3H, overlapping m), 10.89 (1 H, s), 12.22 (1 H, s).

Example 10: (Z)-Methyl 5-methyl-3-(((4-(methyl(2-(4-(2-(methylamino)-2- oxoethyl)piperazin-1 -yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2- oxoindoline-6-carboxylate

Intermediate X: ferf-Butyl (4-(/V-(2-hydroxyethoxy)-/V- methylsulfamoyl)p

To a solution of 2-((methylamino)oxy)ethanol (Intermediate U) (0.1 16 g, 1.27 mmol) in DMF (3 mL) was added Hunig's base (0.445 ml, 2.55 mmol) followed by te/f-butyl (4- (chlorosulfonyl)phenyl)carbamate (0.371 g, 1.27 mmol). The mixture was stirred at rt for 16 h. The solvent was removed under reduced pressure and the residue was partitioned between DCM (10 mL) and 1 M aqueous HCI solution (10 mL). The layers were separated and the aqueous extracted with DCM (2 x 10 mL). The organic extracts were combined and filtered through a phase separator. The solvent was removed under reduced pressure and the crude product was purified by flash column chromatography (S1O2, 24 g, 0-70% EtOAc in hexanes, gradient elution) to afford the subtitle compound te/f-butyl (4-(/V-(2-hydroxyethoxy)-/V- methylsulfamoyl)phenyl)carbamate as a pink gum, which solidified on standing overnight (246 mg, 41 %); R< 1.97 min (Method 2); m/z 347 (M+H) + (ES + ); 1 H NMR δ: 1.49 (9H, s), 2.71 (3H, s), 3.54 (2H, q), 3.93 (2H, dd), 4.68 (1 H, t), 7.69-7.76 (4H, overlapping m), 9.97 (1 H, s).

Intermediate Y: 4-Amino-W-(2-hydroxyethoxy)- V-methylbenzenesulfonamide, trifluoroacetate s

To a solution of te/f-butyl (4-(/V-(2-hydroxyethoxy)-/V-methylsulfamoyl)phenyl)carbamate (Intermediate X) (202 mg, 0.583 mmol) in DCM (3 ml_) was added TFA (0.449 ml,

5.83 mmol). The mixture was stirred at rt for 4 h. The solvent was removed under reduced pressure and the residue was re-dissolved in MeOH (3 ml_), and filtered through an SAX column, washing with MeOH (75 ml_). The solvent was removed under reduced pressure to afford the subtitle compound 4-amino-A/-(2-hydroxyethoxy)-A/-methylbenzenesulfonamide, T trifluoroacetate salt as as an orange gum (0.23 g, 95%); R' 1.13 min (Method 2); m/z 247 (M+H) + (ES + ).

Intermediate Z: (Z)-Methyl 1 -acetyl-3-(((4-( V-(2-hydroxyethoxy)- V- methylsulfamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-o xoindoline-6- c

A mixture of (£)-methyl 1-acetyl-3-(ethoxy(phenyl)methylene)-5-methyl-2-oxoindoline- 6- carboxylate (Intermediate D) (0.231 g, 0.61 mmol) and 4-amino-/V-(2-hydroxyethoxy)-/\/- methylbenzenesulfonamide, trifluoroacetate salt (Intermediate Y) (0.219 g, 0.608 mmol) in DMF (3 ml_) was treated with Hunig's base (0.106 ml, 0.608 mmol) and heated at 80 °C for 18 h. The solvent was removed under reduced pressure and the crude product was purified by flash column chromatography (S1O2, 24 g, 0-2% MeOH in DCM, gradient elution) to afford the subtitle compound (Z)-methyl 1-acetyl-3-(((4-(/V-(2-hydroxyethoxy)-/\/- methylsulfamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-o xoindoline-6-carboxylate as an orange gum (226 mg, 63%); R< 2.58 min (Method 2); m/z 580 (M+H) + (ES + ).

Intermediate AA: (Z)-Methyl 1 -acetyl-3-(((4-(N-(2-bromoethoxy)-/V- methylsulfamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-o xoindoline-6- c

To a solution of (Z)-methyl 1-acetyl-3-(((4-(/V-(2-hydroxyethoxy)-/\/- methylsulfamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-o xoindoline-6-carboxylate (0.226 g, 0.390 mmol) (Intermediate Z) in DCM (4 ml_) were added triphenylphosphine (0.112 g, 0.429 mmol) and CBr 4 (41 μΙ_, 0.429 mmol). The mixture was stirred at rt for 16 h. Further portions of PPh 3 (0.1 12 g, 0.429 mmol) and CBr 4 (41 μΙ_, 0.429 mmol) were added and the mixture was stirred at rt for 2 h. The solvent was removed under reduced pressure and the crude product was purified by flash column chromatography (S1O2, 12 g, 0-2% MeOH in DCM, gradient elution) to afford the subtitle compound (Z)-methyl 1-acetyl-3-(((4- (A/-(2-bromoethoxy)-/\/-methylsulfamoyl)phenyl)amino)(phenyl )methylene)-5-methyl-2- oxoindoline-6-carboxylate as a yellow solid (161 mg, 64%); R' 3.01 min (Method 2); m/z 642/644 (M+H) + (ES + ).

(Z)-Methyl 3-(((4-( V-(2-((2-hydroxyethyl)(methyl)amino)ethoxy)- V- methylsulfamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-o xoindoline-6-

A solution of (Z)-methyl 1-acetyl-3-(((4-(/V-(2-bromoethoxy)-/\/- methylsulfamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-o xoindoline-6-carboxylate (Intermediate AA) (77 mg, 0.120 mmol) and 2-(methylamino)ethanol (0.096 ml, 1.20 mmol) in DMF (2 mL) was heated at 60 °C for 16 h. After cooling to rt, the solvent was removed under reduced pressure and the residue was partitioned between EtOAc (10 mL), and

water/saturated aqueous NaHCC>3 solution (1 : 1 , 10 mL). The layers were separated and the aqueous extracted with EtOAc (3 x 10 mL). The organic extracts were combined and filtered through an SCX column, washing with MeOH (50 mL). The filtrate was discarded before the column was washed with 1 % NH3 in MeOH (50 mL). The solvent was removed under reduced pressure and the crude product was purified by flash column chromatography (S1O2, 12 g, 0-10% 0.7 M ammonia in MeOH in DCM) to afford the title compound (Z)-methyl 3- (((4-(/V-(2-((2-hydroxyethyl)(methyl)amino)ethoxy)-/\/- methylsulfamoyl)phenyl)amino)(phenyl)methylene)-5-methyl-2-o xoindoline-6-carboxylate as a yellow solid (41 mg, 55%); R< 1.72 min (Method 1); m/z 595 (M+H) + (ES + ); 1 H NMR δ: 2.14 (3H, s), 2.17-2.30 (4H, overlapping m), 2.68 (3H, s), 3.43-3.54 (2H, overlapping m), 3.76 (3H, s), 3.96-4.08 (2H, overlapping m), 4.40 (1 H, m), 5.65 (1 H, s), 6.96 (2H, m), 7.36 (1 H, s), 7.53-7.75 (7H, overlapping m), 10.96 (1 H, s), 12.29 (1 H, s), (Missing 3H-presumed overlap with solvent).

The following compound examples (Table 2) may be prepared by similar synthetic methods to the aforementioned examples or by methods described elsewhere herein:

Table 2: Additional Compound Examples of the Invention

*Route code index:

Route 1A: see Example 1 or Example 2

Route 1 B: see Example 3, Example 4, Example 5 or Example 6

Route 1 C: see Example 7 or Example 8

Route 1 D: see Example 9

Route 1 E: see Example 10 Example No., Structure, Name, Route code, LC-MS Analysis and 1 H NMR Spectral Data Example 11 : , ,

(Z)-Methyl 3-(((4-(/V-methoxy-/V- methylsulfamoyl)phenyl)amino)(phenyl)methylene)-2- oxoindoline-6-carboxylate

Route code*: 1A

Example 12:

R' 1.56 min (Method 1); m/z 543 (M+H) + (ES + ); 1 H NMR δ: 2.39-2.46 (4H, overlapping m), 2.54 (2H, t), 3.50-3.58 (4H, overlapping m), 3.77 (3H, s), 3.94 (2H, t), 5.87 (1 H, d), 6.86 (2H, d), 7.21 (1 H, dd), 7.42 (1 H, d), 7.50-7.56 (4H, overlapping

m), 7.57-7.68 (3H, overlapping m),

(Z)-Methyl 3-(((4-((2- 1 1.05 (1 H, s), 11.59 (1 H, s), 12.27 morpholinoethoxy)carbamoyl)phenyl)amino)(phenyl)m

(1 H, s).

ethylene)-2-oxoindoline-6-carboxylate

Route code*: 1 B

R' 1.56 min (Method 1); m/z 515 (M+H) + (ES + ); 1 H NMR δ: 2.16 (9H, s), 2.47 (2H, m), 3.77 (3H, s), 3.90 (2H, t), 5.84 (1 H, m), 6.57 (1 H, dd), 6.77 (1 H, d), 7.07 (1 H, d), 7.20 (1 H, dd), 7.42 (1 H, d), 7.52 (2H,

m), 7.55-7.68 (3H, overlapping m),

(Z)-Methyl 3-(((4-((2- 1 1.02 (1 H, s), 12.25 (1 H, s).

(dimethylamino)ethoxy)carbamoyl)-3- methylphenyl)amino)(phenyl)methylene)-2- oxoindoline-6-carboxylate Route code*: 1 B

R' 1.50 min (Method 1); m/z 570 (M+H) + (ES + ); 1 H NMR δ: 2.13 (3H, s), 2.15-2.28 (8H, overlapping m), 2.27 (2H, t), 3.21 (3H, s), 3.75-3.78 (5H, overlapping m), 5.88 (1 H, d), 6.85 (2H, m), 7.21 (1 H, dd), 7.35-

7.46 (3H, overlapping m), 7.49-

(Z)-Methyl 3-(((4-(methyl(2-(4-methylpiperazin-1- 7.56 (2H, m), 7.55-7.69 (3H, yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene

overlapping m), 1 1.03 (1 H, s),

)-2-oxoindoline-6-carboxylate

12.29 (1 H, s).

Route code*: 1 B

Example 15:

(Z)-Methyl 3-(((4-(Λ/-(2- (dimethylamino)ethoxy)sulfamoyl)phenyl)ami

yl)methylene)-2-oxoindoline-6-carboxylate

Route code*: 1A

Example 16: R' 1.59 min (Method 1); m/z 557

(M+H) + (ES + ); 1 H NMR δ: 2.17 (3H, s), 2.40-2.45 (4H, overlapping m), 2.54 (2H, t), 3.54-3.56 (4H, overlapping m), 3.77 (3H, s), 3.95 (2H, t), 5.84 (1H, d), 6.57 (1H, dd),

6.78 (1H, d), 7.08 (1H, d), 7.20

(Z)-Methyl 3-(((3-methyl-4-((2- (1H, dd), 7.42 (1H, d), 7.51-7.53 morpholinoethoxy)carbamoyl)phenyl)amino)(phenyl)m

(2H, overlapping m), 7.56-7.70 (3H, ethylene)-2-oxoindoline-6-carboxylate

overlapping m), 11.02 (1H, s), 11.31 (1H, s), 12.25 (1H, s).

Route code*: 1 B

R' 1.46 min (Method 1); m/z 570 (M+H) + (ES + ); 1 H NMR δ: 2.17 (3H, s), 2.18 (3H, s), 2.28-2.49 (8H, overlapping m), 2.55 (2H, t), 3.77 (3H, s), 3.93 (2H, t), 5.84 (1H, d), 6.57 (1H, dd), 6.78 (1H, d), 7.08

(1H, d), 7.20 (1H, dd), 7.42 (1H, d),

(Z)-Methyl 3-(((3-methyl-4-((2-(4-methylpiperazin-1 - 7.47-7.56 (2H, overlapping m), yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylen)- 7.55-7.69 (3H, overlapping m),

2-oxoindoline-6-carboxylate

11.03 (1H, s), 11.25 (1H, s), 12.26 (1H, s).

Route code*: 1 B

R' 1.53 min (Method 1); m/z 584 (M+H) + (ES + ); 1 H NMR δ: 2.06 (3H, s), 2.11 (3H, s), 2.08-2.28 (10H, overlapping m), 3.18 (3H, s), 3.64- 3.72 (2H, overlapping m), 3.77 (3H, s), 5.87 (1H, d), 6.59 (1H, dd), 6.79 (Z)-Methyl 3-(((3-methyl-4-(methyl(2-(4- (1H, d), 7.02 (1H, d), 7.20 (1H, dd), methylpiperazin-1- 7.42 (1H, d), 7.51 (2H, m), 7.55- yl)ethoxy)carbamoyl)p enyl)amino)(phenyl)methylene 7.68 (3H, overlapping m), 11.01 )-2-oxoindoline-6-carboxylate (1H, s), 12.25 (1H, s).

Route code*: 1 B

Example 19:

R' 1.54 min (Method 1); m/z 515 (M+H) + (ES + ); 1 H NMR δ: 1.69 (2H, m), 2.11 (6H, s), 2.31 (2H, t), 3.77 (3H,s),3.84 (2H,t),5.87(1H, d), 6.86 (2H, m), 7.21 (1H, dd), 7.42 (1H, d), 7.50-7.55 (4H, overlapping m), 7.57-7.68 (3H, overlapping m),

11.03 (1H, s), 12.28 (1H, s).

(Z)-Methyl 3-(((4-((3-

(dimethylamino)propoxy)carbamoyl)phenyl)amino)(ph

enyl)methylene)-2-oxoindoline-6-carboxylate

Route code*: 1 B

Example 20:

R l 1.55 min (Method 1); m/z 519 (M+H) + (ES + ); 1 H NMR δ: 2.17 (6H, s), 2.46-2.48 (2H, overlapping m), 3.78 (3H, s), 3.90 (2H, t), 5.89 (1H, d), 6.58 (1H, d), 6.72-6.80 (1H, overlapping m), 7.22 (1H, dd), 7.35-7.44 (2H, overlapping m),

7.51-7.58 (2H, overlapping m),

(Z)-Methyl 3-(((4-((2- 7.59-7.72 (3H, m), 11.06 (1H, s),

(dimethylamino)ethoxy)carbamoyl)-3- 12.21 (1H, s).

fluorophenyl)amino)(phenyl)methylene)-2-oxoindoline-

6-carboxylate

Route code * : B

Example 21: R* 1.57 min (Method 1); m/z 541

(M+H) + (ES + ); 1 H NMR δ: 1.64-1.69 (2H, overlapping m), 1.82-1.87 (4H, overlapping m), 2.14 (3H, s), 2.79- 2.84 (2H, overlapping m), 3.45 (3H, s), 3.77 (3H, s), 3.80-3.90 (1 H, overlapping m), 5.89 (1 H, d), 6.82- 6.92 (2H, overlapping m), 7.21 (1 H, dd), 7.30-7.38 (2H, overlapping m),

(Z)-Methyl 3-(((4-(methoxy(1 -methylpiperidin-4- 7.42 (1 H, d), 7.50-7.53 (2H, yl)carbamoyl)phenyl)amino)(phenyl)methylene)-2

overlapping m), 7.54-7.67 (3H, oxoindoline-6-carboxylate

overlapping m), 1 1.02 (1 H, s), 12.26 (1 H, s).

Route code*: 1 B

R< 1.58 min (Method 1); m/z 529 (M+H) + (ES + ); 1 H NMR (major rotamer) δ: 1.97 (6H, s), 2.17 (3H, s), 3.17 (3H, s), 3.72-3.79 (5H, overlapping m), 5.86 (1 H, d), 6.60 (1 H, dd), 6.80 (1 H, d), 7.03 (1 H, d),

7.20 (1 H, dd), 7.42 (1 H, d), 7.46-

(Z)-Methyl 3-(((4-((2- 7.54 (2H, overlapping m), 7.55-

(dimethylamino)ethoxy)(methyl)carbamoyl)-3- 7.69 (3H, overlapping m), 10.99 methylphenyl)amino)(phenyl)methylene)-2- (1 H, s), 12.23 (1 H, s). (Missing 2H- oxoindoline-6-carboxylate

presumed overlap with solvent).

Route code*: 1 B

Example 23: R* 1.57 min (Method 1); m/z 515

(M+H) + (ES + ); 1 H NMR δ: 2.09 (6H, s), 2.43 (2H, t), 3.46 (3H, s), 3.66 (2H, t), 3.77 (3H, s), 5.87 (1 H, d), 6.86 (2H, m), 7.21 (1 H, dd), 7.37 (2H, m), 7.42(1 H, d), 7.51 (2H, m), 7.55-7.67 (3H, overlapping m), 1 1.02 (1 H, s), 12.27 (1 H, s).

(Z)-Methyl 3-(((4-((2-

(dimethylamino)ethyl)(methoxy)carbamoyl)phenyl)arni

no)(phenyl)methylene)-2-oxoindoline-6-carboxylate

Route code*: 1 B

Example 24:

R' 1.55 min (Method 1); m/z 513 (M+H) + (ES + ); 1 H NMR δ: 2.33 (3H, s), 2.59-2.61 (2H, overlapping m), 2.68-2.71 (2H, overlapping m), 3.77 (3H, s), 3.74-3.79 (4H, overlapping m), 5.87 (1 H, d), 6.85 (2H, m), 7.21 (1 H, dd), 7.37-7.45

(Z)-Methyl 3-(((4-(5-methyl-1 ,2,5-oxadiazepane-2- (3H, overlapping m), 7.51 (2H, m), carbonyl)phenyl)amino)(phenyl)methylene)-2- 7.56-7.70 (3H, overlapping m), oxoindoline-6-carboxylate 1 1.00 (1 H, s), 12.29 (1 H, s).

Route code*: 1 B

Example 25: R* 1.70 min (Method 1); m/z 606

(M+H) + (ES + ); 1 H NMR δ: 2.30-2.48 (6H, overlapping m), 2.67 (3H, s), 3.78 (3H, s), 4.00 (2H, t), 5.90 (1 H, d), 6.96 (2H, m), 7.23 (1 H, dd), 7.43 (1 H, d), 7.58 (2H, m), 7.61- 7.75 (5H, overlapping m), 11.09 (1 H, s), 12.37 (1 H, s). (Missing 7H- presumed overlap with solvent).

(Z)-Methyl 3-(((4-(A/-methyl-A/-(2-(4-methylpiperazin-1- yl)ethoxy)sulfamoyl)phenyl)amino)(phenyl)methylene)- 2-oxoindoline-6-carboxylate

Route code*: 1A

Example 26:

R* 1.95 min (Method 1); m/z 515 (M+H) + (ES + ); 1 H NMR δ: 2.13 (3H, s), 2.13 (3H, s), 2.17 (3H, s), 2.53 (2H, m), 3.75 (3H, s), 3.93 (2H, t), 5.58 (1 H, s), 6.58 (1 H, dd), 6.79 (1 H, d), 7.08 (1 H, d), 7.36 (1 H, s), 7.52 (2H, m), 7.57-7.71 (3H,

overlapping m), 10.87 (1 H, s),

(Z)-Methyl 3-(((4-((2- 1 1.31 (1 H, s), 12.19 (1 H, s).

(dimethylamino)ethoxy)carbamoyl)phenyl)amino)(phe

nyl)methylene)-5-methyl-2-oxoindoline-6-carboxylate

Route code*: 1 B

Example 27: R< 2.00 min (Method 1); m/z 529

(M+H) + (ES + ); 1 H NMR δ: 1.70 (2H, m), 2.09-2.18 (9H, overlapping m), 2.34 (2H, t), 3.75 (3H, s), 3.84 (2H, t), 5.61 (1 H, s), 6.88 (2H, m), 7.36 (1 H, s), 7.48-7.57 (4H, overlapping m), 7.58-7.70 (3H, overlapping m),

10.88 (1 H, s), 12.22 (1 H, s).

(Z)-Methyl 3-(((4-((3-

(dimethylamino)propoxy)carbamoyl)phenyl)amino)(ph

enyl)methylene)-5-methyl-2-oxoindoline-6-carboxylate

Route code*: 1 B

Example 28:

R* 1.93 min (Method 1); m/z 584 (M+H) + (ES + ); 1 H NMR δ: 2.13 (3H, s), 2.18 (6H, s), 3.75 (3H, s), 3.91 (2H, t), 5.61 (1 H, s), 6.87 (2H, m), 7.36 (1 H, s), 7.49-7.57 (4H, overlapping m), 7.57-7.70 (3H, overlapping m), 10.89 (1 H, s),

12.22 (1 H, s), (Missing 10H-

(Z)-Methyl 5-methyl-3-(((3-methyl-4-((2-(4- presumed overlap with solvent). methylpiperazin-1- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene

)-2-oxoindoline-6-carboxylate

Route code*: 1 B

Example 29:

R< 2.02 min (Method 1); m/z 543 (M+H) + (ES + ); 1 H NMR δ: 1.69 (2H, m), 2.12 (6H, s), 2.13 (3H, s), 2.16 (3H, s), 2.31 (2H, t), 3.75 (3H, s), 3.85 (2H, t), 5.58 (1 H, s), 6.59 (1 H, dd), 6.79 (1 H, d), 7.07 (1 H, d), 7.36 (1 H, s), 7.52 (2H, dd), 7.57-7.71

(3H, overlapping m), 10.87 (1 H, s), 12.18 (1 H, s). (Z)-Methyl 3-(((4-((3-

(dimethylamino)propoxy)carbamoyl)-3- methylphenyl)amino)(phenyl)methylene)-5-methyl-2- oxoindoline-6-carboxylate

Route code*: 1 B

Example 30:

R' 1.96 min (Method 1); m/z 541 (M+H) + (ES + ); 1 H NMR δ: 1.49-1.61 (2H, overlapping m), 1.78-1.87 (2H, overlapping m), 1.96-2.06 (2H, overlapping m), 2.13 (6H, s), 2.55- 2.64 (2H, overlapping m), 3.75 (3H, s), 3.82 (1 H, m), 5.61 (1 H, s), 6.87

(2H, m), 7.36 (1 H, s), 7.50-7.56

(Z)-Methyl 5-methyl-3-(((4-(((1 -methylpiperidin-4- (4H, overlapping m), 7.58-7.69 (3H, yl)oxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2

overlapping m), 10.89 (1 H, s), oxoindoline-6-carboxylate

1 1.36 (1 H, s), 12.22 (1 H, s).

Route code*: 1 B

Example 31 :

R' 1.99 min (Method 1); m/z 555 (M+H) + (ES + ); 1 H NMR δ: 1.50-1.62 (2H, overlapping m), 1.77-1.89 (2H, overlapping m), 1.97-2.07 (2H, overlapping m), 2.10-2.18 (9H, overlapping m), 2.53-2.63 (2H, overlapping m), 3.75 (3H, s), 3.82

(1 H, m), 5.58 (1 H, s), 6.58 (1 H,

(Z)-Methyl 5-methyl-3-(((3-methyl-4-(((1 - dd), 6.79 (1 H, d), 7.08 (1 H, d), 7.36 methylpiperidin-4- (1 H, s), 7.52 (2H, m), 7.57-7.71 yl)oxy)carbamoyl)phenyl)amino)(phenyl)methylene)-2

(3H, overlapping m), 10.87 (1 H, s), oxoindoline-6-carboxylate

1 1.13 (1 H, s), 12.19 (1 H, s).

Route code*: 1 B

Example 32: R' 1.86 min (Method 1); m/z 588

(M+H) + (ES + ); 1 H NMR δ: 2.10-2.15 (6H, overlapping m), 2.19-2.46 (8H, overlapping m), 2.52-2.55 (2H, overlapping m), 3.76 (3H, s), 3.92 (2H, t), 5.63 (1 H, s), 6.59 (1 H, dd), 6.77 (1 H, dd), 7.36 (1 H, s), 7.40

(1 H, t), 7.51-7.59 (2H, overlapping

(Z)-Methyl 3-(((3-fluoro-4-((2-(4-methylpiperazin-1- m), 7.61-7.72 (3H, overlapping m), yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene

10.92 (1 H, s), 12.15 (1 H, s).

)-5-methyl-2-oxoindoline-6-carboxylate

Route code*: 1 B

Example 33:

R' 1.73 min (Method 1); m/z 574 (M+H) + (ES + ); 1 H NMR δ: 2.13 (3H, s), 2.16-2.47 (8H, overlapping m), 2.51-2.56 (2H, overlapping m), 3.78 (3H, s), 3.92 (2H, t), 5.89 (1 H, d), 6.57 (1 H, d), 6.76 (1 H, d), 7.22 (1 H, dd), 7.35-7.44 (2H,

overlapping m), 7.51-7.58 (2H,

(Z)-Methyl 3-(((3-fluoro-4-((2-(4-methylpiperazin-1- overlapping m), 7.58-7.70 (3H, yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene

overlapping m), 11.06 (1 H, s),

)-2-oxoindoline-6-carboxylate

12.21 (1 H, s).

Route code*: 1 B

Example 34:

R* 1.69 min (Method 1); m/z 569

(M+H) + (ES + ); 1 H NMR δ: 1.57-1.69 (2H, overlapping m), 1.74-1.89 (4H, overlapping m), 2.06 (3H, s), 2.10- 2.18 (6H, overlapping m), 2.73- 2.84 (2H, overlapping m), 3.35- 3.45 (3H, br s), 3.75 (3H, s), 5.64 (1 H, s), 6.63 (1 H, d), 6.83 (1 H, s), 7.01 (1 H, d), 7.36 (1 H, s), 7.49 (2H, m), 7.54-7.67 (3H, overlapping m),

(Z)-Methyl 3-(((4-(methoxy(1 -methylpiperidin-4- 10.85 (1 H, s), 12.13 (1 H, s).

yl)carbamoyl)-3- (Missing 1 H-presumed obscured methylphenyl)amino)(phenyl)methylene)-5-methyl-2

by solvent).

oxoindoline-6-carboxylate

Route code*: 1 B

Example 35:

R* 1.67 min (Method 1); m/z 555 (M+H) + (ES + ); 1 H NMR δ: 1.61-1.69 (2H, overlapping m), 1.74-1.89 (4H, overlapping m), 2.13 (3H, s), 2.14 (3H, s), 2.74-2.83 (2H, overlapping m), 3.45 (3H, s), 3.75 (3H, s), 3.85 (1 H, m), 5.64 (1 H, s), 6.89 (2H, d), 7.31-7.39 (3H, overlapping m), 7.51 (2H, m), 7.56-7.68 (3H, overlapping m), 10.87 (1 H, s),

(Z)-Methyl 3-(((4-(methoxy(1 -methylpiperidin-4- 12.20 (1 H, s).

yl)carbamoyl)phenyl)amino)(phenyl)methylene)-5- methyl-2-oxoindoline-6-carboxylate

Route code*: 1 B

Example 36: R' 1.54 min (Method 1); m/z 584

(M+H) + (ES + ); 1 H NMR δ: 0.97 (3H, m), 2.13 (3H, s), 2.22-2.48 (10H, overlapping m), 2.54 (2H, t), 3.75 (3H, s), 3.92 (2H, t), 5.61 (1 H, s), 6.87 (2H, m), 7.36 (1 H, s), 7.49- 7.56 (4H, overlapping m), 7.58-

7.69 (3H, overlapping m), 10.89

(Z)-Methyl 3-(((4-((2-(4-ethylpiperazin-1- (1 H, s), 1 1.57 (1 H, br s), 12.22 (1 H, yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene

s).

)-5-methyl-2-oxoindoline-6-carboxylate

Route code*: 1 B

Example 37:

R< 2.35 min (Method 1); m/z 456 (M+H) + (ES + ); 1 H NMR δ: 3.77 (3H, s), 4.05-4.12 (2H, overlapping m), 4.40-4.47 (2H, overlapping m), 5.88 (1 H, d), 6.85 (2H, m), 7.21 (1 H, dd), 7.39-7.54 (5H, overlapping m), 7.55-7.66 (3H,

overlapping m), 11.02 (1 H, s),

(Z)-Methyl 3-(((4-(1 ,2-oxazetidine-2- 12.26 (1 H, s).

carbonyl)phenyl)amino)(phenyl)methyl

oxoindoline-6-carboxylate

Route code*: 1 C

Example 38:

R' 1.30 min (Method 1); m/z 570 (M+H) + (ES + ); 1 H NMR δ: 1.60 (1 H, m), 1.83 (1 H, m), 2.13 (6H, s), 2.36 (1 H, m), 2.57-2.79 (6H, overlapping m), 3.77 (3H, s), 3.91 (2H, t), 5.87 (1 H, d), 6.86 (2H, m), 7.21 (1 H, dd), 7.42 (1 H, d), 7.50-7.56 (4H,

overlapping m), 7.57-7.68 (3H,

(S,Z)-Methyl 3-(((4-((2-(3-(dimethylamino)pyrrolidin-1- overlapping m), 8.20 (1 H, s), 1 1.04 yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene

(1 H, s), 12.28 (1 H, s).

)-2-oxoindoline-6-carboxylate, formate Route code * : 1C

Example 39:

R l 1.41 min (Method 1); m/z 586 (M+H) + (ES + ); 1 H NMR δ: 2.30-2.47 (8H, overlapping m), 2.53 (2H, m), 3.47 (2H, t), 3.77 (3H, s), 3.92 (2H, t), 5.87 (1 H, d), 6.86 (2H, m), 7.21 (1 H, dd), 7.42 (1 H, d), 7.50-7.56 (4H, overlapping m), 7.57-7.70 (3H,

overlapping m), 8.23 (1 H, s), 1 1.04

(Z)-Methyl 3-(((4-((2-(4-(2-hydroxyethyl)piperazin-1 - (1 H, s), 12.28 (1 H, s). (Missing 2H- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene

presumed obscured by solvent). )-2-oxoindoline-6-carboxylate

Route code*: 1 C

Example 40:

R l 1.59 min (Method 1); m/z 557 (M+H) + (ES + ); 1 H NMR δ: 1.27-1.44 (2H, overlapping m), 1.60-1.76 (2H, overlapping m), 2.00-2.19 (2H, overlapping m), 2.54-2.58 (2H, overlapping m), 2.71-2.81 (2H, overlapping m), 3.43 (1 H, m), 3.61

(1 H, m), 3.77 (3H, s), 3.92 (2H, t),

(Z)-Methyl 3-(((4-((2-(4-hydroxypiperidin-1- 5.86 (1 H, d), 6.86 (2H, m), 7.20 yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene

(1 H, dd), 7.42 (1 H, d), 7.48-7.56

)-2-oxoindoline-6-carboxylate

(4H, overlapping m), 7.56-7.68 (3H, overlapping m), 8.20 (1 H, s), 1 1.03 (1 H, s), 12.28 (1 H, s).

Route code * : 1 C

Example 41 : R' 1.75 min (Method 1); m/z 591

(M+H) + (ES + ); 1 H NMR δ: 2.82-2.89 (2H, m), 3.05-3.17 (8H, overlapping m), 3.77 (3H, s), 3.97 (2H, t), 5.87 (1H, d), 6.87 (2H, m), 7.21 (1H, dd), 7.42 (1H, d), 7.50-7.56 (4H, overlapping m), 7.57-7.68 (3H,

overlapping m), 11.03 (1H, s),

(Z)-Methyl 3-(((4-((2-(1,1- 11.63 (1H, s), 12.28 (1H, s).

dioxidothiomorpholino)ethoxy)carbamoyl)phenyl)amin

o)(phenyl)methylene)-2-oxoindoline-6-carboxylate

Route code*: 1C

Example 42:

R' 1.47 min (Method 1); m/z 556 (M+H) + (ES + ); 1 H NMR δ: 2.60-2.67 (4H, overlapping m), 3.02 (2H, s), 3.09-3.17 (2H, overlapping m), 3.77 (3H, s), 3.95 (2H, t), 5.87 (1H, d), 6.86 (2H, m), 7.21 (1H, dd), 7.42 (1H, d), 7.49-7.56 (4H,

overlapping m), 7.57-7.68 (3H,

(Z)-Methyl 2-oxo-3-(((4-((2-(3-oxopiperazin-1- overlapping m), 7.69-7.75 (1H, m), yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene

11.03 (1H, s), 11.58 (1H, s), 12.28

)indoline-6-carboxylate

(1H, s).

Route code*: 1C

Example 43:

R' 1.55 min (Method 1); m/z 566 (M+H) + (ES + ); 1 H NMR δ: 1.59-2.00 (4H, overlapping d), 2.52-2.96 (6H, overlapping m), 3.77 (3H, s), 3.98 (2H, m), 5.87 (1H, d), 6.87 (2H, m), 7.21 (1H, dd), 7.42 (1H, d), 7.49- 7.57 (4H, overlapping m), 7.57-

7.70 (3H, overlapping m), 8.13 (0.5H, s), 11.02 (1H, d), 12.28 (1H, (Z)-Methyl 3-(((4-((2-(4-cyanopiperidin-1- s). (Missing 1 H-presumed yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene obscured by solvent).

)-2-oxoindoline-6-carboxylate, hemiformate

Route code*: 1 C

Example 44:

R' 1.59 min (Method 1); m/z 569 (M+H) + (ES + ); 1 H NMR δ (mixture of rotamers): 0.80 and 0.87 (3H, 2 x d), 0.92-1.67 (9H, overlapping m), 1.74-1.98 (2H, overlapping m), 3.77 (3H, s), 5.85 and 5.89 (1 H, 2 x d), 6.83 (2H, d), 6.92-7.19 (1 H, m),

7.06 (1 H, m), 7.43 (2H, m), 7.52

(Z)-Methyl 3-(((4-((2-((2S,6 )-2,6-dimethylpiperidin-1- (2H, m), 7.54-7.68 (3H, overlapping yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene

m), 8.45 (1 H, s), 1 1.03 (1 H, m), )-2-oxoindoline-6-carboxylate

12.28 (1 H, m), (Missing 4H- presumed obscured by solvent).

Route code*: 1 C

Example 45:

R' 1.40 min (Method 1); m/z 531 (M+H) + (ES + ); 1 H NMR δ: 2.22 (3H, s), 2.45 (2H, t), 2.62 (2H, t), 3.45 (2H, t), 3.77 (3H, s), 3.91 (2H, t), 5.87 (1 H, d), 6.86 (2H, m), 7.20 (1 H, m), 7.42 (1 H, d), 7.47-7.57 (4H, overlapping m), 7.57-7.70 (3H,

overlapping m), 8.32 (1 H, s), 1 1.03

(Z)-Methyl-3-(((4-((2-((2- (1 H, s), 12.28 (1 H, s).

hydroxyethyl)(methyl)amino)ethoxy)carbamoyl)ph

amino)(phenyl)methylene)-2-oxoindoline-6- carboxylate

Route code*: 1 C

Example 46: R' 1.44 min (Method 1); m/z 600

(M+H) + (ES + ); 1 H NMR δ: 2.31-2.47 (8H, overlapping m), 2.51-2.56 (2H, overlapping m), 3.22 (3H, s), 3.40 (2H, t), 3.77 (3H, s), 3.92 (2H, t), 5.87 (1 H, d), 6.86 (2H, m), 7.21 (1 H, dd), 7.42 (1 H, d), 7.48-7.56

(4H, overlapping m), 7.56-7.69 (3H,

(Z)-Methyl 3-(((4-((2-(4-(2-methoxyethyl)piperazin-1- overlapping m), 8.19 (1 H, s), 1 1.03 yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene

(1 H, s), 12.28 (1 H, s). (Missing 2H- )-2-oxoindoline-6-carboxylate, formate

presumed obscured by solvent).

Route code*: 1 C

Example 47:

R' 1.20 min (Method 1); m/z 584 (M+H) + (ES + ); 1 H NMR δ: 1.25-1.45 (2H, overlapping m), 1.68-1.78 (2H, overlapping m), 1.88-2.03 (2H, overlapping m), 2.27 (6H, s), 2.51- 2.59 (3H, overlapping m), 2.92- 2.99 (2H, overlapping m), 3.77 (3H,

s), 3.92 (2H, t), 5.86 (1 H, d), 6.85

(Z)- Methyl 3-(((4-((2-(4-(dimethylamino)piperidin-1- (2H, m), 7.20 (1 H, dd), 7.42 (1 H, yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene

m), 7.49-7.68 (7H, overlapping m), )-2-oxoindoline-6-carboxylate, formate

8.22 (1 H, s), 11.04 (1 H, s), 12.28 (1 H, s).

Route code*: 1 C Example 48:

R' 1.65 min (Method 1); m/z 551 (M+H) + (ES + ); 1 H NMR δ: 2.09 (6H, s), 2.14 (3H, s), 2.39 (2H, t), 3.76 (3H, s), 3.89 (2H, t), 5.64 (1 H, s), 6.96 (2H, m), 7.36 (1 H, s), 7.52- 7.73 (7H, overlapping m), 8.20 (1 H, s), 10.94 (1 H, s), 12.25 (1 H, s). (Z)-Methyl 3-(((4-(Λ/-(2-

(dimethylamino)ethoxy)sulfamoyl)phenyl)amino)(phen

yl)methylene)-5-methyl-2-oxoindoline-6-carboxylate

Route code*: 1A

Example 49:

R' 1.73 min (Method 1); m/z 555 (M+H) + (ES + ); 1 H NMR δ: 1.33-1.39 (2H, overlapping m), 1.42-1.52 (4H, overlapping m), 2.13 (3H, s), 2.34- 2.44 (4H, overlapping m), 2.53 (2H, m), 3.75 (3H, s), 3.92 (2H, t), 5.61 (1 H, s), 6.87 (2H, m), 7.36 (1 H, s),

7.49-7.56 (4H, overlapping m),

(Z)-Methyl 5-methyl-2-oxo-3-(phenyl((4-((2-(piperidin- 7.57-7.69 (3H, overlapping m), 1- 8.20 (1 H, s), 10.89 (1 H, s), 12.21 yl)ethoxy)carbamoyl)phenyl)amino)methylene)indoline

(1 H, s).

-6-carboxylate

Route code*: 1 C

Example 50:

R' 1.33 min (Method 1); m/z 598 (M+H) + (ES + ); 1 H NMR δ: 1.30-1.43 (2H, overlapping m), 1.68-1.76 (2H, overlapping m), 1.89-2.01 (2H, overlapping m), 2.10-2.19 (5H, overlapping m), 2.22 (6H, s), 2.90- 2.98 (2H, overlapping m), 3.75 (3H,

s), 3.92 (2H, t), 5.61 (1 H, s), 6.87

(Z)-Methyl 3-(((4-((2-(4-(dimethylamino)piperidin-1- (2H, m), 7.36 (1 H, s), 7.49-7.57 yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene

(4H, overlapping m), 7.57-7.68 (3H, )-5-methyl-2-oxoindoline-6-carboxylate, formate

overlapping m), 8.22 (1 H, s), 10.89 (1 H, s), 12.22 (1 H, s). (Missing 1 H- presumed obscured by solvent).

Route code*: 1 C Example 51 :

R* 1.32 min (Method 1); m/z 584 (M+H) + (ES + ); 1 H NMR δ: 1.59 (1 H, m), 1.82 (1 H, m), 2.08-2.17 (9H, overlapping m), 2.35 (1 H, m), 2.55- 2.79 (5H, overlapping m), 3.75 (3H, s), 3.91 (2H, t), 5.61 (1 H, s), 6.87 (2H, m), 7.36 (1 H, s), 7.49-7.56

(4H, overlapping m), 7.57-7.70 (3H,

(S,Z)-Methyl 3-(((4-((2-(3-(dimethylamino)pyrrolidin-1- overlapping m), 8.20 (1 H, s), 10.89 yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene

(1 H, s), 12.22 (1 H, s). (Missing 1 H- )-5-methyl-2-oxoindoline-6-carboxylate, formate

presumed obscured by solvent).

Route code*: 1 C

Example 52:

R< 2.05 min (Method 2); m/z 585 (M+H) + (ES + ); 1 H NMR δ: 1.30-1.43 (2H, overlapping m), 1.75-1.83 (2H, overlapping m), 2.06-2.16 (5H, overlapping m), 2.53 (1 H, m), 2.68- 2.76 (2H, overlapping m), 3.07- 3.17 (2H, overlapping m), 3.21 (3H,

s), 3.75 (3H, s), 3.91 (2H, t), 5.61

(Z)- Methyl 3-(((4-((2-(4-methoxypiperidin-1- (1 H, s), 6.87 (2H, m), 7.36 (1 H, s), yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene

7.49-7.57 (4H, overlapping m),

)-5-methyl-2-oxoindoline-6-carboxylate

7.57-7.68 (3H, overlapping m), 8.34 (1 H, s), 10.89 (1 H, s), 12.21 (1 H, s).

Route code*: 1 C Example 53:

R< 1.77 min (Method 1); m/z 569 (M+H) + (ES + ); 1 H NMR δ 1.32-1.41 (2H, overlapping m), 1.43-1.53 (4H, overlapping m), 1.72 (2H, m), 2.14 (3H, s), 2.26-2.40 (6H, overlapping m), 3.76 (3H, s), 3.85 (2H, t), 5.62

(1 H, s), 6.88 (2H, m), 7.37 (1 H, s),

(Z)- Methyl 5-methyl-2-oxo-3-(phenyl((4-((3-(piperidin- 7.48-7.57 (4H, overlapping m), 1- 7.59-7.69 (3H, overlapping m), yl)propoxy)carbamoyl)phenyl)amino)methylene)indolin

10.89 (1 H, s), 12.22 (1 H, s).

e-6-carboxylate

Route code*: 1 B

Example 54:

R< 1.60 min (Method 1); m/z 571 (M+H) + (ES + ); 1 H NMR δ: 1.27-1.40 (2H, overlapping m), 1.48 (1 H, m), 1.60-1.70 (2H, overlapping m), 1.81 (1 H, m), 2.07 (1 H, overlapping m), 2.13 (3H, s), 2.70-2.84 (2H,

overlapping m), 3.10 (1 H, m), 3.41 (1 H, m), 3.67 (1 H, m), 3.76 (3H, s),

(Z)- Methyl 3-(((4-((2-(4-hydroxypiperidin-1- 3.91 (2H, t), 5.61 (1 H, s), 6.85 (2H, yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene

m), 7.36 (1 H, s), 7.47-7.56 (4H, )-5-methyl-2-oxoindoline-6-carboxylate, formate

overlapping m), 7.58-7.67 (3H, overlapping m), 8.31 (2H, s), 10.89 (1 H, s), 12.22 (1 H, s).

Route code*: 1 C Example 55:

R' 1.74 min (Method 1); m/z 599 (M+H) + (ES + ); 1 H NMR δ: 1.31-1.43 (2H, overlapping m), 1.70 (2H, m), 1.76-1.83 (2H, overlapping m), 1.96-2.09 (2H, overlapping m), 2.14 (3H, s), 2.36 (2H, t), 2.60-2.70 (2H, overlapping m), 3.14 (1 H, m),

3.22 (3H, s), 3.76 (3H, s), 3.84 (2H,

(Z)-Methyl 3-(((4-((3-(4-methoxypiperidin-1- t), 5.62 (1 H, s), 6.88 (2H, m), 7.37 yl)propoxy)carbamoyl)phenyl)amino)(phenyl)methylen

(1 H, s), 7.48-7.57 (4H, overlapping e)-5-methyl-2-oxoindoline-6-carboxylate

m), 7.59-7.69 (3H, overlapping m), 10.89 (1 H, s), 12.22 (1 H, s).

Route code*: 1 B

Example 56:

R* 1.61 min (Method 1); m/z 559 (M+H) + (ES + ); 1 H NMR δ: 2.06 (3H, s), 2.14 (3H, s), 2.32-2.38 (4H, overlapping m), 3.22 (3H, s), 3.39 (2H, m), 3.76 (2H, m), 3.76 (3H, s), 4.26 (1 H, m), 5.62 (1 H, s), 6.86 (2H, m), 7.37 (1 H, s), 7.44 (2H, m),

7.52 (2H, m), 7.57-7.69 (3H,

(Z)-Methyl 3-(((4-((2-((2- overlapping m), 10.84 (1 H, s), hydroxyethyl)(methyl)amino)ethoxy)(methyl)carbamoyl

12.22 (1 H, s).

)phenyl)amino)(phenyl)methylene)-5-methyl-2- oxoindoline-6-carboxylate

Route code*: 1 D

Example 57: R 1 1.51 min (Method 1); m/z 570

(M+H) + (ES + ); 1 H NMR δ: 2.14 (3H, s), 2.26-2.34 (6H, overlapping m), 2.77-2.79 (4H, overlapping m), 3.21 (3H, s), 3.75 (3H, s), 3.79 (2H, t), 5.61 (1 H, s), 6.86 (2H, m), 7.37 (1 H, s), 7.43 (2H, m), 7.52 (2H, m),

7.57-7.69 (3H, overlapping m),

(Z)-Methyl 5-methyl-3-(((4-(methyl(2-(piperazin-1- 10.87 (1 H, s), 12.22 (1 H, s).

yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene

)-2-oxoindoline-6-carboxylate

Route code*: 1 D

Example 58:

R' 1.56 min (Method 1); m/z 614 (M+H) + (ES + ); 1 H NMR δ: 2.14 (s, 3H), 2.17-2.44 (12H, overlapping m), 3.21 (3H, s), 3.48 (2H, m), 3.70-3.83 (5H, overlapping m), 4.36 (1 H, m), 5.62 (1 H, s), 6.86 (2H, m), 7.36 (1 H, s), 7.42 (2H, m),

7.52 (2H, m), 7.56-7.69 (3H,

(Z)-Methyl 3-(((4-((2-(4-(2-hydroxyethyl)piperazin-1 - overlapping m), 10.88 (1 H, s), yl)ethoxy)(methyl)carbamoyl)phenyl)amino)(phenyl)m

12.22 (1 H, s).

ethylene)-5-methyl-2-oxoindoline-6-carboxylate

Route code*: 1 D

Example 59:

R< 1.42 min (Method 1); m/z 582 (M+H) + (ES + ); 1 H NMR δ: 1.38 (1 H, m), 1.53 (1 H, m), 2.14 (3H, s), 2.21 (1 H, m), 2.42-2.54 (2H, overlapping m), 2.58-2.70 (2H, overlapping m), 2.85 (1 H, m), 3.18 (1 H, m), 3.21 (3H, s), 3.44 (1 H, m), 3.70 (2H, t),

3.75 (3H, s), 5.61 (1 H, s), 6.86 (2H,

(Z)-Methyl 3-(((4-((2-((1 S,4S)-2,5- m), 7.36 (1 H, s), 7.45 (2H, m), 7.53 diazabicyclo[2.2.1 ]heptan-2- yl)ethoxy)(methyl)carbamoyl)phenyl)amino)(phenyl)m (2H, m), 7.57-7.68 (3H, overlapping ethylene)-5-methyl-2-oxoindoline-6-carboxylate m), 10.87 (1 H, s), 12.22 (1 H, br. s). Route code*: 1 D

Example 60:

R' 1.47 min (Method 1); m/z 596 (M+H) + (ES + ); 1 H NMR δ: 1.56-1.65 (2H, overlapping m), 1.66-1.75 (2H, overlapping m), 2.13 (3H, s), 2.22 (2H, t), 2.64-2.70 (2H, overlapping m), 2.89-2.94 (2H, overlapping m), 3.22 (3H, s), 3.72-3.78 (5H,

overlapping m), 5.60 (1 H, s), 6.86

(Z)-Methyl 3-(((4-((2-(3,8-diazabicyclo[3.2.1]octan-8- (2H, m), 7.36 (1 H, s), 7.45 (2H, m), yl)ethoxy)(methyl)carbamoyl)phenyl)amino)(phenyl)m

7.53 (2H, m), 7.57-7.70 (3H, ethylene)-5-methyl-2-oxoindoline-6-carboxylate

overlapping m), 10.88 (1 H, s), 12.23 (1 H, s), (Missing 2H- presumed overlap with solvent).

Route code*: 1 D Example 61 :

R' 1.65 min (Method 1); m/z 596 (M+H) + (ES + ); 1 H NMR δ: 1.57-1.67 (4H, overlapping m), 2.04-2.10 (2H, overlapping m), 2.14 (3H, s), 2.27 (2H, t), 2.45-2.48 (2H, overlapping m), 3.21 (3H, s), 3.39-3.45 (2H, overlapping m), 3.74 (2H, t), 3.75

(3H, s), 5.62 (1 H, s), 6.86 (2H, m),

(Z)-Methyl 3-(((4-((2-(3,8-diazabicyclo[3.2.1]octan-3- 7.32-7.42 (3H, overlapping m), yl)ethoxy)(methyl)carbamoyl)phenyl)amino)(phenyl)m

7.52 (2H, m), 7.56-7.70 (3H, ethylene)-5-methyl-2-oxoindoline-6-carboxylate

overlapping m), 10.87 (1 H, s), 12.21 (1 H, s).

Route code*: 1 D

Example 62: R* 1.56 min (Method 1); m/z 584

(M+H) + (ES + ); 1 H NMR δ: 0.88 (3H, d), 1.43 (1 H, m), 1.75 (1 H, m), 2.13 (3H, s), 2.25 (2H, t), 2.73 (1 H, m), 3.21 (3H, s), 3.71-3.83 (5H, overlapping m), 5.61 (1 H, s), 6.85 (2H, m), 7.36 (1 H, s), 7.44 (2H, m),

7.53 (2H, m), 7.56-7.69 (3H,

(S,Z)-Methyl 5-methyl-3-(((4-(methyl(2-(3- overlapping m), 10.87 (1 H, s), methylpiperazin-1- 12.24 (1 H, s), (Missing 4H- yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene

presumed overlap with solvent).

)-2-oxoindoline-6-carboxylate

Route code*: 1 D

Example 63:

R< 1.70 min (Method 1); m/z 580 (M+H) + (ES + ); 1 H NMR δ: 1.60-1.72 (2H, overlapping m), 1.77-1.87 (2H, overlapping m), 2.13 (3H, s), 2.32 (2H, m), 2.52-2.63 (4H, overlapping m), 2.83 (1 H, m), 3.75 (3H, s), 3.92 (2H, t), 5.62 (1 H, s), 6.87 (2H, m),

7.36 (1 H, s), 7.49-7.56 (4H,

(Z)- Methyl 3-(((4-((2-(4-cyanopiperidin-1- overlapping m), 7.58-7.68 (3H, yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene

overlapping m), 8.15 (0.4H, s), )-5-methyl-2-oxoindoline-6-carboxylate, 0.4 formate

10.88 (1 H, s), 12.21 (1 H, s).

R* 1.42 min (Method 1); m/z 584 (M+H) + (ES + ); 1 H NMR δ: 1.70 (2H, m), 2.13 (6H, s), 2.21-2.42 (10H, overlapping m), 3.75 (3H, s), 3.84 (2H, t), 5.61 (1 H, s), 6.86 (2H, m), 7.36 (1 H, s), 7.48-7.56 (4H, overlapping m), 7.57-7.68 (3H, overlapping m), 10.88 (1 H, s),

(Z)-Methyl 5-methyl-3-(((4-((3-(4-methylpiperazin-1 - 12.22 (1 H, s).

yl)propoxy)carbamoyl)phenyl)amino)(phenyl)methylen

e)-2-oxoindoline-6-carboxylate

Route code*: 1 B

Example 65:

R' 1.54 min (Method 1); m/z 584 (M+H) + (ES + ); 1 H NMR δ: 0.88 (3H, d), 1.43 (1 H, m), 1.75 (1 H, m), 2.13 (3H, s), 2.24 (2H, t), 2.51-2.60 (2H, overlapping m), 2.72 (1 H, m), 3.21 (3H, s), 3.72-3.83 (5H, overlapping m), 5.61 (1 H, s), 6.84 (2H, m), 7.36

( ,Z)-Methyl 5-methyl-3-(((4-(methyl(2-(3- (1 H, s), 7.44 (2H, m), 7.53 (2H, m), methylpiperazin-1- 7.57-7.69 (3H, overlapping m), yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene 10.87 (1 H, s), 12.24 (1 H, s), )-2-oxoindoline-6-carboxylate (Missing 2H-presumed overlap with solvent).

Route code*: 1 D

Example 66:

R' 1.57 min (Method 1); m/z 582 (M+H) + (ES + ); 1 H NMR δ: 1.70 (1 H, m), 2.14 (3H, s), 2.25 (1 H, m), 2.51-2.57 (3H, overlapping m), 2.79-2.87 (2H, overlapping m), 3.23 (3H, s), 3.42-3.48 (2H, overlapping m), 3.75 (3H, s), 3.81

(Z)-Methyl 3-(((4-((2-(3,6-diazabicyclo[3.1.1 ]heptan-3- (2H, t), 5.62 (1 H, s), 6.83 (2H, m), yl)ethoxy)(methyl)carbamoyl)phenyl)amino)(phenyl)m 7.37 (1 H, s), 7.42 (2H, m), 7.53 ethylene)-5-methyl-2-oxoindoline-6-carboxylate (2H, m), 7.57-7.69 (3H, overlapping m), 10.87 (1 H, s), 12.17 (1 H, br s). (Missing 1 H-presumed overlap with solvent).

Route code*: 1 D Example 67:

R 1 2.05 min (Method 1); m/z 598 (M+H) + (ES + ); 1 H NMR δ: 0.95 (6H, d), 1.77 (2H, t), 2.04-2.17 (8H, overlapping m), 2.76 (2H, m), 3.76 (3H, s), 3.92 (2H, t), 5.62 (1 H, s), 6.88 (2H, m), 7.37 (1 H, s), 7.49- 7.58 (4H, overlapping m), 7.58- 7.70 (3H, overlapping m), 8.25 (1 H,

(Z)-Methyl 5-methyl-2-oxo-3-(phenyl((4-((2-((3ft,5S)- s), 10.89 (1 H, s), 12.22 (1 H, s), 3,4,5-trimethylpiperazin-1- (Missing 2H-presumed overlap with yl)ethoxy)carbamoyl)phenyl)amino)methylene)indoline

-6-carboxylate

Route code*: 1 C

Example 68:

R 1 1.57 min (Method 1); m/z 584 (M+H) + (ES + ); 1 H NMR δ: 1.13 (3H, t), 2.13 (3H, s), 2.16-2.24 (4H, overlapping m), 2.27 (2H, t), 2.60- 2.68 (4H, overlapping m), 3.64 (2H, m), 3.73-3.78 (5H, overlapping m), 5.61 (1 H, s), 6.86 (2H, m), 7.37 (1 H, s), 7.41 (2H, m), 7.52 (2H, m),

(Z)-Methyl 3-(((4-(ethyl(2-(piperazin-1- 7.57-7.69 (3H, overlapping m), yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene 10.87 (1 H, s), 12.24 (1 H, s).

)-5-methyl-2-oxoindoline-6-carboxylate

Route code*: 1 B

Example 69: R 1 1.59 min (Method 1); m/z 598

(M+H) + (ES + ); 1 H NMR δ: 1.18 (6H, d), 2.14 (3H, s), 2.16-2.25 (4H, overlapping m), 2.26 (2H, t), 2.60- 2.69 (4H, overlapping m), 3.71 (2H, t), 3.75 (3H, s), 4.30 (1 H, m), 5.61 (1 H, s), 6.87 (2H, m), 7.29-7.39 (3H, overlapping m), 7.53 (2H, m),

(Z)-Methyl 3-(((4-(isopropyl(2-(piperazin-1 - 7.56-7.69 (3H, overlapping m), yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene 10.87 (1 H, s), 12.23 (1 H, s).

)-5-methyl-2-oxoindoline-6-carboxylate

Route* : 1 B

Example 70

R* 1.61 min (Method 1); m/z 570 (M+H) + (ES + ); 1 H NMR δ: 2.13 (3H, s), 2.59-2.67 (4H, overlapping m), 3.02 (2H, br s), 3.13 (2H, m), 3.75 (3H, s), 3.95 (2H, m), 5.62 (1 H, s), 6.88 (2H, m), 7.36 (1 H, s), 7.51- 7.56 (4H, overlapping m), 7.59- 7.69 (3H, overlapping m), 7.72 (1 H,

(Z)-Methyl 5-methyl-2-oxo-3-(((4-((2-(3-oxopiperazin- br s), 10.89 (1 H, s), 11.58 (1 H, br 1- s), 12.22 (1 H, s).

yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene

)indoline-6-carboxylate

Route* : 1 C

Example 71 :

R< 1.56 min (Method 1); m/z 627 (M+H) + (ES + ); 1 H NMR δ: 2.24-2.36 (10H, overlapping m), 2.59 (3H, d), 2.87 (2H, s), 3.21 (3H, s), 3.75- 3.80 (5H, overlapping m), 5.87 (1 H, d), 6.85 (2H, m), 7.21 (1 H, dd), 7.40-7.44 (3H, overlapping m), 7.51-7.55 (3H, overlapping m),

(Z)-Methyl 3-(((4-(methyl(2-(4-(2-(methyl 7.57-7.67 (3H, overlapping m), oxoethyl)piperazin-1- 1 1.03 (1 H, s), 12.28 (1 H, s). yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene

)-2-oxoindoline-6-carboxylate

Route*: 1 D

Example 72

R' 1.52 min (Method 1); m/z 613 (M+H) + (ES + ); 1 H NMR δ: 2.35-2.58 (10H, overlapping m), 2.60 (3H, d), 2.86 (2H, br s), 3.77 (3H, s), 3.93 (2H, t), 5.87 (1 H, d), 6.87 (2H, m), 7.21 (1 H, dd), 7.42 (1 H, d), 7.51- 7.55 (4H, overlapping m), 7.57-

(Z)-Methyl 3-(((4-((2-(4-(2-(methylamino)-2- 7.68 (4H, overlapping m), 1 1.04 oxoethyl)piperazin-1- (1 H, s), 1 1.56 (1 H, br s), 12.28 (1 H, yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene

s).

)-2-oxoindoline-6-carboxylate

Route*: 1 C

Example 73

Rt 1.57 min (Method 1); m/z 627 (M+H)+ (ES+); 1 H NMR δ: 2.14 (3H, s), 2.34-2.54 (8H, overlapping m), 2.55 (2H, t), 2.60 (3H, s), 2.70 (2H, s), 3.76 (3H, s), 3.94 (2H, t), 5.62 (1 H, s), 6.38 (2H, m), 7.37 (1 H, s), 7.50-7.55 (4H, overlapping

(Z)-Methyl 5-methyl-3-(((4-((2-(4-(2-(methylamino)-2- m), 7.57-7.68 (4H, overlapping m), oxoethyl)piperazin-1- 10.90 (1 H, s), 11.56 (1 H, br s), yl)ethoxy)carbamoyl)phenyl)amino)(phenyl)methylene

12.22 (1 H, s).

)-2-oxoindoline-6-carboxylate

Route*: 1 C Biological Testing: Experimental Methods

Enzyme Inhibition Assays The enzyme inhibitory activities of compounds disclosed herein were determined using the ADP-Glo™ assay (Promega, UK). Assays for FGFR1 , PDGFRa, PDGFRβ and VEGFR2 were performed in buffer containing 40 mM Tris pH 7.5, 20 mM MgC , 0.1 mg/mL BSA and 1 mM DTT; whilst assays for FGFR3 and VEGFR1 were performed in the above buffer supplemented with 2 mM MnCI 2 .

FGFR1 Enzyme Inhibition

The inhibitory activities of compounds of the invention against FGFR1 (FGFR1 Kinase Enzyme System: Promega), were evaluated by mixing the FGFR1 protein (3.12 ng/mL, 2 μΙ_), substrate (Poly (4: 1 Glu 4 , Ty ), 100 ng/mL, 2 μΙ_) with the test compound (2 μΙ_ at either 3 μΜ, 0.67 μΜ, 0.15 μΜ, 0.033 μΜ, 0.0073 μΜ, 0.0016 μΜ, 0.0036 μΜ or 0.00008 μΜ) for 90 min at 25°C. The kinase reaction was then initiated by adding ATP (50 μΜ, 2 μΙ_) and the mixture was incubated for 1 hr at 25°C. ADP-Glo™ reagent was added for 40 min (8 μΙ_) then development reagent (16 μΙ_) was added for 40 min prior to detection in a microplate reader (EnVision® Multilabel Reader, Perkin Elmer).

FGFR3 Enzyme Inhibition

The inhibitory activities of compounds of the invention against FGFR3 (FGFR3 Kinase Enzyme System: Promega), were evaluated by mixing the FGFR3 protein (12.5 ng/mL, 2 μί), substrate (Poly (Ala6, GIU2, Lyss, Ty ), 100 ng/mL, 2 μί) with the test compound (2 μί at either 3 μΜ, 0.67 μΜ, 0.15 μΜ, 0.033 μΜ, 0.0073 μΜ, 0.0016 μΜ, 0.0036 μΜ or 0.00008 μΜ) for 90 min at 25°C. The kinase reaction was initiated by adding ATP (50 μΜ, 2 μί) and the mixture was incubated for 90 min at 25°C. ADP-Glo™ reagent was added for 40 min (8 μΙ_) then development reagent (16 μΙ_) was added for 40 min prior to detection in a microplate reader (EnVision® Multilabel Reader, Perkin Elmer).

PDGFRa Enzyme Inhibition The inhibitory activities of compounds of the invention against PDGFRa (PDGFRa Kinase Enzyme System: Promega), were evaluated by mixing the PDGFRa protein (12.5 ng/mL, 2 μί), substrate (Poly (4: 1 Glu 4 , Ty ), 100 ng/mL, 2 μί) with the test compound (2 μί at either 3 μΜ, 0.67 μΜ, 0.15 μΜ, 0.033 μΜ, 0.0073 μΜ, 0.0016 μΜ, 0.0036 μΜ or 0.00008 μΜ) for 90 min at 25°C. The kinase reaction was initiated by adding ATP (25 μΜ, 2 μΙ_) and the mixture was incubated for 1 hr at 25°C. ADP-Glo™ reagent was added for 40 min (8 μΙ_) then development reagent (16 μΙ_) was added for 40 min prior to detection in a microplate reader (EnVision® Multilabel Reader, Perkin Elmer).

ΡΩΘΕΡΙβ Enzyme Inhibition

The inhibitory activities of compounds of the invention against PDGFF^ (PDGFF^ Kinase Enzyme System: Promega), were evaluated by mixing the PDGFF^ protein (6.25 ng/mL, 2 μΙ_), substrate (Poly (4: 1 Glu 4 , Ty ), 100 ng/mL, 2 μΙ_) with the test compound (2 μΙ_ at either 3 μΜ, 0.67 μΜ, 0.15 μΜ, 0.033 μΜ, 0.0073 μΜ, 0.0016 μΜ, 0.0036 μΜ or 0.00008 μΜ) for 90 min at 25°C. The kinase reaction was initiated by adding ATP (25 μΜ, 2 μΙ_) and the mixture was incubated for 1 hr at 25°C. ADP-Glo™ reagent was added for 40 min (8 μΙ_) then development reagent (16 μΙ_) was added for 40 min prior to detection in a microplate reader (EnVision® Multilabel Reader, Perkin Elmer).

VEGFR1 Enzyme Inhibition The inhibitory activities of compounds of the invention against VEGFR1 (VEGFR1 Kinase Enzyme System: Promega), were evaluated by mixing the VEGFR1 protein (12.5 ng/mL, 2 μΙ_), substrate (IGFRI Rtide, 100 ng/mL, 2 μΙ_) with the test compound (2 μΙ_ at either 3 μΜ, 0.67 μΜ, 0.15 μΜ, 0.033 μΜ, 0.0073 μΜ, 0.0016 μΜ, 0.0036 μΜ or 0.00008 μΜ) for 90 min at 25°C. The kinase reaction was initiated by adding ATP (50 μΜ, 2 μΙ_) and the mixture was incubated for 90 min at 25°C. ADP-Glo™ reagent was added for 40 min (8 μΙ_) then development reagent (16 μΙ_) was added for 40 min prior to detection in a microplate reader (EnVision® Multilabel Reader, Perkin Elmer).

VEGFR2 Enzyme Inhibition

The inhibitory activities of compounds of the invention against VEGFR2 (VEGFR2 Kinase Enzyme System: Promega), were evaluated by mixing the VEGFR2 protein (1.56 ng/mL, 2 μΙ_), substrate (Poly (4: 1 Glu 4 , Ty ), 100 ng/mL, 2 μΙ_) with the test compound (2 μΙ_ at either 3 μΜ, 0.67 μΜ, 0.15 μΜ, 0.033 μΜ, 0.0073 μΜ, 0.0016 μΜ, 0.0036 μΜ or 0.00008 μΜ) for 90 min at 25°C. The kinase reaction was initiated by adding ATP (50 μΜ, 2 μΙ_) and the mixture was incubated for 1 hr at 25°C. ADP-Glo™ reagent was added for 40 min (8 μΙ_) then development reagent (16 μΙ_) was added for 40 min prior to detection in a microplate reader (EnVision® Multilabel Reader, Perkin Elmer).

In all cases, the kinase converts ATP to ADP and the ADP-Glo™ reagent then depletes any remaining ATP. The detection reagent converts the ADP that has been produced back into ATP and generates luciferase which can be detected as luminescence. Therefore the luminescent signal is directly proportional to the amount of ADP produced by the enzyme reaction and a reduction in this signal after compound treatment demonstrates inhibition. The percentage inhibition produced by each concentration of compound was calculated using the equation shown below:

(Mean Min - Mean Inh )

% Inhibition = 1— x 100

(Mean Min - Mean Max)

Percentage inhibition was then plotted against compound concentration, and the relative 50% inhibitory concentration (RIC50) was determined from the resultant concentration-response curve. Once this had been determined Ki was calculated using the equation below:

RIC50

Ki = -

1 + Km)

Cellular and Other In Vitro Assays PDGF-BB induced Normal Human Lung Fibroblast (NHLF) proliferation

NHLF's (Lonza group Ltd) are expanded up to 90% confluence in FGM-2 growth media supplemented with 2% FBS (plus SingleQuot™ growth factors; Lonza). Fibroblasts are harvested (Trypsin/EDTA), suspended at 25x10 3 per ml in growth media and 200μΙ is added per well (5x10 3 cells/well) of a 96 well tissue culture plate. After 24 hr incubation (37 C/ 5%C02/95%C>2), cells are serum starved (24 hr) by reducing the FBS concentration to 0.1 % in the culture media. Cells are pre-incubated with test compound for 1 hr, then stimulated with rhuPDGF-BB (100ng/ml, R&D Systems) for 48 hr. Cell proliferation is assessed by BrdU incorporation (Cell Proliferation colorimetric ELISA, Roche). The percent inhibition of rhuPDGF-BB-induced NHLF proliferation by test compound at each concentration is calculated as a percentage of that achieved by rhuPDGF-BB at each concentration of test compound by comparison against vehicle control (basal proliferation). The relative 50% inhibitory concentration (RIC50) is determined from the resultant concentration-response curve. PDGF-BB / FGF-Basic induced MRC-5 Fetal human lung fibroblast) proliferation

MRC-5 fibroblasts (LGC Standards) are expanded up to 90% confluence in DMEM media supplemented with 10% FBS. Cells are harvested (Trypsin/EDTA), suspended at 25x10 3 per ml in growth media and 200μΙ is added per well (5x10 3 cells/well) of a 96 well tissue culture plate. After 24 hr incubation (37 C/ 5%C02/95%02), cells are serum starved (3 hr) by replacing the growth media with media containing 0.1 % FBS. Cells are then pre-incubated with test compound for 1 hr followed by stimulation with rhuPDGF-BB (100ng/ml, R&D Systems) or rhuFGF-basic (5ng/ml; R&D Systems) for 48 hr. Cell proliferation is assessed by BrdU incorporation (Cell Proliferation colorimetric ELISA, Roche). The percent inhibition of rhuPDGF-BB/ rhuFGF -induced MRC-5 proliferation by test compound at each concentration is calculated as a percentage of that achieved by rhuPDGF-BB / FGF-basic at each concentration of test compound by comparison against vehicle control (basal proliferation). The relative 50% inhibitory concentration (RIC50) is determined from the resultant concentration-response curve.

VEGF165 /FGF-Basic induced endothelial cell proliferation

TeloHAEC (telomerase immortalized Human Aortic Endothelial Cells; ATCC) are seeded in to 96 well tissue culture plates at a cell density of 4000 cells per well (100μΙ) in endothelial cell starvation medium (0.5% FBS, without FGF and VEGF growth factors) and cultured for 3 hr (37 C/ 5%C02/95%02). Cells are pre-incubated with test compound for 1 hr followed by stimulation with rhuVEGFi65 (10 ng/ml, R&D Systems) or rhuFGF-basic (5ng/ml, R&D Systems) for 48 hr. Cell proliferation is assessed by BrdU incorporation (Cell Proliferation colorimetric ELISA, Roche). The percent inhibition of rhuVEGFi6s/ rhuFGF-basic -induced TeloHAEC proliferation by test compound at each concentration is calculated as a percentage of that achieved by rhuVEGFi6s/ FGF-basic at each concentration of test compound by comparison against vehicle control (basal proliferation). The relative 50% inhibitory concentration (RIC50) is determined from the resultant concentration-response curve.

PDGF-BB induced phosphorylation οίΡΩΰΡΡβ in fibroblasts

MRC-5 / NHLF / NIH-3T3s (mouse embryonic fibroblasts, LGC Standards) were used to evaluate the inhibitory effect of test compound on PDGFRβ phosphorylation using the HTRF (Homogeneous Time Resolved Fluorescence) phospho-PDGFRβ (Tyr751) cellular assay kit (Cisbio). MRC-5 / NHLFs were seeded in to 96 well tissue culture plates at a cell density of 10000 cells per well in DMEM growth media (10% FBS) or FGM-2 growth media (2% FBS) respectively and cultured for 48 hr (37 C/ 5%C0 2 /95%0 2 ). NIH-3T3s were seeded in to 96 well tissue culture plates at a cell density of 7000 cells per well in DM EM growth media (10% FBS) and cultured for 48 hrs (37 C/ 5%C02/95%02). Cell media was replaced with the respective starvation medium containing 0.1 % FBS and plates further incubated for 24 hr (37 C/ 5%C0 2 /95%0 2 ) for MRC-5/ NHLFs and for 3 hr for NIH-3T3s. Cells were pre-incubated with test compound for 1 hr and then stimulated with rhuPDGF-BB (25-50 ng/ml, R&D Systems) for 5 min and rmPDGF-BB (25 ng/ml, Life Technologies) for NIH-3T3s. Media was aspirated off and cells immediately lysed by addition of 50μΙ lysis buffer provided in the HTRF assay kit. 16μΙ of cell lysate from each well was transferred to a white low volume 384 well plate to which propriety kit reagents were added as per kit instructions. Phosphorylation of the PDGFRβ was quantitated by calculating the ratio of fluorescence read at 665nm and 620nm. The percent inhibition of rPDGF-BB induced PDGFRβ phosphorylation by test compound was calculated as a percentage of that achieved by rPDGF-BB at each concentration of test compound by comparison against vehicle control. The relative 50% inhibitory concentration (RIC50) was determined from the resultant concentration-response curve.

VEGF165 induced phosphorylation of VEGFR2 in endothelial cells

TeloHAECs (telomerase immortalized Human Aortic Endothelial Cells; ATCC) were used to evaluate the inhibitory effect of test compound on VEGFR2 phosphorylation using the HTRF (Homogeneous Time Resolved Fluorescence) phospho-VEGFR2 (Tyr1175) cellular assay kit (Cisbio). TeloHAECs were seeded in to 96 well tissue culture plates at a cell density of 12000 cells per well in endothelial growth medium (ATCC; 2% FBS) and cultured for 48 hr (37 C/ 5%C02/95%C>2). Cell media was replaced with starvation medium (without VEGF and FGF growth factors) containing 0.5% FBS and plates further incubated for 24 hr (37 C/ 5%C02/95%C>2). Cells were pre-incubated with test compound for 1 hr followed by stimulation with rhuVEGFi65 (50 ng/ml, R&D Systems) for 5 min. Media was aspirated off and cells immediately lysed by addition of 50μΙ lysis buffer provided in the HTRF assay kit. 16μΙ of cell lysate from each well was transferred to a white low volume 384 well plate to which propriety kit reagents were added as per kit instructions. Phosphorylation of the VEGFR2 was quantitated by calculating the ratio of fluorescence read at 665nm and 620nm. The percent inhibition of rhuVEGFi65 induced VEGFR2 phosphorylation by test compound was calculated as a percentage of that achieved by rhuVEGFi6s at each concentration of test compound by comparison against vehicle control. The relative 50% inhibitory concentration (RIC50) was determined from the resultant concentration-response curve. Fibroblast gel contraction assay

NHLF's are expanded up to 90% confluence in FGM-2 growth media (Lonza) supplemented with 2% FBS (plus SingleQuot™ growth factors). Fibroblasts are harvested (Trypsin/EDTA) and suspended at 1x10 6 per ml in serum free media. Based on the cell contraction assay kit (Cell Biolabs) a cell lattice is prepared by mixing 1 part cell suspension + 4 parts collagen gel solution as per kit instructions. 0.9ml aliquots of the collagen lattice is added to 1.5 ml centrifuge tubes and treated with final assay concentrations of test compound. 250μΙ of compound treated lattice is then pipetted into each well of a 48 well tissue culture plate (triplicate per test concentration). The plate is incubated for 90 min (37 C/ 5%C02/95%02) to allow the gels to polymerize. 250μΙ of serum free media containing final assay concentrations of test compound are then added to each corresponding gel. After further 30 min incubation, the gels are stimulated with TGF^ (10ng/ml; R&D Systems). Following a 24 hr (37 C/ 5%C02/95%C>2) incubation period, each individual gel is removed and weighed on a precision balance. The effect of the test compound at each concentration is expressed as the percent reversal of the ΤΘΡβΙ induced contraction relative to the vehicle treated basal contraction.

Fibroblast IL-6 release assay NHLF's are expanded up to 90% confluence in FGM-2 growth media (Lonza) supplemented with 2% FBS (plus SingleQuot™ growth factors). Fibroblasts are harvested (Trypsin/EDTA), suspended at 50x10 3 per ml in growth media and 200μΙ added per well (10x10 3 cells/well) of a 96 well tissue culture plate. After 24 hr incubation (37 C/ 5%C02/95%02), cells are serum starved (24 hr) by reducing the media FBS concentration to 0.1 %. Cells are pre-incubated with test compound for 1 hr, then stimulated with ΤΘΡβΙ (5ng/ml, R&D Systems) for 24 hr. Cell free supernatants are recovered for determination of IL-6 concentrations by sandwich ELISA (Duo- set, R&D systems). The inhibition of IL-6 production is calculated as a percentage of that achieved by 5ng/ml ΤΘΡβΙ at each concentration of test compound by comparison against vehicle control. The relative 50% inhibitory concentration (RIC50) is determined from the resultant concentration-response curve.

Mast cell apoptosis

Mast cells are differentiated from cord blood CD34+ cells (Lonza) for 8 weeks in growth media supplemented with 100 ng/ml SCF and 10 ng/ml IL-6. Mast cells are seeded in 384 well white clear bottom plates between 2500 to 10000 cells/well in growth media containing SCF (100 ng/ml). As a positive control for apoptosis, 8 wells are incubated in growth media without SCF. Cells are incubated with test compounds or vehicle for 24 hr (37 C/ 5%C02/95%02). Caspase- 3/7 luminogenic substrate (Caspase-Glo 3/7 Assay, Promega) is added to the cells and incubated at room temperature for 30 min, before reading the luminescence signal. The induction of apoptosis by test compounds is calculated as a percentage of that achieved by cells incubated in the absence of SCF (maximal apoptotic response) for each concentration of test compound compared to vehicle (baseline apoptosis). The relative 50% inhibitory concentration (RIC50) is determined from the resultant concentration-response curve.

The Effect of Test Compounds on Cell Viability

MRC-5 cells were seeded into white clear bottom (for fluorescence/ luminescence reads) or clear (for colorimetric reads) 96 well tissue culture plates at a cell density of 12 x 10 3 cells per well in DMEM growth media (10% FBS). After 24 hr incubation (37 C/ 5%C0 2 /95%0 2 ), the growth media was replaced with media containing 0.1 % FBS plus test compound/ vehicle and incubated for a further 48 hrs. For the colorimetric MTT assay (assessment of cellular metabolic activity), supernatants from each well were aspirated off, replaced with 100 μΙ/well fresh media (0.1 % FBS) and 10 μΙ/well of 5mg/ml MTT. After a 1 hr incubation (37 C/ 5%C0 2 /95%0 2 ) period the media were aspirated off and 100% DMSO (100 μΙ) added to each well. The plates were lightly shaken for 15 minutes prior to reading the absorbance at 550 nm. The percentage loss of cell viability (represented by a reduction in absorbance values) was calculated for each compound concentration relative to vehicle (0.5% DMSO) treated cells. The MultiTox-Fluor Multiplex Cytotoxicity assay in conjunction with the Caspase-Glo 3/7 assay (Promega) were used to measure cellular cytotoxicity/ viability and apoptosis. The MultiTox- Fluor Multiplex Cytotoxicity assay is a single-reagent-addition fluorescent assay that simultaneously measures the relative number of live and dead cells in cell populations. The assay gives ratiometric, inversely correlated measures of cell viability and cytotoxicity. The ratio of viable cells to dead cells is independent of cell number and, therefore, can be used to normalize data. Addition of the single Caspase-Glo® 3/7 reagent in an "add-mix-measure" format results in cell lysis, followed by caspase cleavage of a substrate and generation of a "glow-type" luminescent signal. For this multiplex cytotoxicity assay, 100μΙ of cell supernatants from the 48 hr compound treated cells were carefully removed from each well then 50μΙ MultiTox reagent added (working solution of proprietary MultiTox reagents were prepared by diluting 10μΙ of GF-AFC and bis AAF-R110 into 10ml assay buffer as per kit instructions). Cells were incubated in the dark for 30 minutes before taking two separate fluorescence readings at; 400E x / 505ΕΓΠ (Live cell read) and 485E x / 520ΕΓΠ (Dead cell read). Next, 100μΙ of supernatant was carefully removed from each well and 50μΙ of Caspase 3/7 Glo reagent added to the cell plate and incubated for 30 minutes in the dark. Caspase 3/7 activity was quantitated by reading the luminescence signal. An increase in signal above the vehicle treated control cells represented an increase cell apoptosis.

Fibroblast-to-myofibroblast transition assay

To assess anti-fibrotic activity of test compounds, two alternate protocols were used. In the first, isolated lung fibroblasts are seeded on 96-well plates at 3000 cells/well. Five (5) days post-seeding, cells are refreshed and test compounds or vehicle are added to the cells. After one (1) hour, TGF-βΙ (1.25 ng/mL) is added to induce fibroblast-to-myofibroblast transition. Expression of aSMA, a marker of myofibroblast transition, is measured after 72 hours by immunostaining, assessed by high content imaging on the IN Cell Analyzer 2200 (GE Healthcare) and quantified using a proprietary (BioFocus) algorithm with the IN Cell developer software (GE Healthcare). The output of the algorithm represents the staining intensity multiplied by the stained area (DxA levels). Co-staining of cell nuclei with 4',6-diamidino-2- phenylindole (DAPI) is performed in order to quantify cell number, as a measure of potential toxicity and/or to normalize aSMA for differences in cell density.

In the alternate protocol, isolated lung fibroblasts are seeded on 96-well plates at 5100 cells/well. After 24 hours, cells are refreshed with starve media for a further 24 hours. Test compounds or vehicle are added to the cells. After one (1) hour, TGF-βΙ (0.1 ng/mL) is added to induce fibroblast-to-myofibroblast transition. Expression of aSMA, a marker of myofibroblast transition, is measured after 48 hours by immunostaining, assessed by high content imaging on the ImageXpres micro (Molecular Devices) and quantified using MetaXpress software (Molecular Devices). The percent positive cells is used to evaluate aSMA staining and therefore the degree of fibroblast to myofibroblast transition. Co-staining of cell nuclei with 4', 6- diamidino-2-phenylindole (DAPI) is performed in order to quantify cell number, as a measure of potential toxicity and/or to normalize aSMA for differences in cell density.

Evaluation of duration of action of test compounds against PDGF-BB induced phosphorylation of PDGFRfi in human fetal lung fibroblast cells To determine the relative persistence of the effects of drug exposure on PDGF-BB induced PDGFF^ phosphorylation, a washout experiment is used utilising MRC-5 human fetal lung fibroblast cells and a Homogenous HTRF (Homogeneous Time Resolved Fluorescence) phospho-PDGFRβ (Tyr751) cellular assay kit (Cisbio).that detects receptor modulation of the PDGF-BB induced phosphorylation of PDGFRβ. Washout protocol

MRC-5 cells are initially dislodged with trypsin and neutralized with full media (DMEM growth media, 10% FBS). 1.2x10 6 cells are placed into microfuge tubes, centrifuged at 10,000 rpm for 30 sec utilising a bench top centrifuge, supernatant is removed and cells are washed in 1 ml of pre-warmed (37°C) wash buffer (HBSS pH7.4, 0.1 % BSA and 0.04% Pluronic acid) to remove any residual FBS. Cells are then centrifuged again, supernatant removed and incubated with 500 μΙ_ of vehicle, test control compound and test compound (at concentration giving 70% inhibition) for 1 hrwith gentle shaking at 37°C. Following incubation, cells are dispersed evenly and 100 μΙ_ aliquot is removed from each tube, for a no wash control. The remaining cells are then subjected to 5 repeated wash steps in which cells are centrifuged, supernatant removed and re-suspended in 1 ml of fresh pre-warmed (37°C) wash buffer. To avoid effects of compound carry over due to sticking to plastic, cells are transferred to fresh tubes following each wash step and placed in a 37°C shaker (900 rpm) for 10 minutes incubation between washing to allow for re-equilibration between cells and buffer. Following the final wash step, the cells are re-suspended in 360ul of pre-warmed wash buffer. 5 μΙ_ from both no wash and washed tubes is placed into a 384 white polypropylene microtitre plate (Greiner) and incubated at 37°C for 15 minutes. The cells are then stimulated with 5 μΙ_ of rhuPDGF-BB (3ng/ml, R&D Systems) for 5 min, after which cells are immediately lysed by the addition of 10μΙ of lysis buffer provided in the HTRF assay kit. The cells are placed on a plate shaker (rpm 1450) for 1 hr, after which the plate is briefly centrifuged for 30 sec (3000rpm) and the proprietary HTRF kit reagents added as per kit instructions. Phosphorylation of the PDGFRβ is quantitated by calculating the ratio of fluorescence read at 665nm and 620nm. The percentage inhibition of rhuPDGF-BB induced PDGFRβ phosphorylation by test compound is calculated as a percentage of that achieved at 3 μg/ml rhuPDGF-BB against the test vehicle control. Table 7 presents data as percentage phosphorylation after wash-out relative to maximum phosphorylation of control (which is 100 minus the aforementioned percentage inhibition figure).

PAMPA permeability assay

This assay measures permeability across an artificial membrane and was performed by Cyprotex using a parallel artificial membrane permeability assay. It is an in vitro model of passive, transcellular permeation through an artificial hexadecane membrane, (Wohnsland F et al., Med. Chem., 2001 44; 923-930). The compounds can be categorised into low and high permeability. Generally, compounds which have a P app < 10 x 10 "6 cm/s are classified as low permeability and compounds with a P app > 10 x 10 "6 cm/s are classified as high permeability. Compounds which have low permeability are believed to be likely to have long residency time in the lung due to their slower absorption (Tronde Ann et al., J Pharm. Sci., 2003, 92(6), 1216- 33).

Protocol Summary

Test compound is added to the donor side of a filter coated artificial PAMPA membrane, and permeability is measured by monitoring the appearance of the test compound on the acceptor side of the cell membrane using LC- MS/MS.

Experimental Procedure

A solution of hexadecane in hexane was prepared (5 % v/v) and an aliquot was added onto the membrane of each well in the filter (donor) plate (Multiscreen filter plate for permeability, Millipore). The donor plates were then allowed to dry to ensure evaporation of hexane.

Buffer (pH 7) containing DMSO (5 %) was added to each well of the acceptor plates. Test compound solutions were prepared by diluting 10 mM DMSO concentrates in buffer which gave a final test compound concentration of 10 μΜ (final DMSO concentration 5 %). The fluorescent integrity marker lucifer yellow was also included in the test compound solution. The donor plate was inserted into the acceptor plate and the plates were then incubated at room temperature for 5 hr. Analytical standards were prepared from test compound solutions. Test compound permeability was assessed in quadruplicate. On each plate compounds of known permeability characteristics were run as controls.

At the end of the incubation period the donor plate was removed from the acceptor plate. The donor and acceptor samples for test and control compounds were quantified by LC-MS/MS cassette analysis using a 5 - point calibration with appropriate dilution of the samples. The experimental recovery was calculated from both donor and acceptor compartment concentrations.

If the lucifer yellow permeation was above QC limits for one, two or three individual test compound wells, then n = 3, 2 or 1 results were reported.

Data Analysis

The apparent permeability coefficient (P apP ) for each compound was calculated from the following equation:

ί Λ [dr g

Papp = {C x - ln(l -— v - )}

yarug \ e q U nn, r i U m where C =

(V D + V A )Area x time

where VD and VA are the volumes of the donor and acceptor compartments, respectively, area is surface area of the membrane multiplied by the porosity and the equilibrium drug concentration is the concentration of test compound in the total volume of the donor and acceptor compartments. In Vivo Screening: Pharmacodynamics and Pharmacokinetics

Bleomycin-induced fibrosis in mice (a mouse model of lung fibrosis) C57BL6/J mice are dosed by the intra tracheal route with either vehicle or bleomycin sulphate (MP Biomedicals, 2 U/kg) on day 0. Compounds are administered intra-nasally (as aqueous solutions or suspensions, 25-50 ul) once daily from day 5 until day 20. After a further 24 hr the animals are anesthetized, their tracheas cannulated and animals are mechanically ventilated using a computer-controlled piston ventilator (flexiVent, SCIREQ Inc., Montreal, Canada). Following lung function measurements, mice are exsanguinated and bronchoalveolar lavage fluid (BALF) extracted. Total and differential white cell counts in the BALF samples are measured using a Neubaur haemocytometer. Cytospin smears of the BALF samples are prepared by centrifugation and stained using a DiffQuik stain system (Dade Behring). Cells are counted using oil immersion microscopy. Leftover BALF supernatants are saved for cytokine analysis.

Whole lungs are inflated under 25 cm H2O pressure with 10% neutral buffered formalin through the tracheal cannula and immersed in formalin for at least 24 h. After being processed into paraffin blocks, the lungs are sectioned (5 μηι) and stained with either hematoxylin and eosin (H&E) or picro-sirius red (PSR). A constant number of pictures are randomly taken of 4 transversal sections. Pictures are scored from 0 (no PF) to 8 (maximal PF) by 2 blinded investigators to assess fibrotic changes in the lungs. Additionally, staining for alpha-smooth muscle actin (Thermo Scientific, Freemont, CA) is done using standard methodologies. Soluble collagen in whole lung homogenate is assessed by Sircol collagen assay (Biocolor Ltd, Carrickfergus, UK).

PDGFBB-induced PDGFR phosphorylation in mice

7 to 12-week-old C57BL/6 mice (Charles River) are housed under a 12-hour light/dark cycle and receive food and water ad libitum. Aqueous solutions or suspensions of compounds are prepared to deliver indicated doses (mg/kg) based on the average weight of the mice in the groups. The animals are anesthetized and compounds or vehicles are administered intra- nasally (50 ul). After recovery at indicated time points, the animals are anesthetized again and recombinant mouse PDGF-BB (50 ug/animal, Cambridge Biosciences) or vehicle is administered intra-tracheally, and then, 5 to 30 minutes after PDGF-BB instillation, terminal blood samples are taken and whole lungs are excised. Plasma is isolated from the blood by centrifugation. Approximately one half of the left lobes are homogenised using a FastPrep-24 5G instrument in Matrix D lysing tubes (MP Biomedicals). Phosphorylation levels of PDGFRp are measured by Western blot using anti-phospho-PDGFRa(Y849)/p(Y857), anti-total PDGFRp/a and anti GAPDH antibodies (Cell Signaling). Phosphorylation levels of PDGFRp are reported as ratios to Total PDGFRp levels or to GAPDH. The percentage inhibition of PDGFRp phosphorylation is calculated for each treatment relative to vehicle treatment. Measurements of compound levels in the plasma or lung homogenates are determined by LC- MS/MS.

Pharmacokinetic measurements in rodents Male CD rats or C57BL/6J mice were used for pharmacokinetic studies where animals were dosed intra-tracheally, orally or intravenously for rats and intra-nasally, orally or intravenously for mice.

Animals were dosed intravenously via the lateral tail vein. Animals dosed via the intra-tracheal or intra-nasal route were anaesthetised prior to dosing using isoflurane, and nitrous oxide in rat studies, or oxygen in mouse studies.

Animals were cannulated in the lateral tail vein and placed in a hot-box (37-40°C) approximately 5 minutes before each sampling time-point to dilate the tail veins. Compounds were formulated as solutions for intravenous administration and as solutions or suspensions for all other methods. Animals were weighed on the day of dosing and received a single administration of the formulation with the volume adjusted according to the individual's bodyweight (Table 3). For serial samples, 150-200μΙ rat blood samples or 20 μΙ mouse blood samples (with same volume addition of water) were taken from the cannula port into K2EDTA- coated microtainers at pre-determined time-points over 24 hours. Plasma was isolated by centrifugation (8200rcf for 5 minutes), in rat studies only.

At termination, blood samples were collected through the descending vena cava into K2EDTA- coated microtainers and BAL fluid was obtained by flushing the lungs via the trachea with 3 x 4ml instillations of 4% BSA in PBS for rats and 3 x 0.4ml instillations of 4% BSA in PBS for mice. The lungs were excised, weighed and the weights recorded, and snap-frozen in liquid nitrogen.

Measurements of compound levels in the plasma or blood, lung homogenates and BAL fluid were determined by LC-MS/MS. Drug was extracted by protein precipitation using an excess of solvent containing an appropriate internal standard. Drug concentrations were determined against an external matrix- matched standard curve.

The drug concentrations were plotted against time on a semi-logarithmic plot (see Figures 1 , 2 and 3). Standard pharmacokinetic parameters were calculated using non-compartmental analysis. The concentrations in the lung and BAL fluid at each time point were expressed as the percentage of dose administered. Table 3. Volume of administration

Route of administration Mouse Rat

Intranasal (i.n.) 1 .5ml/kg Not used

Intra-tracheal (i.t.) Not used 0.5ml/kg

Oral (p.o.) 10ml/kg 10ml/kg

Intravenous (i.v.) 2ml/kg 1 ml/kg

Results

Results of testing in the enzyme inhibition assays and in the cellular and other in vitro assays are shown in Tables 4 to 10 below and Figures 1 to 4.

Table 4 The inhibitory activities of compounds of the invention and nintedanib against FGFR1 , FGFR3, PDGFRa, PDGFRp, VEGFR1 and VEGFR2.

134 34.2 19 21.7 341 102

228 15.7 14.9 14.1 313 83.8

75.5 14.6 16 1 1.7 100 37.4

166 7.42 14 1 1 177 38.6

68.7 1 1.7 15.9 9.62 126 33.7

63.7 16.7 15.2 1 1.1 125 39.8

80.8 13.2 13.8 10.2 121 31.7

66 8.14 7.54 10 145 29.8

49.9 13.3 17.8 12.2 85.7 32

67.8 15.2 18.1 15.5 146 51

314 16.2 25.5 19.8 304 76.8

17.8 12.6 12 8.61 96.3 1 1.4

20 10.2 9.1 1 4.77 71.5 13.8

29.1 10.7 12.4 7.88 125 18.2

25 9.56 9.33 7.48 87.8 22.5

27.3 9.56 9.37 7.32 49 20.4

27 10.7 8.74 6.58 76.5 20

28.7 10.7 10.5 4.92 106 19.6

94 20.8 14.4 8.47 259 44.5

91.9 26.4 19.6 15.8 158 43.9

94.2 30.6 39.6 22.1 99.6 58.4

52.3 23.7 13.6 8.46 11 1 37.5

1330

1070 347 238 244 661

(n=1)

90.7 7.68 13.6 6.91 115 43.3

63.9 7.06 6.71 5.39 101 45

68.7 17.9 10.6 7.66 123 49.5

187 27.6 20 15 270 91.1

118 20.2 9.62 7.06 186 49.1

350 50.3 34.6 32.4 329 155

858 105 137 103 534 409

160 29.4 26.2 14.1 256 73.2

346 45.8 53.3 28.1 664 194

122 21.9 15.3 10.2 172 42.6

53.7 18.3 16.6 18.3 231 70

91.6 28 31.3 31.1 206 61.3 50 26.7 9.69 1 1 9.24 77.7 18.4

51 38.3 9.39 1 1.3 9.44 72.3 31.6

52 79.2 33 27.3 17 241 52

53 97.3 36.4 33.4 20.3 184 70.3

54 56.7 25.7 20.2 18.1 120 37.4

55 96.5 23.1 30 21.3 166 71.1

56 29.6 12.4 1 1.4 7.19 66.7 12.8

57 38.4 1 1.2 1 1.2 9.84 57.5 5.36

58 53.3 8.96 1 1.3 8.87 74.8 3.86

59 34.1 1 1.5 14.7 9.19 44.7 5.43

60 54.2 12.2 16.6 13 63.7 5.24

61 97.5 16.4 27.3 18.3 110 1 1.8

62 47.7 6.78 1 1.6 1 1.2 72.6 5.74

63 67.5 22 16.4 15.7 182 16.7

64 ND ND ND ND ND ND

65 25 7.9 13 13 79 79

66 50 6.3 20 16 100 20

67 ND ND ND ND ND ND

68 63 63 ND ND 126 32

69 79 32 ND ND 200 40

70 32 13 ND ND 200 25

71 126 25 ND ND 398 100

72 126 16 ND ND 200 79

73 40 7.9 13 7.9 100 16

ND: Not determined; n=2 unless otherwise indicated (mean); *n=34

Table 5 The inhibitory effects of the compounds of the invention and nintedanib on phosphorylation of PDGFRp in fibroblasts induced by PDGFBB in MRC5 cells and on phosphorylation of VEGFR2 in endothelial cells induced by VEGF165.

1 ND 32.2

2 39.6 ND

3 7.14 13.9 (n=2)

4 15.2 (n=3) 23.9 (n=2)

5 7.74 4.17 (n=2)

6 6.95 (n=2) 6.58 (n=6)

7 10.4 (n=2) 12.5

8 21.5 39.5

9 8.45 ND

10 13.6 ND

11 15.3 22.6 (n=2)

12 ND 31.1

13 4.4 13.9 (n=2)

14 ND 9.32

15 19.9 30.1 (n=1)

16 ND ND

17 ND ND

18 3.68 4.47

19 5.95 54.7 (n=2)

20 ND ND

21 6.79 25

22 6.68 8.26

23 ND ND

24 ND 8.21

25 19.4 52.1

26 5.39 5.47 (n=2)

27 10.4 ND

28 5.03 5.1

29 3.18 30.7

30 7.43 10.2

31 7.72 46.4

32 3.09 11.4

33 8.99 83.5

34 13.4 24.2

35 26.9 94.2

36 9.44 12.6 37 24.1 71.1

38 14.6 56.7

39 33.8 38.1

40 29.2 157

41 37.4 ND

42 56.3 103

43 25.7 33.8

44 73.9 ND

45 27.2 124

46 31.7 128

47 39 371

48 38.8 ND

49 40.2 23.5

50 9.42 26.8

51 9.39 19.1

52 21.8 17

53 43.8 33.6

54 34.1 38.1

55 17.3 17

56 3.95 7.54

57 5.04 ND

58 4.06 ND

59 25.4 ND

60 5.6 ND

61 7.84 ND

62 4.61 ND

63 4.74 ND

64 ND 32.2

65 9.3 15.5

66 15.5 17.8

67 ND ND

68 16.5 ND

69 14.3 12.9

70 9.98 8.87

71 23.2 102

72 36.7 172 73 12.7 7.83

ND: Not e etermined; n=1 unless otherwise indicated

Table 6 The effect of the compounds of the invention and nintedanib on cell viability of MRC5 cells (MTT assay).

6 88 111.4 111.8 (n=3) 86.4 88.6 94.9 8 104.8 117 110.2 9 91.8 85.8 92 0 26 87.7 96.1 1 95.7 97.3 111.7 2 66.3 82.3 93.5 (n=2) 91.3 101.3 110.7 4 61.5 80 114.8 5 75.7 77.2 92.9 6 65.9 93 108.8 7 106 98.9 95.2 8 93 90.8 89.3 9 90.8 90.1 89.9 0 110.5 103.8 103.8 1 86.8 96.1 112.3 2 118.3 109.6 114.5 3 83.9 91.8 96.5 4 109.8 105.9 103 5 80.7 104.7 98.9 6 86.3 109.5 120.7 7 93 102.4 112 8 103.5 121 132.6 9 69.4 110.8 124.5 0 69.7 92.6 107.5 1 71.2 85.8 86.5 2 59.8 50.9 62 3 97.8 96.8 124.2 4 92.9 99.8 101 5 92.3 111.1 106.7 6 70.5 121 132 7 91.2 123.2 123.5 8 55.6 116.9 110.2 9 103.8 107.7 123.5 0 0.2 84.4 88.8 1 35.2 86.6 83 62 -0.2 73.8 80.7

63 12.1 1 16.9 99

64 73.7 88.6 86.5

65 70.5 1 10.3 1 18.6

66 53.8 92.9 102.8

67 ND ND ND

68 ND ND ND

69 74.0 93.1 92.5

70 94.0 99.7 91.7

71 ND ND ND

72 ND ND ND

73 96.8 104 82.9

ND: Not determined; n=1 unless otherwise indicated

Table 7 Duration of action of example compounds as inhibitors of PDGF-BB induced phosphorylation of PDGFF^ in human fetal lung fibroblast cells. Percentage phosphorylation after wash-out relative to maximal phosphorylation (100%) of 3 μg/ml rhuPDGF-BB of test vehicle control.

Nintedanib 30.9 2 4.8

57 73.1 2 7.3

73 69.3 2 1.7

4 15.9 1

Since lower values indicate higher percentage inhibition of phosphorylation of PDGFF^, the data of Table 7 reveals that Example compounds 4 and 6 have longer duration of action as inhibitors of PDGFF^ than nintedanib in this assay.

Table 8 Determination of P app for compounds of the invention in the PAMPA permeability assay

17 0.0142 0.00675 4 91.9

18 0.424 0.138 4 51.5

19 0.00329 0.000375 4 54.6

20 0.0305 0.0153 4 91.7

20 0.0296 0.0159 4 66.5

21 ND ND ND ND

22 ND ND ND ND

23 20 2.73 4 51.8

24 44.8 13.6 4 71.8

25 30 6.72 4 41.5

26 0.0156 0.0047 4 40.2

27 0.00146 0.000782 4 61.2

28 0.00407 0.00136 4 69.0

29 0.00415 0.00238 4 65.1

30 0.00225 0.000845 4 87.0

31 ND ND ND ND

32 0.0192 0.0103 4 42.8

33 0.00743 0.00316 3 71.9

34 ND ND ND ND

35 ND ND ND ND

36 0.0301 0.00466 3 53.1

37 ND ND ND ND

38 0.00569 0.00194 3 55.4

39 ND ND ND ND

40 0.00954 0.00451 4 75.0

41 0.00258 ND 1 59.6

42 0.0264 ND 1 76.8

43 0.00798 0.00642 4 75.3

44 ND ND ND ND

45 ND ND ND ND

46 ND ND ND ND

47 ND ND ND ND

48 0.220 0.107 4 67.4

49 0.0768 0.0158 4 75.2

50 0.0566 0.0253 2 179

51 0.00306 0.00084 4 62 51 0.00474 0.00333 4 74.7

52 0.279 0.0567 4 76.2

53 ND ND ND ND

54 ND ND ND ND

55 0.154 0.0407 2 59.1

56 0.719 0.154 4 69.7

57 0.688 ND 1 50.5

58 0.0296 0.00422 2 49.5

59 0.0364 0.0120 4 69.6

60 0.0503 0.0105 3 69.2

61 0.0205 0.00130 3 64.4

62 0.00628 0.000584 4 72.8

63 ND ND ND ND

64 0.0679 0.0169 2 59.4

ND = not determined *values from a repeat are shown for some compounds

Table 9 Pharmacokinetic measurements over a period of 24 hours in rats dosed intravenously (i.v.) and intra-tracheally (i.t.) with compounds of the invention or nintedanib.

24 NC 98 198 BLQ NC 103 ND

BAL % administered

dose at time (h)

0.1 0.06 0.04 0.06 0.01 0.03 0.09 BLQ

1.5 0.03 0.01 0.03 BLQ 0.02 0.02 BLQ

6 ND 0.01 ND ND ND ND BLQ

24 BQL 0.01 BQL BLQ BLQ 0.01 BLQ i.t. dosing studies

i.t. dose 0.42 0.1 0.5 0.1 0.1 0.1 0.1 i.t. BAV (%) 43 70 115 67 325 ND ND lung concentration (nM)

at time (h)

0.1 12396 3593 30413 5543 6693 3433 2100

1.5 7699 1710 11876 1538 1684 1370 881

6 ND 629 ND ND ND ND 1005

24 152 747 4724 218 247 135 519 lung/ plasma cone ratio

at time(h)

0.1 1227 74 288 279 176 177 233

1.5 1607 362 2893 1321 1426 884 114

6 ND 461 ND ND ND ND 114

24 NC 1519 3282 NC 503 314 ND

BAL % administered

dose at time (h)

0.1 2 3.1 6 3 3 2.4 5.3

1.5 1 0.6 3 0.3 0.3 0.6 0.2

6 ND 0.3 ND ND ND ND 0.3

24 0.1 0.2 0.1 0.1 0.1 0.1 0.1

Lung cone i.t./ i.v.

Ratio at time (h)

0.1 5.4 7.6 10.2 13.2 9.3 6.9 6

1.5 5.5 6.8 6.3 5.8 5.2 6.6 5

6 ND 6.8 ND ND ND ND 15

24 2.3 14.4 27.3 18.2 8.8 2.0 17

Oral dosing studies

Oral dose (mg/kg) - 3 3 - - - 3 Oral BAV (%) - 4 <1 - - - ND

ND: Not determined

NC: Not calculable due to one parameter being BLQ

BLQ: Below level of Quantitation Table 10 Pharmacokinetic measurements over a period of 24 hours in mice dosed intravenously (i.v.) and intra-nasally (i.n.) with compounds of the invention or nintedanib.

1.5 804 1006 986 593 1728 ND

6 ND ND 258 219 1322 ND

24 15 2 84 17 363 ND lung/ plasma cone ratio at time(h)

0.1 42 ND 79 ND 209 ND

1.5 27 ND 398 ND 688 ND

6 ND ND 607 ND 1 127 ND

24 NC ND NC ND NC ND

BAL % administered dose at time

(h)

0.1 0.5 ND 1 ND 1 ND

1.5 0.1 ND 0.3 ND 0.1 ND

6 ND ND 0.1 ND 0.3 ND

24 NC ND 0.04 ND 0.1 ND

Lung cone i.n. / i.v. Ratio at time

(h)

0.1 1.2 ND 2.6 ND 4.0 ND

1.5 0.6 ND 5.9 ND 4.2 ND

6 ND ND 5.6 ND 4.3 ND

24 1.5 ND 6.5 ND 18.2 ND

Oral dosing studies

oral dose (mg/kg) 60 ND 10 ND 10 ND

BAV (%) 32 5 <5 13 5

ND: Not determined

NC: Not calculable due to one parameter being BLQ

BLQ: Below level of Quantitation

sd: Standard deviation

Summary of results

The compounds of the invention, as disclosed herein, demonstrate inhibitory activity against FGFR1 , FGFR3, PDGFRa, PDGFRp, VEGFR1 and VEGFR2 (Table 4 and Table 5) and nanomolar potency. A vast majority of the tested compounds show no adverse effects in the cell viability assays (Table 6). Furthermore, the compounds generally have low permeability across membranes (Table 8 and Figure 1). Certain compounds showed evidence of increased duration of action with respect to nintedanib as determined with respect to inhibition of PDGF- BB-induced phosphorylation of human fetal lung fibroblast cells (see Table 7). The pharmacokinetic measurements show that generally the compounds of the invention are found to have a higher relative content remaining in the lung 24 hours following topical delivery after the subject receives the dose than nintedanib (Tables 9 and 10 and Figures 2 to 4). The low BAL levels noted together with the reducing amounts over a 24 h period indicate rapid dissolution and absorption into the lung tissue is occurring with compounds of the invention.

This profile indicates that compounds of the invention are expected to be suitable as medicines inter alia for the treatment of fibrotic diseases or interstitial lung diseases, such as IPF, or respiratory disorders, especially when delivered topically to the lung.

References

Castriotta RJ, et al., Chest, 2010, 138(3):693-703

Du Bois RM., Nat Rev Drug Discov., 2010, 9(2): 129-40

Fehrenbach. H., et al., Virchows Arch., 1999, 435(1):20-31

King TE Jr, et al., Lancet, 2011 , 3;378(9807): 1949-61

Lindroos. P., Am J Physio Lung Cell Mol Physiol., 2001 , 280:L354-L362

Tronde Ann et al., J Pharm. Sci., 2003, 92(6), 1216-33

Selman M, et al., Ann Intern Med., 2001 , 16;134(2): 136-51

Wohnsland F et aL, Med Chem., 2001 , 44;923~930

Throughout the specification and the claims which follow, unless the context requires otherwise, the word 'comprise', and variations such as 'comprises' and 'comprising', will be understood to imply the inclusion of a stated integer, step, group of integers or group of steps but not to the exclusion of any other integer, step, group of integers or group of steps.

All patents and patent applications referred to herein are incorporated by reference in their entirety.