Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOUNDS
Document Type and Number:
WIPO Patent Application WO/2016/185160
Kind Code:
A1
Abstract:
A first aspect of the- invention relates to a compound of formula (I), or a pharmaceutically acceptable salt or ester thereof, (Formula (I)) wherein: R1 is (CH2)mNR11R12;R2 is selected from H, halo, OR13, ΝΗR13, alkyl, alkenyl and alkynyl; R3 is selected from alkyl, alkenyl, alkynyl, aryl, halo, aryloxy, NHCO2R4, NHCONR5R6, NHCOR7, NH-alkyl, NH-alkenyl, NH(CH2);n-aryl, (CH2)p-heteroaryl, (CH2)qCO2R8, (CH2)rCOR9 and NHSO2R10, wherein each alkyl, alkenyl, aryl or heteroaryl moiety in the aforementioned list is optionally further substituted by one or more groups selected from alkyl, halo OH, NH2, alkoxy, aryloxy, alkylamino, arylamino, carboxyl and carboxamide; R4 to R10 and R13 are each independently selected from alkyl, alkenyl and aryl; R11 and R12 are each independently selected from alkyl and alkenyl; or R11 and R:12 are linked together with the nitrogen to which they are attached to form a heterocycloalkyl or heterocycloalkenyl group; n, m, p, q and r are each independently selected from 0, 1, 2,.3. 4, 5 and 6. Further aspects relate to pharmaceutical compositions, therapeutic uses and process for preparing according to the invention.

Inventors:
UNCITI-BROCETA ASIER (GB)
FRASER CRAIG (GB)
O CARRAGHER NEIL (GB)
Application Number:
PCT/GB2016/051057
Publication Date:
November 24, 2016
Filing Date:
April 15, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV COURT UNIV OF EDINBURGH (GB)
International Classes:
C07D487/04; A61K31/519; A61P25/28; A61P31/12; A61P35/00
Domestic Patent References:
WO2013177668A12013-12-05
WO2002076986A12002-10-03
WO2015018333A12015-02-12
WO1984003564A11984-09-13
Other References:
"Handbook of Pharmaceutical Excipients", 1994
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING CO.
BERGE ET AL., J PHARM SCI, vol. 66, 1977, pages 1 - 19
"Advanced Organic Chemistry", 1985, JOHN WILEY AND SONS
HUGHES, J. P.; REES, S.; KALINDJIAN, S. B.; PHILPOTT, K. L.: "Principles of early drug discovery", BR. J. PHARMACOL., vol. 162, 2011, pages 1239 - 1249
SMITH, A: "Screening for drug discovery", NATURE, vol. 418, 2002, pages 451 - 463
HOLDGATE, G ET AL.: "Biophysical Methods in Drug Discovery from Small Molecule to Pharmaceutical", METHODS MOL. BIOL., vol. 1008, 2013, pages 327 - 355
MCLNNES, C: "Virtual screening strategies in drug discovery", CURRO OPIN. CHEM. BIOL., vol. 11, 2007, pages 494 - 502
VISTOLI, G.; PEDRETTI, A.; TESTA, B: "Assessing drug-likeness-what are we missing?", DRUG DISCOV. TODAY, vol. 13, 2008, pages 285 - 294, XP022594177, DOI: doi:10.1016/j.drudis.2007.11.007
KAMB, A.; WEE, S.; LENGAUER, C.: "Why is cancer drug discovery so difficult?", NAT. REV. DRUG DISCOV., vol. 6, 2007, pages 115 - 20
CHIN, L.; GRAY, J. W.: "Translating insights from the cancer genome into clinical practice", NATURE, vol. 452, 2008, pages 553 - 563
STOMMEL, J. M. ET AL.: "Coactivation of receptor tyrosine kinases affects the response of tumor cells to targeted therapies", SCIENCE, vol. 318, 2007, pages 287 - 290, XP002484746, DOI: doi:10.1126/science.1142946
CARRAGHER, N.; UNCITI-BROCETA, A.; CAMERON, D.: "Advancing cancer drug discovery towards more agile development of targeted combination therapies", FUT. MED. CHEM., vol. 4, 2012, pages 87 - 105
KNIGHT, Z. A.; LIN, H.; SHOKAT, K. M.: "Targeting the cancer kinome through polypharmacology", NAT. REV. CANCER., vol. 10, 2010, pages 130 - 137, XP055186104, DOI: doi:10.1038/nrc2787
KOLA, I.; LANDIS, J.: "Can the pharmaceutical industry reduce attrition rates?", NAT. REV. DRUG DISCOV., vol. 3, 2004, pages 711 - 716
SASSOON, I.; BLANC, V: "Antibody-drug conjugate (ADC) clinical pipeline: a review", METHODS MOL. BIOL., vol. 1045, 2013, pages 1 - 27
VELEMA, W. A.; SZYMANSKI, W.; FERINGA, B. L.: "Photopharmacology: beyond proof of principle", J. AM. CHEM. SOC., vol. 136, 2014, pages 2178 - 2191
VERSTEEGEN, R. M.; ROSSIN, R.; TEN HOEVE, W.; JANSSEN H. M.; ROBILLARD, M. S.: "Click to Release: Instantaneous Doxorubicin Elimination upon Tetrazine Ligation", ANGEW. CHEM. INT. ED. ENGL., vol. 52, 2013, pages 14112 - 14116, XP055105238, DOI: doi:10.1002/anie.201305969
CLAVEL, C. M.: "Thermoresponsive Chlorambucil Derivatives for Tumour Targeting", ANGEW. CHEM., INT. ED., vol. 50, 2011, pages 7124 - 7127
WEISS, J. T: "Extracellular palladium-catalyzed dealkylation of 5-fluoro-1-propargyl-uracil as a bioorthogonally-activated prodrug approach", NAT. COMMUN., vol. 5, 2014, pages 3277
WEISS, J. T. ET AL.: "Development and Bioorthogonal Activation of Palladium-Labile Prodrugs of Gemcitabine", J. MED. CHEM., vol. 57, 2014, pages 5395 - 5404, XP055408793, DOI: doi:10.1021/jm500531z
LEE, J. A.; UHLIK, M. T.; MOXHAM, C. M.; TOMANDL, D.; SAIL, D. J.: "Modern phenotypic drug discovery is a viable, neoclassic pharma strategy", J. MED. CHEM., vol. 55, 2012, pages 4527 - 4538
EDER, J.; SEDRANI, R.; WIESMANN, C: "The discovery of first-in-class drugs: origins and evolution", NAT. REV. DRUG DISCOV., vol. 13, 2014, pages 577 - 587
CARRAGHER, N. 0.; BRUNTON, V. G.; FRAME, M. C.: "Combining imaging and pathway profiling: an alternative approach to cancer drug discovery", DRUG DISCOV. TODAY, vol. 17, 2012, pages 203 - 214, XP028403167, DOI: doi:10.1016/j.drudis.2012.02.002
COHEN, P: "Protein kinases-the major drug targets of the twenty-first century?", NAT. REV. DRUG. DISCOV., vol. 1, 2002, pages 309 - 315, XP008125595, DOI: doi:10.1038/nrd773
ZHANG, J.; YANG, P. L.; GRAY, N. S.: "Targeting cancer with small molecule kinase inhibitors", NAT. REV. CANCER, vol. 9, 2009, pages 28 - 39, XP002570143, DOI: doi:10.1038/NRC2559
LU, L. ET AL.: "Hippo signaling is a potent in vivo growth and tumor suppressor pathway in the mammalian liver", PROC. NATL. ACAD. SCI. USA, vol. 107, 2010, pages 1437 - 1442
ZHOU, B.-B. S.; BARTEK, J.: "Targeting the checkpoint kinases: chemosensitization versus chemoprotection", NAT. REV. CANCER, vol. 4, 2004, pages 216 - 225, XP002382043
FUJIMOTO, H. ET AL.: "Regulation of the antioncogenic Chk2 kinase by the oncogenic Wip1 phosphatase", CELL DEATH DIFFER., vol. 13, 2006, pages 1170 - 1180
WILHELM, S. ET AL.: "Discovery and development of sorafenib: a multikinase inhibitor for treating cancer", NAT. REV. DRUG DISCOV., vol. 5, 2006, pages 835 - 844, XP002693738, DOI: doi:10.1038/NRD2130
GREUBER, E. K.; SMITH-PEARSON, P.; WANG, J.; PENDERGAST, A. M.: "Role of ABL family kinases in cancer: from leukaemia to solid tumours", NAT. REV. CANCER, vol. 13, 2013, pages 559 - 571, XP055215234, DOI: doi:10.1038/nrc3563
NOREN, N. K.; FOOS, G.; HAUSER, C. A.; PASQUALE, E. B.: "The EphB4 receptor suppresses breast cancer cell tumorigenicity through an Abl-Crk pathway", NAT. CELL BIOI., vol. 8, 2006, pages 815 - 825, XP055496904, DOI: doi:10.1038/ncb1438
ALLINGTON, T. M.; GALLIHER-BECKLEY, A. J.; SCHIEMANN, W. P.: "Activated Abl kinase inhibits oncogenic transforming growth factor-beta signaling and tumorigenesis in mammary tumors", FASEB J., vol. 23, 2009, pages 4231 - 4243
GIL-HENN, H. ET AL.: "Arg/Abl2 promotes invasion and attenuates proliferation of breast cancer in vivo", ONCOGENE, vol. 32, 2013, pages 2622 - 2630
HANKE, J. H. ET AL.: "Discovery of a novel, potent, and Src family-selective tyrosine kinase inhibitor. Study of Lck- and FynT-dependent T cell activation", J. BIOL. CHEM., vol. 271, 1996, pages 695 - 701
TATTON, L; MORLEY, G. M.; CHOPRA, R.; KHWAJA, A: "The Src-selective kinase inhibitor PP1 also inhibits Kit and Bcr-Abl tyrosine kinases", J. BIOL. CHEM., vol. 278, 2003, pages 4847 - 4853, XP002350399, DOI: doi:10.1074/jbc.M209321200
JESTER, B. W.; GAJ, A.; SHOMIN, C. D.; COX, K. J.; GHOSH, I: "Testing the promiscuity of commercial kinase inhibitors against the AGC kinase group using a split-luciferase screen", J. MED. CHEM., vol. 55, 2012, pages 1526 - 1537, XP055080234, DOI: doi:10.1021/jm201265f
DINER, P.; ALAO, J. P.; SODERLUND, J.; SUNNERHAGEN, P.; GRATLI, M: "Preparation of 3-substituted-1 -isopropyl-1 H-pyrazolo[3,4-d]pyrimidin-4-amines as RET kinase inhibitors", J. MED. CHEM., vol. 55, 2012, pages 4872 - 4876, XP055363610, DOI: doi:10.1021/jm3003944
APSEL, B. ET AL.: "Targeted polypharmacology: discovery of dual inhibitors of tyrosine and phosphoinositide kinases", NAT. CHERN. BIOL., vol. 4, 2008, pages 691 - 699, XP007909493, DOI: doi:10.1038/nchembio.117
ANTONELLI, A. ET AL.: "CLM29, a multi-target pyrazolopyrimidine derivative, has antineoplastic activity in medullary thyroid cancer in vitro and in vivo", MOL. CELL ENDOCRINOL., vol. 393, 2014, pages 56 - 64
SCHINDLER, T. ET AL.: "Crystal structure of Hck in complex with a Src family-selective tyrosine kinase inhibitor", MOL. CELL., vol. 3, 1999, pages 639 - 648
KNOWLES, P. P. ET AL.: "Structure and chemical inhibition of the RET tyrosine kinase domain", J. BIOL. CHEM., vol. 281, 2006, pages 33577 - 87, XP055421767, DOI: doi:10.1074/jbc.M605604200
LIU, Y.; GRAY, N. S.: "Rational design of inhibitors that bind to inactive kinase conformations", NAT. CHEM. BIOL., vol. 2, 2006, pages 358 - 364, XP002487804, DOI: doi:10.1038/nchembio799
LIU, Y. ET AL.: "Structural basis for selective inhibition of Src family kinases by PP1", CHEM BIOI., vol. 6, pages 671 - 678, XP026906323, DOI: doi:10.1016/S1074-5521(99)80118-5
DAR, A. C.; LOPEZ, M. S.; SHOKAT, K. M.: "Small molecule recognition of c-Src via the Imatinib-binding conformation", CHEM. BIOL., vol. 15, 2008, pages 1015 - 1022, XP026046172, DOI: doi:10.1016/j.chembiol.2008.09.007
HANN, M. M.; KESERU, G. M.: "Finding the sweet spot: the role of nature and nurture in medicinal chemistry", NAT. REV. DRUG DISCOV., vol. 11, 2012, pages 355 - 365
HSIEH, A. C. ET AL.: "The translational landscape of mTOR signalling steers cancer initiation and metastasis", NATURE, vol. 485, 2012, pages 55 - 61, XP055120586, DOI: doi:10.1038/nature10912
KERKELA R; GRAZETTE L; YACOBI R; LLIESCU C; PATTEN R; BEAHM C; WALTERS B; SHEVTSOV S; PESANT S; CLUBB FJ: "Cardiotoxicity of the cancer therapeutic agent imatinib mesylate", NATURE MED, vol. 12, no. 8, August 2006 (2006-08-01), pages 908 - 16, XP007905631, DOI: doi:10.1038/nm1446
BOYCE, B.F.; XING, L.; YAO, Z.; YAMASHITA, T.; SHAKESPEARE, W.C.; WANG, Y.; METCALF, C.A., 3RD; SUNDARAMOORTHI, R.; DALGARNO, D.C.: "SRC inhibitors in metastatic bone disease", CLIN CANCER RES, vol. 12, 2006, pages 6291S - 6295S, XP008136816, DOI: doi:10.1158/1078-0432.CCR-06-0991
BRUNTON, V.G.; FRAME, M.C.: "Src and focal adhesion kinase as therapeutic targets in cancer", CURR OPIN PHARMACOL, vol. 8, 2008, pages 427 - 432, XP025428943, DOI: doi:10.1016/j.coph.2008.06.012
CHATURVEDI, D.; GAO, X.; COHEN, M.S.; TAUNTON, J.; PATEL, T.B.: "Rapamycin induces transactivation of the EGFR and increases cell survival", ONCOGENE, vol. 28, 2009, pages 1187 - 1196
CREEDON, H.; BRUNTON, V.G.: "Src kinase inhibitors: promising cancer therapeutics?", CRIT REV ONCOG, vol. 17, 2012, pages 145 - 159
GIROTTI R ET AL.: "Paradox-Breaking RAF Inhibitors that Also Target SRC Are Effective in Drug-Resistant BRAF Mutant Melanoma", CANCER CELL, vol. 27, no. 1, 2015, pages 85 - 96, XP029131331, DOI: doi:10.1016/j.ccell.2014.11.006
KERKELA R; GRAZETTE L; YACOBI R; LLIESCU C; PATTEN R; BEAHM C; WALTERS B; SHEVTSOV S; PESANT S; CLUBB FJ: "Cardiotoxicity of the cancer therapeutic agent imatinib mesylate", NATURE MED, vol. 12, no. 8, 2006, pages 908 - 16, XP007905631, DOI: doi:10.1038/nm1446
QIU Z; GANG Y; GOFF SP: "Abl family tyrosine kinases are essential for basement membrane integrity and cortical lamination in the cerebellum", J NEUROSCI., vol. 30, no. 43, 27 October 2010 (2010-10-27), pages 14430 - 9
DE WISPELAERE, M.; LACROIX, A.J.; YANG, P.L.: "The small molecules AZD0530 and dasatinib inhibit dengue virus RNA replication via Fyn kinase", J VIROL, vol. 87, 2013, pages 7367 - 7381, XP055318961, DOI: doi:10.1128/JVI.00632-13
DUXBURY, M.S.; !TO, H.; ZINNER, M.J.; ASHLEY, S.W.; WHANG, E.E.: "Inhibition of SRC tyrosine kinase impairs inherent and acquired gemcitabine resistance in human pancreatic adenocarcinoma cells", CLIN CANCER RES, vol. 10, 2004, pages 2307 - 2318
GIL-HENN, H.; PATSIALOU, A.; WANG, Y.; WARREN, M. S.; CONDEELIS, J. S.; KOLESKE, A. J.: "Arg/Abl2 promotes invasion and attenuates proliferation of breast cancer in vivo", ONCOGENE, vol. 32, 2013, pages 2622 - 2630
HAWTHORNE, V.S.; HUANG, W.C.; NEAL, C.L.; TSENG, L.M.; HUNG, M.C.; YU, D.: "ErbB2-mediated Src and signal transducer and activator of transcription 3 activation leads to transcriptional up-regulation of p21Cip1 and chemoresistance in breast cancer cells", MOL CANCER RES, vol. 7, 2009, pages 592 - 600
KOPETZ, S.; LESSLIE, D.P.; DALLAS, N.A.; PARK, S.I.; JOHNSON, M.; PARIKH, N.U.; KIM, M.P.; ABBRUZZESE, J.L.; ELLIS, L.M.; CHANDRA,: "Synergistic activity of the SRC family kinase inhibitor dasatinib and oxaliplatin in colon carcinoma cells is mediated by oxidative stress", CANCER RES, vol. 69, 2009, pages 3842 - 3849
MAYER, E.L.; KROP, I.E.: "Advances in targeting SRC in the treatment of breast cancer and other solid malignancies", CLIN CANCER RES, vol. 16, 2010, pages 3526 - 3532
MCCARTHY, S.D.; JUNG, D.; SAKAC, D.; BRANCH, D.R.: "c-Src and Pyk2 protein tyrosine kinases play protective roles in early HIV-1 infection of CD4+ T-cell lines", J ACQUIR IMMUNE DEFIC SYNDR, vol. 66, 2014, pages 118 - 126
MURRILLS, R.J.; FUKAYAMA, S.; BOSCHELLI, F.; MATTEO, J.J.; OWENS, J.; GOLAS, J.M.; PATEL, D.; LANE, G.; LIU, Y.B.; CARTER, L.: "Osteogenic effects of a potent Src-over-Abl-selective kinase inhibitor in the mouse", J PHARMACOL EXP THER, vol. 340, 2012, pages 676 - 687
MYOUI, A.; NISHIMURA, R.; WILLIAMS, P.J.; HIRAGA, T.; TAMURA, D.; MICHIGAMI, T.; MUNDY, G.R.; YONEDA, T.: "C-SRC tyrosine kinase activity is associated with tumor colonization in bone and lung in an animal model of human breast cancer metastasis", CANCER RES, vol. 63, 2003, pages 5028 - 5033
NOREN, N. K.; FOOS, G.; HAUSER, C. A.; PASQUALE, E. B: "The EphB4 receptor suppresses breast cancer cell tumorigenicity through an Abl-Crk pathway", NAT. CELL BIOL., vol. 8, 2006, pages 815 - 825, XP055496904, DOI: doi:10.1038/ncb1438
PARK, G.B.; KIM, D.; KIM, Y.S.; KIM, S.; LEE, H.K.; YANG, J.W.; HUR, D.Y.: "The Epstein-Barr virus causes epithelial-mesenchymal transition in human corneal epithelial cells via Syk/src and Akt/Erk signaling pathways", INVEST OPHTHALMOL VIS SCI, vol. 55, 2014, pages 1770 - 1779
SAWYERS, C.L.; MCLAUGHLIN, J.; GOGA, A.; HAVLIK, M.; WITTE, O.: "The nuclear tyrosine kinase c-Abl negatively regulates cell growth", CELL, vol. 77, 1994, pages 121 - 131, XP024245808, DOI: doi:10.1016/0092-8674(94)90240-2
WHEELER, D.L.; LIDA, M.; KRUSER, T.J.; NECHREBECKI, M.M.; DUNN, E.F.; ARMSTRONG, E.A.; HUANG, S.; HARARI, P.M.: "Epidermal growth factor receptor cooperates with Src family kinases in acquired resistance to cetuximab", CANCER BIOL THER, vol. 8, 2009, pages 696 - 703
ZHANG, S.; HUANG, W.C.; LI, P.; GUO, H.; POH, S.B.; BRADY, S.W.; XIONG, Y.; TSENG, L.M.; LI, S.H.; DING, Z.: "Combating trastuzumab resistance by targeting SRC, a common node downstream of multiple resistance pathways", NAT MED, vol. 17, 2011, pages 461 - 469
SUMMY JM; GALLICK GE: "Src family kinases in tumor progression and metastasis", CANCER METASTASIS REV., vol. 22, no. 4, 2003, pages 337 - 58
FRAME MC ET AL.: "v-Src's hold over actin and cell adhesions", NAT REV MOL CELL BIOL., vol. 3, no. 4, 2002, pages 233 - 45
YOO SK ET AL.: "Early redox, Src family kinase, and calcium signaling integrate wound responses and tissue regeneration in zebrafish", J CELL BIOL, vol. 199, 2012, pages 225 - 234
TYRYSHKIN A ET AL.: "SRC kinase is a novel therapeutic target in lymphangioleiomyomatosis", CANCER RES, vol. 74, no. 7, 1 April 2014 (2014-04-01)
WALCHER D ET AL.: "C-Peptide induces vascular smooth muscle cell proliferation: involvement of SRC-kinase, phosphatidylinositol 3-kinase, and extracellular signal-regulated kinase 1/2", CIRCULATION RESEARCH, vol. 99, 2006, pages 1181 - 1187, XP002517639, DOI: doi:10.1161/01.RES.0000251231.16993.88
CHO HM ET AL.: "The Src/PLC/PKC/MEK/ERK signaling pathway is involved in aortic smooth muscle cell proliferation induced by glycated LDL", MOLECULES AND CELLS, vol. 19, no. 1, 2005, pages 60 - 66
YANG K1; BELROSE J; TREPANIER CH; LEI G; JACKSON MF; MACDONALD JF: "Fyn, a potential target for Alzheimer's disease", J ALZHEIMERS DIS., vol. 27, 2011, pages 243 - 52
NYGAARD HB; VAN DYCK CH; STRITTMATTER SM: "Fyn kinase inhibition as a novel therapy for Alzheimer's disease", ALZHEIMERS RES THER., vol. 6, 2014, pages 8, XP021193618, DOI: doi:10.1186/alzrt238
KAUFMAN AC1; SALAZAR SV; HAAS LT; YANG J; KOSTYLEV MA; JENG AT; ROBINSON SA; GUNTHER EC; VAN DYCK CH; NYGAARD HB: "Fyn inhibition rescues established memory and synapse loss in Alzheimer mice", ANN NEUROL., 2015
NYGAARD HB; WAGNER AF; BOWEN GS; GOOD SP; MACAVOY MG; STRITTMATTER KA; KAUFMAN AC; ROSENBERG BJ; SEKINE-KONNO T; VARMA P: "A phase lb multiple ascending dose study of the safety, tolerability, and central nervous system availability of AZD0530 (saracatinib) in Alzheimer's disease", ALZHEIMERS RES THER., vol. 7, 2015, pages 35, XP021215783, DOI: doi:10.1186/s13195-015-0119-0
H. MA; S. DEACON; K. HORIUCHI: "The challenge of selecting protein kinase assays for lead discovery optimization", EXPERT OPIN. DRUG DISCOV., vol. 3, no. 6, 2008
PATTON, E. E.; DHILLON, P.; AMATRUDA, J. F.; RAMAKRISHNAN, L.: "Spotlight on zebrafish: translational impact", DIS. MODEL. MECH., vol. 7, 2014, pages 731 - 733
GALLARDO, V. E.; VARSHNEY, G. K.; LEE, M.; BUPP, S.; XU, L.; SHINN, P.; CRAWFORD, N. P.; INGLESE, J.; BURGESS, S. M.: "Phenotype-driven chemical screening in zebrafish for compounds that inhibit collective cell migration identifies multiple pathways potentially involved in metastatic invasion", DIS. MODEL. MECH., vol. 8, 2015, pages 565 - 576, XP055392628, DOI: doi:10.1242/dmm.018689
XIAO, T.; ROESER, T.; STAUB, W.; BAIER H: "A GFP-based genetic screen reveals mutations that disrupt the architecture of the zebrafish retinotectal projection", DEVELOPMENT, vol. 132, 2005, pages 2955 - 2967
QIU, Z.; CANG, Y.; GOFF, S. P.: "c-Abl tyrosine kinase regulates cardiac growth and development", PROC. NATL, ACAD. SCI. USA, vol. 107, 2010, pages 1136 - 1141
CHISLOCK, E. M.; RING, C.; PENDERGAST, A. M.: "Abl kinases are required for vascular function, Tie2 expression, and angiopoietin-1-mediated survival", PROC. NATL. ACAD. SCI. USA, vol. 110, 2013, pages 12432 - 12437
Attorney, Agent or Firm:
CLYDE-WATSON, Zoe (120 HolbornLondon Greater, London, EC1N 2DY, GB)
Download PDF:
Claims:
CLAIMS

1. A compound of formula i, or a pharmaceutically acceptable salt or ester thereof,

wherein:

Ri is (CH2)mNRiiRi2;

R2 is selected from H, halo, OR 3l NHR13, alkyl, alkenyl and alkynyl;

R3 is selected from alkyl, alkenyl, alkynyl, aryl, haio, aryioxy, NHC02R4, NHCONR5R6,

NHCORy, NH-alkyl, NH-alkenyl, NH(CH2)n-aryl, (CH2)p-heteroaryl, (CH2)qC02R8,

(CH2)rCOR9 and NHSO2R 0, wherein each alkyl, alkenyl, aryl or heteroaryl moiety in the aforementioned list is optionally further substituted by one or more groups selected from alkyl, halo, OH, NH2, alkoxy, aryioxy, alkylamino, arylamino, carboxyl and carboxamide;

R4 to R10 and R13 are each independently selected from aikyl, alkenyl and aryi;

Rn and R12 are each independently selected from alkyl and alkenyl; or Rn and R12 are linked together with the nitrogen to which they are attached to form a heterocycioalkyi or heterocycioalkenyl group;

n, m, p, q and r are each independently selected from 0, 1 , 2, 3, 4, 5 and 6.

2. A compound according to claim 1 wherein and R12 are alkyl.

3. A compound according to claim 1 wherein Rn and R12 are linked together with the nitrogen to which they are attached to form a heterocycioalkyi group.

4. A compound according to any preceding claim wherein R is selected from NMe2, CH2NMe2, pyrrolidin-1-yi and piperidin-1 -yl.

5. A compound according to any preceding ciaim wherein. f¾ s selected from H and alkoxy, more preferably, H and OMe.

8. A compound according to any preceding claim wherein Ra is selected from aikyi, HCD2R , NHCOR7, HiQHaJri-aryl, MHCONR5R6i (GH2)p-heteroaryl and (OH2)qCO2R8,

7. A compound according to any preceding claim wherein R4 to t0 are each indepenefently aikyi,

8. A compound according to an preceding claim wherein ¾ is selected from Me, NHGOa-aikyl, NHCQ-alkyl, MHtGH ^-ar i, NHCGNH-aJkyS, (CH2)p»heteroaryl and

9. A compound according to an preceding claim wherein each of n, p, q and r is 1.

!O. A compound according to any preceding ciaim wherein R3 is selected from Me, MHGGy'Bu, NHCQCH2C(Me)3, NHCH2phenyi) NHCQHH~Bu, GHa-(4-methylOxa ol-2-

11. A compound according to any preceding claim wherein R3. is selected from Me and fsIHGQg-teu.

12. A compound which is selected from the following:

and pharmaceuticall accepiabie salts thereof.

13. A pharmaceutical composition comprising a compound according to any one of claims 1 to 12 and a pharmaceuticafly acceptable carrier, diluent or excipient.

14. A compound according to any one of claims 1 to 12 for use in medicine.

15. A compound according to any one of claims 1 to 12 for use in treating cancer.

16. A compound according to any one of claims 1 to 12 for use in treating a viral disorder.

17. A compound according to claim 16 wherein the viral disorder is selected from Epstein Barr Virus, Dengue Virus infection and HIV.

18. A compound according to any one of claims 1 to 12 for use in treating osteoporosis.

19. A compound according to any one of claims 1 to 12 for use in treating a neurological disorder.

20. A compound according to claim 9 wherein the neurological disorder is

Alzheimer's disease.

21. Use of a compound according to any one of claims 1 to 12 in the preparation of a medicament for treating or preventing a disorder selected from cancer, a neurological disorder, a viral disorder and osteoporosis.

22. A compound according to any one of claims 1 to 12 in the preparation of a medicament for the prevention or treatment of a disorder caused by, associated with or accompanied by abnormal Src kinase activity.

23. A method of treating a mammal having a disease state alleviated by the selective inhibition of a Src family kinase, wherein the method comprises administering to a mammal a therapeutically effective amount of a compound according to any one of claims 1 to 12.

24. A method according to claim 23 wherein the disease state is alleviated by the selective inhibition of a Src family kinase over Abi-ksnase.

25. Use of a compound according to any one of claims 1 to 12 in an assay for identifying further candidate compounds capable of selectively inhibiting a Src family kinase, preferably, selectively inhibiting: a Src family kinase over Abl-kirsase.

28. A process for preparing a compound of formula i as defined in claim T, said proce

(i) converting a compound of formula (ill) to a compound of formula (IV);

'(if) converting said CQmpound of formula (IV) to a compound of formula (V);

(iii) converting said compound of formula (V) to a compound of formula (VI); and

(iv) converting said compound of formula (VI) to a compound of formula (I),

27. A combination comprising a compound according to any on© Of c!aims 1 to 12 and a further therapeutic agent.

28. A pharmaceutical composition according to claim 13 which further comprises a second therapeutic agent.

29. A compound of formula (!l), or a pharmaceutically acceptable salt thereof,

wherein:

X is selected from amino, alkylamino, arylamino, hydroxy!, alkoxy and aryloxy;

L is an alkylene linker group having from 1 to 6 carbon atoms, wherein said alkylene linker group is optionally substituted by one or more R" groups;

Z is a piperidinyl or piperazinyl group that is optionally substituted by one or more groups selected from R" and {CH2)mNRiiRi2;

Y is an aryl or heteroaryi group, wherein said aryl or heteroaryi group is optionally substituted by one or more groups selected from halo, OR 3, afkyl, aryl, alkenyl, alkynyl, NHC02R4, NHCONR5Re, NHCOR7l NH-alkyi, NH-alkeny!, NH(CH2)n-aryl, (CH2)P- heteroaryl, (CH2)qC02R8, (CH2)rCOR9 and NHSO2Ri0, wherein each alkyl, alkenyl, aryl or heteroaryi moiety in the aforementioned list is optionally further substituted by one or more groups selected from alkyl, halo, OH, NH2, alkoxy, aryloxy, alkylamino, arylamino, carboxyl and carboxamide;

R' is selected from H, alkyl, aryl, heteroaryi and halo, wherein said alkyl, aryl and heteroaryi groups may be optionally substituted by one or more R" groups;

R4 to R 0 and R1 3 are each independently selected from alkyl, alkenyl and aryl; and n, m, p, q and r are each independently selected from 0, 1 , 2, 3, 4, 5 and 6;

each R" is independently selected from aikyl, OH, alkoxy and halo;

R and R 2 are each independently selected from alkyl and alkenyl; or Rn and R12 are linked together with the nitrogen to which they are attached to form a heterocycioalkyi or heterocycloalkenyl group;

for use in treating a disorder selected from Alzheimer's disease, Epstein Barr Virus and Dengue fever.

Description:
COMPOUNDS

The present invention relates to pyrazolopyrimidine compounds that are capable of inhibiting, more preferably, selectively inhibiting one or more tyrosine kinases. The compounds find applications in the treatment of a variety of disorders, including proliferative disorders such as cancer, bone, neurological, and viral disorders.

BACKGROUND TO THE INVENTION

Most drug discovery programs begin with a screening campaign (e.g. biochemicai, virtual or biophysical) for agonists, antagonists or inhibitors of a nominated target associated to a particular disease [1-4]. After hit identification, subsequent chemical optimization is fundamentally based upon "on-target" potency [1]. Generation of so- calied lead compounds (= high-affinity iigands) is followed by medicinal chemistry refinement into derivatives of superior potency and/or selectivity, and desirable pharmacokinetic properties (= druglikeness) [1 , 5]. Selected drug candidates are then validated in vivo and, upon verified safety and efficacy, progressed to human trials [5]. This well-defined process, which typically consumes over a decade of research work and tens of millions of pounds on its path to the clinic, is finding a particularly low success rate in the development of anticancer drugs [6], This is because, on top of the enormous difficulties of translating a drug discovery program from target identification - through preclinical and clinical development - into regulatory approval and marketing, it has become apparent that conventional approaches are not appropriately tailored to pathologies generated by the concurrent or sequential action of multiple etiologic factors such as cancer [6-8]. High attrition during late-stage drug development has underlined that elucidating cancer heterogeneity across patients and adaptive drug resistance mechanisms are the major obstacles to the development of effective targeted anticancer therapies [9-11]. These challenges are stimulating out-of-the-box thinking in pharmacotherapy research (e.g. targeted polypharmaco!ogy [10], antibody-drug conjugates [12], innovative prodrug approaches [13-17], etc.) and the reexamination of the core principles of drug discovery in oncology [18-20]. The rise of modern phenotypic drug discovery [18, 19] together with the use of clinically-relevant cancer models to guide early drug development [20], are representative examples of the paradigm shift initiated in the field to trigger a positive inflection point. Protein kinases are integral components of intracellular slgnai transduction cascades. They govern a wide range of basic cellular functions and coordinate cell-to-cell and extracellular matrix (ECiM)-tG-ceil communications to influence ce!i and tissue physiology. Thereby, kinases are directly involved in progressive diseases including cancer and inflammation [21]. Advances in the understanding of cancer cell biology along with the approval of several kinas inhibitors for cancer treatment have demonstrated the validity of a number of kinases a anticancer targets [22], while, on the contrary, other protein kinases have been shown to pla an essential role in tumor suppresso pathways (ahfi-targets) [23-26],

The vast majority of kinase inhibitors target the kinase ATP pocket and because all kinases (>500) necessarily possess this relatively well-conserved catalytic site, there is a great potential for cross-reactivity [10]. in fact, even if most clinically-approved kinase inhibitors have been developed from a single target hypothesis, they typically display a broad selectivity profile which, in some cases, have resulted in unanticipated clinical applications (e.g. sorafenih) [28]. An inhibitor's, promiscuity may also be advantageous for anticancer therapy when off-target activities assist to address bioactivit issues related to pathway redundancies , molecular heterogeneity o resistance mechanisms

[9, 10, 26], OR the contrary, if these activities result in the inhibition of anti-oncogenic pathways or lead to severe side effects, drug promiscuity becomes a major drawback. Paradoxically, there is strong evidence indicating that some kinases may behave as a target or an antifarget depending of the cancer context. By way of illustration, the expression of the activated fusion oncoprotein Bcr-Ahl is a genetic abnormality associated with chronic myeloid leukemia (C L) and Abi inhibitors (imatinib, dasatinib) are clinically used in chronic phase CIVIL treatment [27]. in addition, Abi family kinases are abnormally activated in various solid tumors, supporting their Involvement in oncogenesis [27], However, Abi (Abi1) and Arg (Ab|2) have been found to negatively modulate breast cancer progression in Vivo [28-30], indicating that Abi inhibition could be counterproductive for its treatment (5= breast cancer anti-target). This example serves to delineate the complexity of cancer etiology and highlights the necessity of developing kinase inhibitors with tailor-made pharmacodynamic profiles for the effectiv targeting of each cancer subtype, Unfortunately, despite the vast amount of small molecule inhibitors and biomedical: knowledge built over the years, the limited:

understanding of cancer biology prevents the appropriate targeting of orchestrated actions tha generate, maintain and progress most neoplastic processes, Acknowledging these limitations, many research groups are frontloading the search of robust empirical data to progress anticancer drug development programs away from classical black-and-white anticancer hypotheses. The present invention seeks to provide tyrosine kinase inhibitors having potent antiproliferative properties. The invention is founded on three hypotheses: (i) the use of phenotypic screening in designated models of cancer can be used to generate target- agnostic structure-bioactivity relationships and guide ligand optimization tailored to particular cancer types/subtypes; (ii) targeting the kinase ATP pocket with compounds derived from promiscuous kinase inhibitors can enable "rationally-biased" serendipitous discoveries; and (iii) early improvement of druglikeness on promiscuous ligands can be concurrently used to explore pharmacodynamic diversity. By means of this pragmatic approach to kinase inhibitor discovery, target deconvolution of identified hits and leads was largely facilitated, thereby enabling the rapid identification of the molecular targets and antitargets involved in the observed phenotype.

STATEMENT OF INVENTION

A first aspect of the invention relates to a compound of formula (I), or pharmaceutically acceptable salt thereof,

R 2 is selected from H, halo, OR 13 , NHR 3 , a!kyl, alkenyl and alkynyl;

R 3 is selected from alkyl, alkenyl, alkynyl, aryl, halo, aryloxy, NHCO 2 R 4 , NHCONR 5 R 6 , NHCOR 7> NH-alkyl, NH-alkenyl, NH(CH 2 ) n -aryl, (CH 2 ) p -heteroaryl, (CH 2 ) q C0 2 R 8 , (CH 2 ) r CORg and NHSO 2 R 10 , wherein each alkyl, alkenyl, aryl or heteroaryl moiety in the aforementioned list is optionally further substituted by one or more g ouses selected from alkyi, halo, OH, NH ¾ , aikoxy, aryloxy, alkylamiHO, arylamino, earboxyS and carboxamide; R 4 to f¾io and R a e each independently selected from alkyi, alkenyj and aryl;

R ri and f½ are each independently selected from alkyi and alkenyl; or ' R t1 and R 12 are linked together with the nitrogen to which they are attached to form a heterocycloalkyl or heterocyeioalkenyl group;

ft, m t p, q and are each independently selected fro 0, 1 , 2, 3, 4, 5 and 6.

A second aspect of the invention relates to a pharrnaoeutica! composition comprising at least one compound as described above and a pharmaceutica!iy acceptable carrier, diluent or exoiplent

A third aspect of the invention relates to a compound as described above for use in medicine.

A fourth aspect of the invention relates to a compound as described above for use in treating a disorder selected from a proliferative disorder and a viral disorder,

A fifth aspect of the invention relates to the use of a compound as described above in the preparation of a medicament for treating or preventing a disorder selected from proliferative .disorder, osteoporosis, Alzheimer's and Parkinson's disease and a viral disorder.

A seventh aspect of the invention relates to a method of treating a mammal having a disease state alleviated by inhibition, or preferably the selective inhibition, of a tyrosine kinase, wherein the method comprises administering to a mammal a therapeutically effective amount of a compound as described above.

An eighth aspect of the invention relates to the use of a compound as described above in an assay for identifying further candidate compounds capable of inhibiting, or selectively inhibiting, a tyrosine kinase.

A ninth aspect of the invention relates to a combination comprising a compound as described above and a second therapeutic agent. A tenth aspect of the invention relates to a process for preparing compounds as described herein.

An eleventh aspect of the invention relates to a compound of formula (II), or a pharmaceutically acceptable

wherein:

X is selected from amino, alkylamino, arylamino, hydroxy!, alkoxy and aryloxy;

L is an alkylene linker group having from 1 to 6 carbon atoms, wherein said alkylene linker group is optionally substituted by one or more R" groups;

Z is a piperidinyl or piperazinyl group that is optionally substituted by one or more groups selected from R" and (CH 2 ) m NR 11 R i2 ;

Y is an aryl or heteroaryl group, wherein said aryl or heteroaryl group is optionally substituted by one or more groups selected from halo, OR 3 , alkyl, aryl, alkenyl, alkynyl, NHC0 2 R 4[ NHCONR 6 R 6 , NHCOR 7 , NH-alkyl, NH-alkenyl, NH(CH 2 ) n -aryl, (CH 2 ) P - heteroaryl, (CH 2 ) q C0 2 R 8 , (CH 2 ) r COR 9 and NHSO 2 R 0 , wherein each alkyl, alkenyl, aryl or heteroaryl moiety in the aforementioned list is optionally further substituted by one or more groups selected from alkyl, halo, OH, NH 2 , alkoxy, aryloxy, alkylamino, arylamino, carboxyl and carboxamide;

R' is selected from H, alkyl, aryl, heteroaryl and halo, wherein said alkyl, aryl and heteroaryl groups may be optionally substituted by one or more R" groups;

R 4 to R 10 and R 13 are each independently selected from alkyl, alkenyl and aryl; and n, m, p, q and r are each independently selected from 0, 1 , 2, 3, 4, 5 and 6;

each R" is independently selected from alkyl, OH, alkoxy and halo;

R-H and R 12 are each independently selected from alkyl and alkenyl; or Rn and R 2 are linked together with the nitrogen to which they are attached to form a heterocycloalkyi or heterocyc!oalkenyl group;

for use in treating a disorder selected from Epstein Barr Virus, Alzheimer's disease and Dengue fever. DETA!LED DESCRIPTION

The present invention relates to pyrazolopyrimidine compounds that are capable of inhibiting, or more preferably selectively inhibiting, one or more kinases, preferably one or more tyrosine kinases, even more preferabiy Src-kmase. in particular, the nvention relates to substituted pyrazolopyrimidi e compounds having a specific substitution pattern.

"Alky!" is defined herein as a straight-chain or branched alkyi radical, for example, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyi, pentyl, hexy!. Preferabiy, the aikyl group is a C ; 12 ~alkyi group, more preferably, a C^s-alkyl group, even more preferably a C M -alkyi group.

"Aikenyi" is defined herein as a straight-chain or branched radical, containing one or more oarbon-carbon double bonds, Preferabiy, the aikenyi group is a

more preferably, a C ajky! group, even more preferably a C 2 - -afe l group.

"AlkynyS" is defined herein as a straight-chain or branched radical, containing one or more carbon-carbon triple bonds. Preferably, the alkynyl group is a i¾.i2-alk j group, more preferably, a C e-aikynyl group, even more preferably a Ca- aik n l group. "Cycloalkyl" is defined herein as a monocyclic a!kyl ring, such as, cyclopropyi, cyclobutyl, cyclopentyl, cyclohexyl or cyeloneptyi, or a fused bicyclic ring system such as norbornane. more

preferably a Cs-s-c cloalk i group, ''Cycloalkenyl" is defined herein as a cyclic grou as defined above for cyoSoaikyS, but containing one or more carbon-carbon double bonds. Preferably, the cycloalkenyl group is a G 3 .B-cyoloaikenyi group, more preferably a Qs-e-cyc!oalkeny! group.

"Halogen" is defined herein as chloro, fluoro, bromo or iodo,

As used herein, the term "aryf refers to a G e . z aromatic group, which may be benzocandensed, for example, phenyl or naphthyl.

"Heteroaryl" is defined herei as a monocyclic or bicyclic C 2 .n aromatic ring comprising one or more heteroatoms (that may be the same or different), such as oxygen, nitrogen or sulphur. Examples of suitable heteroaryl groups include thienyl, furanyl, pyrrolyl, pyridinyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazoiyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl etc. and benzo derivatives thereof, such as benzofuranyl, benzothienyl, benzimidazoiyi, indolyl, isoindolyf, indazo!yl etc.; or pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl etc. and benzo derivatives thereof, such as quinolinyi, isoquinolinyl, cinnoiinyl, phthalazinyl, qutnazolinyl, quinoxalinyl, naphthyridinyl etc.

In one preferred embodiment n, m, p, q and r are each independently selected from 0, 1 , 2 and 3.

In one preferred embodiment, R-n and R 2 are alkyl. in one preferred embodiment, m is 0. In another preferred embodiment, m is 1.

In another preferred embodiment, R-n and R 12 are linked together with the nitrogen to which they are attached to form a heterocycloalkyi group. Preferably, for this embodiment, m is 0.

Preferably, R and R 12 are linked together with the nitrogen to which they are attached to form a 6-membered heterocycloalkyi group or a 5-membered heterocycloalkyi group.

In a more preferred embodiment, Ri is selected from NMe 2 , CH 2 NMe 2 , pyrrolidin-1-yl and piperidin-1-yl.

In one preferred embodiment, R 2 is selected from H and alkoxy, more preferably, H and OMe. In one preferred embodiment, R 3 is selected from alkyl, NHC0 2 R 4 , NHCOR 7 , NH(CH 2 ) n - aryl, NHCONR 5 R 6 , (CH 2 ) p -heteroaryl and (CH 2 ) q C0 2 R 8 .

In one preferred embodiment, R 4 to R 10 are each independently alkyl. In one preferred embodiment, f¾ is selecied from Me, NHG0 2 -a!kyi, ' NHCO-aik !, NH(CHs)a-aryl, NHGQNH-a!kyl, (CHa) p -heteroar i and (CH 2 ) q CO alkyl.

!n one preferred embodiment, each of n, p, q and r is 1.

In one preferred embodiment, f¾ is selected from Me, HCQ 2 - t Bu, NHCGCH 2 C ' (M ' e) ¾ HGHa iienyl, NHCOMH- Bu, CH 2 -{4-metf yl-oxazol-2-yi) and GBsCO^Bu. in one preferred embodiment 3 is selected from Me and NHCOj-'Bu.

In one preferred embodiment, Ra ¾ Me and 2 is H.

I one preferred embodiment, F¾ is aikoxy, and j is selected from NHGG2R4,

NHCOR 7. , NH{CH 2 )n-aryl, NHCONR s 8! iCH 2 )p-heteroaryi and (CH 2 )<,CQ ? R 8 . in one preferred embodiment; R 2 is OMe and R 3 is selected from NHCCy'Bu,

HGGCH 2 C(fySe) 3 , NHCHzphenyl, NHCO H-feu. CH (4~mefhyl-o)fa2oi-2-yi) and

In one highly preferred embodiment of the invention the compound of formula seieoted from the following:

and pSiarmaceutica!iy acceptable saiis thereof. In one highly preferred embodiment, the compound is selected from compounds 506, 518, 519, 533, 553 and 565.

In another highly preferred embodiment, the compound is selected from compounds 506 and 565.

Compounds of Formula (II)

Another aspect of the invention relates to a compound of formula (II), or a

pharmaceutically acceptable

wherein:

X is selected from amino, alkylamino, arylamino, hydroxyl, alkoxy and aryloxy;

L is an alkylene linker group having from 1 to 6 carbon atoms, wherein said alkylene linker group is optionally substituted by one or more R" groups;

Z is a piperidinyl or piperazinyl group that is optionally substituted by one or more groups selected from R" and R-iz;

Y is an aryl or heteroaryl group, wherein said aryl or heteroaryl group is optionally substituted by one or more groups selected from halo, ORi 3 , a!kyl, aryl, alkenyl, a!kynyl, NHC0 2 R 4 , NHCONR 5 Re, NHCOR 7 , NH-alkyl, NH-alkenyl, NH(CH 2 ) n -aryl, (CH 2 ) P - heteroaryl, (CH 2 ) q C0 2 R3, (CH 2 ) r COR 9 and NHSO 2 Ri 0 , wherein each alkyl, alkenyl, aryl or heteroaryl moiety in the aforementioned list is optionally further substituted by one or more groups selected from alkyl, halo, OH, N H 2 , alkoxy, aryloxy, alkylamino, arylamino, carboxyl and carboxamide;

R" is selected from H, alkyl, aryl, heteroaryl and halo, wherein said alkyl, aryl and heteroaryl groups may be optionally substituted by one or more R" groups;

R 4 to R 0 and R 13 are each independently selected from alkyl, alkenyl and aryl; and n, m, p, q and r are each independently selected from 0, , 2, 3, 4, 5 and 6;

each R" is independently selected from alkyl, OH , alkoxy and halo; Rn and R 12 are each independently selected from alky! and alkenyl; of Rn and R, 2 are linked together with the nitrogen to which they are attached to form a heterocycloalkyi or heteFQcycioalkany! group;

for use in treating a disorder selected from Epstein Barr Virus, Alzheimer's disease and Dengue fever. in one preferred embodiment, L is a Cg-C alkylen linker, which may be substituted or unsubstituted. In a more preferred embodiment, L is an ethylene linker, which may be substituted or unsubstituted. in one preferred embodiment, X is selected from amino, alkyiamino and aryiamino. More preferably, X is amino.

In one preferred embodiment, R' is selected from H and alkyl, or preferably, R ! is H,

In one preferred embodiment, 2 is a piperidin-1-yi or piperazin~1~yi group that is optionaliy substituted by one or more groups selected from R" and {CH2) m R: S1 t2 . in a more preferred embodiment, 2 is a piperidin-1-yi group that is optionall substituted by one or more groups selected from R" and (CHj mNR ' M ' Riz- in a more preferred embodiment, Z is a piperidin-1-yl group that is substituted by one or more (CH ¾ } rf ,N n -)a groups, . Mare preferably, 2 is a piperldin-l-yl group that is substituted in the 4~position by a group seieoted from dialkylamino, eyGloalfcylamino and dialkylaminoa!kyl in one preferred embodiment, Y is an optionaliy substituted aryl group, more preferably, an optionali substituted phenyl group. in a preferred embodiment, Y is a phenyl group optionally substituted in the 3-posstion with a group selected from halo, alkoxy, arylpxy, aikyfamino and alkyl. In a preferred embodiment, Y is a phenyl group optionally substituted in the 4-position with a group selected from alkyl, aryl, halo, aryloxy, alkylamino, arylamino, NHCO 2 R 4 , NHCONRsRe, NHCOR 7l NH-alkyl, NH(CH 2 ) n -aryl, (CH 2 ) p -heteroaryl, (CH 2 ) q C0 2 R 8 , (CH 2 ) r COR 9 and NHSO 2 R 0 , wherein each instance of alkyl, aryl or heteroaryl in the aforementioned list is optionally further substituted by one or more groups selected from alkyl, halo, OH and NH 2 .

More preferably still, Y is a phenyl group substituted in the 3-position with a group selected from halo, alkoxy, aryloxy, alkylamino and alkyl, and substituted in the 4- position by a group selected from alkyl, aryl, halo, aryloxy, alkylamino, arylamino, NHC0 2 R 4 , NHCONRsRe, NHCOR 7 , NH-alkyl, NH(CH 2 ) n -aryl, <CH 2 ) p -heteroaryl, (CH 2 ) q C0 2 R 8l (CH 2 ) r COR 9 and NHSO 2 R 10 , wherein each instance of alkyl, aryl or heteroaryl in the aforementioned list is optionally further substituted by one or more groups selected from alkyl, halo, OH and NH 2 .

Highly preferred embodiments are as described above for compounds of formula (I).

Design, synthesis and screening of novel pyrazolopyrimidines

In the search for inhibitors that could potentially target a wide range of kinases with relevance in cancer, a ligand-based drug development program was conducted using the multikinase inhibitor PP1 as the core structure (Figure 1). PP1 indiscriminately targets tyrosine protein kinases, many of which are involved in oncogenesis such as Src family kinases (SFK), Ret, Kit and Abl [31-34], Moreover, related derivatives developed thereafter [35, 36] have shown strong inhibition against other groups of kinases with relevance in cancer including VEGFRs, PDGFRs, PI3Ks and mTOR. According to the co-crystal structure of PP1 with Hck [37] and Ret [38] kinases, this small molecule is an archetypical type I kinase inhibitor [39, 40], with its Λ/5 and 4-NH 2 groups forming multiple hydrogen bonds with the hinge region of the kinase catalytic site [39-41]. The C3 p-tolyl group is located in a hydrophobic region well-conserved across most tyrosine kinases, thus being responsible for the partial selectivity of the inhibitor over other families of kinases. Although PP1's potent inhibition of disease-associated kinases (e.g. SFK, Ret, etc.) make it a valuable tool for biological studies, its clinical use is limited by very low solubility in water and poor selectivity, major limiting factors for the clinical translation of many drug candidates [42]. Th© substitution of PP1's tert-buty! group at the A/1 position with flexible water- soiubjlfeing groups can be used to both improve its drug-like properties and explore the accessible sugar/phosphate regions occupied by the natural ligand ATP (Figure 1) in the search for novel binding affinity profiles. As show in the lower panel of Figure 1, compounds were designed to display a cyclic tertiary amine connected to .the. A 1 position of the pyrazoiopyrimidine ring through an ethylene linker (see general synthesis in Figure 2). Following this synthetic procedure, structural diversity was readily implemented by coupling a selection of cyclic secondary amines to the corresponding aldehyde derivative via reductive amSnation,

Several investigations have reported that substitution of the p-tolyl group at the C3 position of PP1 by substituted aryi moieties (eve closely related ones) significantly impac on protein-Sigand binding, Medicinal chemistry campaigns on that position has generated inhibitors for a wide range of kinases, including receptor and non-receptor tyrosine kinases (e.g. c-Src, Ret, PDGFRs, VECaFRs, c- . it and Abl) [31-35] and non- tyrosin kinases (e.g. P!S s and rh ' TOR) [38, 43], To expand the prospective pharmacodynamic scope of the novei compounds, alongside the modifications impiemented On the N3 position, a broad selection of aryi bo ' rohic acids were employed to funotionalize the G3 position of the pyrazo!opyrimidine ring by palladium -catalyzed cross-coupling chemistry.

The human breast adenocarcinoma ceil line MCF1 was selected as a cell-based model and phenotypic screening used to evaluate the novel compounds and classify hits against different aniioncDgenic activities, Highly-discriminate cell-based assays do not only allow the identification of compounds that target proteins involved in MCF7 cell growth, migration and survival but also exclude chemicals with low ceil penetrability (and therefore deficient drug-likeness). Searching for antiproliferative properties as the primar o tput; EG S0 values in MCF7 cells were calculated for the novel compounds using a 10-point half-log dose response stud (100 μΜ to 0.01 μΜ). Cell viability was determined at day 5 using PrestoBlue® reagent, Spectrofluorometry analyses were carried out in a Envision© plate reader.

Among all the structures synthesized and tested (see Tables of activities 1 and 2), the introduction of a 4-(W-Boc-am!no)-3~metho)iyphenyl at the C3 position was one of the most successful (Figure 3a), On the other hand the presence of dimef hy!amino- containing piperidinyl groups at the position N1 were found optimai for the

antiproliferative potency of the compounds, with the dimethyfamino-piperidinylethyl group being the best moiety found. Antiproliferative properties of compounds 503 and 506 were tested in cells using dasatinib as positive control. Dasatinib is a potent inhibitor of both Abl and Src, is currently used in the treatment of chronic phase CML and is in several clinical trials for different types of cancer including breast cancer. Together with MCF7 cells, triple negative breast cancer MDA-MB-231 cells were tested alongside to explore the antiproliferative potency of the compounds against different breast cancer subtypes. As shown in Figure 3b, compound 506 displayed a superior antiproliferative effect to compound 503 over both MCF7 and MDA-MB-231 breast cancer ceils. Remarkably, compound 506 also outperformed the gold-standard Src inhibitor dasatinib in both cell lines.

To identify the target s that could be responsible of the phenotype induced by lead compound 506 in MCF7 cells, IC 50 values were determined against a selection of human protein kinases involved in cancer. The recombinant proteins of the assay were chosen in accordance with the kinase profile of related pyrazoiopyrimidines found on the literature [31-36]. As shown in Table 3 (note that values are expressed in nM), both compounds 503 and 506 possessed sub-nano molar potency against Src and Yes (proto-oncogenes that play a critical role in signal transduction pathways involved in tumor growth, angiogenesis, invasion, and dissemination) [30] with compound 506 having an improved selectivity over Abl (>950 fold difference in potency).

Pharmacologically speaking this is very noteworthy since evidence in the literature strongly suggests that Abl inhibition could be counterproductive for the treatment of some types of breast cancer [28-30] and Abl inhibition has also been associated with cardiotoxicfty [44]. Importantly, this unique selectivity profile differentiates the compounds of the invention from compound PP20 (Figure 3a) [35], which is a known unspecific Abl/Src kinase inhibitor displaying subnanomoiar potency for both Abl and Src. Without wishing to be bound by theory, introduction of the polyamine moiety at Λ/1 position of the pyrazoiopyrimidines is believed to play a part in the unprecedented selectivity profile of these derivatives.

To further assess the selectivity profile shown by compounds 506 and 503 on a noncancerous cell modei, dose response studies were carried out in murine SYF fibroblasts (which cells deficient for Src, Yes, and Fyn) with compound 506, 503 and dasatinib. Cell viability determined at day 5 using PrestoB!ue® reagent and analyzed by

spectrGfiUGrometry (Figure 3c), Remarkably, compounds 506 and 503 showed much lower antiproliferative activity than dasatinifo in ' S ' YF ceils, Indicating that the novel compounds are more selective than dasatinib and might lead to reduced side effect caused by off-target activities.

Modifications of compound 506 led to a number of derivatives with similar properties (Table 1} and many compounds with Sow selectivity and/or antiproliferative activity {Table s},

Highly Selective Src Kinase Inhibitors

Src is a non-receptor tyrosine kinase and the most widely studied member of the Src family kinases (SFKs), which include ' Lyn, Fyn, Lck, Hck, Pgr, Blk, Frk and Yes, Upregiilated Src expression and/or activity has been reported in many tumour types, but is best described in colon and breas cancer where Src activity correlates with malignancy potential and poor clinical prognosis [47, 49],

Acquired drug resistance to numerous anticancer agents including cetuximab, oxatipiatin, gemcitabine, rapamycin, taxanes and 8-RAF inhibitors has been associated with dysreguiated Src signaling [65, 57, 54, 48, 56, SO]. These studies indicate that Src activity represent a common drug resistance mechanism and targeted inhibition of Src may sensitize resistant tumours to a number of therapeutic interventions or provide an alternate treatment -option, in late stage relapsed disease. For example, Src activity has also been identified as a common signalling mechanism in trastuzumab resistance, indicating that Src inhibitors may provide an alternative treatment for trastuzumab- resistant breast tumours 661.

The strong disease linkage and correlation between Src activity with poor cancer prognosis, morbidity and acquired resistance to existing therapy all support the premise that Src represents an important anti-cancer therapeutic target. Several Src inhibitors (Dasatanib, Bos-utinib, Saracatinib, AZD0424) have entered the clinic and a number of Phase l/l I trials are underwa across distinct cancer indications [58, 49}. However, clinical outcome data published to date has not been compelling and the unresolved challenge is identifying which patients are most likely to benefit from targeted Src inhibition and which are the most appropriate clinical settings to demonstrate efficacy, e.g advanced disease endpoints or combination with existing agents or rationally designed novel combination therapies.

All current Src kinase inhibitors (Dasatanib, Bosutinib, Saracatinib, AZD0424) represent non-selective tyrosine kinase inhibitors with significant off-target activities contributing to adverse events and dose-!imiting toxicities. Ail existing Src inhibitors also demonstrate potent activity against Abl1 (Abelson murine leukemia viral oncogene homolog 1) a cytoplasmic and nuclear protein tyrosine kinase. The t(9;22) translocation results in the head-to-tail fusion of the BCR and ABL1 genes, leading to a constitutively-active fusion gene present in many cases of chronic myelogenous leukemia, which are successfully treated with BCR-Abl1 inhibitors.

The off-target activities of known Src inhibitors were neither selected or optimized by rational design and may limit their use in advanced cancer disease setting as combination therapy, where unnecessary off-target activity contributes to dose-limiting and combination-limiting toxicity. Evidence exists indicating that therapeutic targeting of Abl may have detrimental effects on cardiotoxicity [51 , 56] and non-tumourigenic tissue through its dual function as a tumour suppressor, thus potentially contributing to neoplasia [64, 62, 45, 55]. Furthermore, evidence suggests that targeting Abl contributes to osteopenia [60], Thus, current non-selective dual Src/Abl inhibitors are not appropriate for long-term application in both cancer and non-cancer (e.g.

osteoporosis or virus infection) indications.

A phenotypic screening strategy was employed in order to identify and optimise novel and highly potent c-Src kinase inhibitors with improved target selectivity profiles compared with existing Src inhibitors on the market. Phenotypic screening strategy favours the discovery of small molecules with optimal biophysical properties. Using this method, pyrazolopyrimidines with potent antiproliferative activities were found.

Unexpectedly, after target identification, it was demonstrated that the presently claimed compounds (including, e.g. compounds 506, 518, 519, 533, 553 and 565) are the only highly potent c-Src kinase inhibitors reported to date that do not target Abl but exert anti-proliferative, anti-migratory and cytotoxic properties upon cancer cells in vitro with comparable potency to existing non-selective Src inhibitors (Tables 4 and 5). I summary, all of the Src kinase inhibitors under current clinical development are not appropriately tailored to the optimal . ' Use of a Src inhibitor as drug combination therapy in advanced disease settings or in the chronic treatment of non-cancerous disease indications as a result of significant off-target activity, including adverse inhibition of AbH . The presently claimed compounds are the first selective Src kinase inhibitors reported that do not inhibit Abil . For example, compounds 506 and 565, which are highly preferred, are superior to existing Src inhibitors for use in the advanced cancer disease setting as a combination therapy, As shown In Table 5 a d Figure 4, the novel Src in ibitors are specific for the "Src family" kinases over other members of the broader kinome family. Th Src family is composed of several closely related homologues (e.g. Lyrs, Fyn. Lck, Hck, Fgr, Blk, Frk and Yes). The homplogues are highly conserved arid it would be extremely difficult to Selectively target distinct family members by small-molecules. In contrast to ail of the Src inhibitors published to date, and in the clinic, the presently claimed compounds are the only molecules that specifically target Src family members at low/sub-nanomojar potenc and not other kinases including Abl. Full kinome panel screening data for the compound 506 supports this claim (see Figure 4). Analysis of the solubility properties found that the presently claimed compounds possess good solubility in PBS (>1QQmg/rrsL), in addition, they show good stability in the presence of liver microsomes and plasma binding at the level of 81-91% (see Table 6), hERG channel inhibition was studied for selected compounds of the invention. A Life Technologies fluorescence-based kit (Predictor® hERG Fluorescence Polarization Assay Kit) was used. All of the compounds showed iG S0 ^IO μΜ, The most potent of the series was compound 533 with iC 60 (1-0-1 S \M). A second hERG Safety Assay utilizing the lonworks™ high -through put eiecfrcphysioiogy screen for selected compounds of the invention was also performed, along with plasma stability analysis (mouse, rat and human). The results are shown in Table 7. These experiments corroborated the safety of the compounds. Plasma stability studies demonstrate the high stability of the compounds in human plasma. In vivo PK (iv and po) were subsequently studied in female mice for compounds 506 and 565. Blood and plasma levels were studied for 8 hours and the compounds showed no toxic effects. Oral administration demonstrated that the compounds are orally available. While 506 displayed moderate oral bioavailability, 565 showed excellent oral bioavailability (52%). The half-life of compound 506 was superior than for compound 565.

As observed in Table 8, CYP P450 inhibition was shown to be residual. Given the potency of the compounds, anything less than 50% inhibition at concentrations above 10μΜ is not considered to be significant.

PD studies were done in vitro with MDA-MB-231 cells, which showed strong inhibition of phospho-Src at low nanoM concentration (see Figure 5 below). To test whether inhibition of Abl could influence the phenotypic effect mediated by the compounds, combination studies were performed with compound 506 and the specific Abl allosteric inhibitor GNF-2. As shown in Figure 6, titration of the Abl inhibitor antagonizes activity of the compound on cancer cell viability in vitro. A lack of clinical efficacy is observed with existing Src inhibitors when used as monotherapy in cancer, thus for some time it has been argued that Src inhibitors must be used in combination therapy. It is believed that the absence of detrimental off-target activity including Abi activity (unique to the presently claimed compounds) will enhance both efficacy and minimize adverse events resulting from off-target activity. Src family kinases are well known to have active roles in cell migration and invasion and are involved metastasis in cancer patients [67, 68]. To test whether compound 506 could inhibit cell migration in vitro, a scratch wound assay was set up using the Incucyte Zoom system. Cells were imaged across 30 minute intervals over 24 hours. The migration of MDA-MB-231 ceils was inhibited as early as 6 hours into the study and potency retained until the end of the assay, 24 hours (see Figure 7). Compound 506 showed comparable results to Dasatinib.

As a toxicity study and phenotypic in vivo study, a zebrafish tail regeneration assay was performed [69]. As demonstrated by Yoo and coworkers [69], the action of Src family kinases (in particular Fynb, similar to human Fyn) is required for regeneration of amputated fins. This was used to assess and compare the in vim activity of compound 508 and Dasatinib. As shown in Figure 8 : compound 506 and dasatinib inhibited regeneration of the fail fin. However, compound compound 506 did not show any other phenotypic or toxic effect on the embryo, while Dasatinib clearly induced cardiofoxicity (a weil-established dose-limiting side effect of Abl inhibitors including Dasatinib).

Phenotypic screening of SRC inhibitor 506 in zebrafish

Developing zebrafish provides a rapid phenotypic assay to simultaneously test safety and efficacy of novel compounds in a living vertebrate [783- Small molecule phenotypic based screens in zebrafish have recently implicated SRC kinase in the migration of the posterior lateral line primofdium [78], a cohesive cluster of ceils that migrates horizontally under the skin along the myoseptum to the end of the tail, periodically depositing neuromasts. To determine the effects of 508 On cell migration in vivo,, we treated Tg(hni3G: QFP) transgenic zebrafish [80] that express green fluorescent protein (GFP) in the mechanGsensory hair cells of the iateral line (which form part of the neuromasts) with 508 and dasatinib for 2d and measured the distance of the last neuromast to the tip of the tail (marked by the end of the notochord and the presence of black melanocytes, Fig 9a, vertical dashed line), 506 significantly reduced neuroblast migration (> 100 microns in average) with minimal effect on the development of the embryos (Fig 9a-e). In contrast, dasatinib treatment at >10 μΐνΐ resulted in severe cardiotoxicit and death of most embryos. At concentrations that wer compatible with embryo survival (1 -10 μΜ , dasatinib did not inhibit the migration , of neuroblasts whereas it did still induce a patent card iofoxic phenotype {note heart enlargement in Fig 9c). Further safety studies showed that dual ABL/SRC inhibitor PP20 also induces severe cardiotoxicity in zebrafish even after short treatment. These results, which correlates with the essential role of ABL in heart development and healing 81|, [82]. suggests that the selectivity of 506 over ABL might be ad vantageous for therapy when ABL inhibition is not required.

In vivo SRC Inhibition study In a tumor xenograft model

The presence of active (phosphoryiated) SRC in human colorectal cancer HCT116 cells and its inhibition under 506 treatment was verified by western blot. Subsequently, an in vivo PD study was performed in a xenograft mode! of HCT1 6 cells in mice [83].

HCT118 cells were injected subcutaneousf and tumors allowed to grow u to 3-4 mm in diameter. Subsequently, mice were dosed daily for 3 d with 506 (50 mg/kg, in nanopure water) or vehicle (nanopure water) by oral gavage and culled 3 h after the last dose (n=4). Tumors were excised, fixed and sections labelled for phospho-SRC Y416 and stained with hematoxylin. As shown in Figure 10, microscopy analysis demonstrated significant reduction of phospho-SRC Y416 in the xenograft sections from mice treated with 506 relative to the untreated anima! controls.

THERAPEUTIC APPLICATIONS

A further aspect of the invention relates to a compound as described above for use in medicine.

Another aspect of the invention relates to a compound as described above for use in treating a disorder selected from a proliferative disorder, a viral disorder, neurological disorders and osteoporosis.

In one preferred embodiment, the proliferative disorder is cancer. Preferably, the cancer is selected from solid cancers at any stage. Preferably, the cancer is in a late-stage, with metastatic lesions. Preferably, the primary cancer is selected from breast cancer, colon cancer, prostate melanoma, bladder, pancreatic, head and neck and ovarian cancer, with or without metastasis, and haemato!ogical cancers such as acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), acute lymphocytic leukemia (ALL), multiple myeloma (MM) and non-Hodgkins lymphoma.

Particularly preferred primary cancer disease indications include, but are not limited to: triple negative breast cancer, Trastuzumab and/or Lapatinib resistant Her2 positive breast cancer, advanced melanoma including Vemurafenib resistant disease and multiple myeloma.

In one preferred embodiment, the proliferative disorder is Lymphangioleiomyomatosis (LAM), a progressive lung disease in which atypical ceils, originating somewhere in the body, spread throughout the lungs, gradually blocking small airways and producing cysts. Typically, the disease progresses slowly, but eventually it can restrict breathing to cause death. Currently there is no proven cure for LAM. Src kinase is active in LAM cells and is important for eel! growth and cells' ability to move around and invade the !ung tissue [70]. Clinical trials to investigate the tolerahiiity of saracatinib in Lam patients is currently in progress. In one preferred embodiment, the proliferative disorder Is atherosclerosis and restenosis characterized by migration and hyperproiiferaiioh of vascular smooth muscle cells. Src Kinase signalling has previously been implicated in aortic smooth muscle ceil proliferation induced by clinical risk factors detected in the circulation; G-peptid© and glycated LDL [71 , 72J.

The compounds described here are also suitable for chronic administration in several non cancer disease indication such as osteoporosis, Parkinson's disease, Alzheimer's disease and dengue virus infection. in one preferred embodiment, the disorder is osteoporosis or bone metastasis. Src activity is implicated in metastatic bone disease, which represents the primar metastatic site for patients with breast cancer. The structural integrity and normal functions of the bone are governed by the carefully controlled balance of bone resorption mediated by osteoclast cells and bone production mediated by osteoblasts. Src activit plays a key role in bot osteoclast function and tumour colonization of the bone [81], Thus, targeted inhibition of Src activity can induce net bone formation [46] and alleviate the significant morbidity associated with metastatic bone disease and potentially osteoporosis. In one particularly preferred embodiment the viral disorder is Epstein Barr Virus. The Epstetn-Barr virus (EBV), also called human herpesvirus 4 (HHV~4), is one of eight viruses in the herpes family, and is one of the most common viruses In humans. It is best known as the cause of infectious mononucleosis (glandular fever). It is also associated with particular forms of cancer, such as Hodgkin's lymphoma, Burkstt's lymphoma, nasopharyngeal carcinoma, and conditions associated with human immunodeficiency virus (HIV), such as hairy leukoplakia and central nervous system lymphomas. There is evidence that infection with E8V is associated with a higher risk of certain autoimmune diseases, especially dermatomyosifis, systemic lupus erythematosus, rheumatoid arthritis, Sjogren's syndrome and multiple sclerosis. Some 200,000 cancer cases per year are thought to be attributable to EBV. EBV infects B cells of the immune system and epithelial cells. Once EBV's initial lytic infection is brought under control, EBV latently persists in the individual's B cells for the rest of the individual's life. In one particularly preferred embodiment, the viral disorder is Dengue Virus infection. Dengue fever cases have increased dramatically over the last 4 decades with between 50 and 528 million people infected yearly. Symptoms include, sudden high fever, severe headaches, pain behind the eyes, severe joint and muscle pain, nausea, vomiting, skin rash, mild bleeding (such as nose bleed, bleeding gums, or easy bruising).

Approximately, 5% of people suffer more severe and life threatening symptoms (e.g. dengue shock syndrome and dengue hemorrhagic fever). Dengue fever is directly related to infection by the dengue virus, a mosquito-borne single positive-stranded RNA virus. Developing a vaccine against the disease is compounded by five distinct serotypes of the dengue virus that can cause the disease, thus, any vaccine must immunize against all five types to be effective. To date no vaccines or curative treatments have been developed. A recent study published in the Journal of Virology identified the Src family member, Fyn Kinase as an important host-cell target required for RNA replication of the Dengue Virus in cells [53]. Treatment of cells infected with dengue virus with small molecule Src inhibitors, AZD0530 and Dasatanib inhibited viral replication, however, serial passaging of dengue virus in the presence of Dasatinib led to the identification of a mutation in the transmembrane domain 3 of the NS4B protein that overcomes the inhibition of RNA replication by AZD0530 and Dasatinib. Thus, similar to cancer ceils, viruses can rapidly acquire resistance to targeted therapy and, thus, combination therapies represent the standard of care. In contrast to other Src inhibitors, compound 518 selectivity is tightly restricted to Src family members of the kinome including Fyn (Table 5) and, thus, may represent an ideal component of combination therapy for dengue fever or other virus infections where Src family members are implicated e.g. Epstein Barr Virus and HIV1 [63, 59].

In one particularly preferred embodiment, the viral disorder is HIV1.

In one preferred embodiment, the neurological disorder is Alzheimers disease. Recent studies have demonstrated the involvement of Fyn, a member of the Src family, in signaling pathways that lead to severe brain pathologies, such as Alzheimer's and Parkinson's diseases [73, 74]. Fyn plays a role in the regulation of amyloid-β (Αβ) plaques production and mediates Αβ-induced synaptic deficits and neurotoxicity. Fyn also induces tyrosine phosphorylation of tau [75j. A phase lb study of saracatinsb (AZD05305, a small molecule inhibitor with high potency for Src and Fyn, has been recently completed for the treatment of AD, with encouraging results that supports a larger ongoing Phase Sla clinical trial. Notably, at " the highest dose used one of the subjects treated with Sarasaiinib developed congestive heart failure, whic was linked to the treatment (76]. This dose-limiting adverse effect underscores the importance of highl specific Src family inhibitors in the treatment of different disease.

Another aspeci reiates to the use of a compound as described above in the preparation of a medicament for treating or preventing disorder selected from osteoporosis, a neurological disorder, a proliferative disorder and a viral disorder.

Another aspect relates to the use of a compound as described above in the preparation of a medicament for treating or preventing a proliferative disorder, for example, cancer,

Preferably, the compound Is administered in an amount sufficient to inhibit one or more tyrosine kinases. Mor preferably, the tyrosine kinase is a Src family kinase. As used herein the terra "Src kinase" refers to a member of the Src family of non receptor tyrosine kinases.

Another aspect relates to a compound of the invention for use in the prevention or treatment of a disorder caused by, associated with or accompanied by any abnormal activity against a biological target, wherein the target is a tyrosine kinase, more preferably a Src family kinase,

Yet another aspeci relates to the use of a compound of the invention in the preparatio Of a medicament for the prevention or treatment of a disorde caused by, associated ith or accompanied by any abnormal activity against a biological target, wherein the target is a tyrosine kinase, more preferably a Src family kinase.

Another aspect of the invention relates to a method of treating a iyrosine kinase related disease o disorder, more preferably, a Src kinase related disease or disorder. The method according to this aspect of the present invention is effected by administering to a subject in need thereof a therapeutically effective amount of a compound of the present invention, as described hereinabove, either per se, or, more preferably, as a part of a pharmaceutical composition, mixed with, for example, a pharmaceutically acceptable carrier, as is detailed hereinafter.

Yet another aspect of the invention relates to a method of treating a mammal having a disease state alleviated by inhibition of a tyrosine kinase, more preferably a Src kinase, wherein the method comprises administering to a mammal a therapeutically effective amount of a compound according to the invention.

Another aspect of the invention relates to a method of treating a mammal having a disease state alleviated by the selective inhibition of c-Src kinase, wherein the method comprises administering to a mammal a therapeutically effective amount of a compound according to the invention. Preferably, the disease state is alleviated by the selective inhibition of c-Src kinase over Abl-kinase. Preferably, the mammal is a human.

The term "method" refers to manners, means, techniques and procedures for accomplishing a given task including, but not limited to, those manners, means, techniques and procedures either known to, or readily developed from known manners, means, techniques and procedures by practitioners of the chemical, pharmacological, biological, biochemical and medical arts.

The term "administering" as used herein refers to a method for bringing a compound of the present invention and a protein kinase together in such a manner that the compound can affect the enzyme activity of the protein kinase either directly; i.e., by interacting with the protein kinase itself or indirectly; i.e., by interacting with another molecule on which the catalytic activity of the protein kinase is dependent. As used herein, administration can be accomplished either in vitro, i.e. in a test tube, or in vivo, i.e., in cells or tissues of a living organism.

Herein, the term "treating" includes abrogating, substantially inhibiting, slowing or reversing the progression of a disease or disorder, substantially ameliorating clinical symptoms of a disease or disorder or substantially preventing the appearance of clinical symptoms of a disease or disorder. 2$

Herein, the term "preventing" refers to a method for barring an organism from acquiring a disorder or disease in the first place.

The term "therapeutically effective amount" refers to that amount of the compound being administered which will relieve to some extent one or more of the symptoms of the disease or disorder being treated.

For any compound used in this invention, a therapeutically .effective amount, also referred to herein as a therapeutically effective dose, can be estimated initially from eel Culture assays. For example, a dose can be formuiaied in animal models to achieve a circulating concentratio range that includes the ICso or the IC 0 c as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Initial dosages can also be estimated from in vivo data. Using these initial guidelines one of ordinary skill in the art could determine an effective dosage in humans.

Moreover, toxicity and therapeutic efficacy of the compounds described herein can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., by determining the LD 50 and the ED S0 . The dose ratio between toxic and therapeutic effect is the therapeutic index and can be expressed as the ratio between LDfto and ED 50 . Compounds which exhibit high therapeutic indices are preferred. The data obtained from these ceil cultures assays and animal studies can be used in formulating a dosage range that is not toxic for use in human. The dosage of such compounds lies preferably within a range of circulating concentrations thai include the ED 5 Q with, little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition, (see, e.g. , Fingl stal, 975, in: The

Pharmacological Basis of Therapeutics, chapter 1, page 1).

Dosage amount and interval may be adjusted individually to provide plasma levels of the active compound which are sufficient to maintain therapeutic effect. Usuai patient dosages for oral administration range from about 50-2000 mg/kg/day, commonly from about 100-1000 mg/kg/day, preferably from about 150-700 mg/kg/day and most preferably from about 250-500 mg/kg/day. Preferably, therapeutically effective serum levels will be achieved by administering multiple doses each day. In cases of local administration or selective uptake, the effective local concentration of the drug may not be related to plasma concentration. One skilled in the art will be able to optimize therapeutically effective local dosages without undue experimentation.

As used herein, "tyrosine kinase related disease or disorder" refers to a disease or disorder characterized by inappropriate kinase activity or over-activity. Inappropriate activity refers to either; (i) kinase expression in cells which normally do not express said kinase; (ii) increased kinase expression leading to unwanted cell proliferation, differentiation and/or growth; or, (iii) decreased kinase expression leading to unwanted reductions in cell proliferation, differentiation and/or growth. Over-activity of kinase refers to either amplification of the gene encoding a particular kinase or production of a level of kinase activity, which can correlate with a eel! proliferation, differentiation and/or growth disorder (that is, as the level of the kinase increases, the severity of one or more of the symptoms of the cellular disorder increases). Over activity can also be the result of ligand independent or constitutive activation as a result of mutations such as deletions of a fragment of a kinase responsible for ligand binding.

Preferred diseases or disorders that the compounds described herein may be useful in preventing, include cancer, osteoporosis, neurological disorders such as Parkinson's and Alzheimer's disease and viral disorders such as Epstein Barr Virus, Dengue Virus infection and HIV.

Thus, the present invention further provides use of compounds as defined herein for the manufacture of medicaments for the treatment of diseases where it is desirable to inhibit a tyrosine kinase, more preferably a Src kinase. Such diseases include proliferative disorders, osteoporosis and viral disorders.

SELECTIVITY

Advantageously, selected compounds according to the invention exhibit selectivity for one or more protein kinases, more preferably, one or more tyrosine kinases.

In a preferred embodiment, the compounds of the invention exhibit selectivity for Src- kinase over one or more other protein kinases as measured by an appropriate kinase screening assay. The skilled person would e familiar with such assays, further details of which are provided in the accompanying examples section 177].

More preferably, the compounds of the invention exhibit at least a 2-foid seiectivity for Src-kinase over one or more other protein kinases, preferably, at least 5-fold selectivity, more preferably at least a 10-fold, at least a 25-fold, at least a 50-fo!d, at least a DO-foid, at least a 250-fo!d, at least a 500-foid s or at least a 1000-foid selectivity for Src-kinase over one or more other protein kinases. In a preferred embodiment, the compounds of the invention exhibit at least a 2-fold selectivity for Src-kinase over Ab! kinase, preferably, at least a S-fold selectivity, more preferably at least a 10-fdid, at least a 25-fold, at least a 5G~Told, at least a 100-foid, at least a 250-foid, at ieasi a SQG-fofd, or at least a 1000~foid selectivity for Src-kinase over Abi kinase, for example, as measured by the ratio of C 50 ft0! IC6o 5rc .

PHARMACEUTICAL COiVIPOSiTrOMS

For use according to the present invention, the compounds or physiologically acceptable salt, ester or other physiologically functional derivative thereof, described herein, may be presented as a pharmaceutical formulation, comprising the compounds or physiologically ' acceptable salt, ester or other physiologically functional derivative the eof, tog ther with one or more pharmaceutically acce ta le carriers therefore nd optionally other therapeutic and/or prophylactic ingredients. The carrier{s) must be acceptable in the sense of being compatible with th other ingredients of the formulation and not deleterious to the recipient thereof. The pharmaceuticai compositions may be fo human or animal usage in human and veterinary medicine,

Examples of such suitable excipients for the various different forms of pharmaceutical compositions described herein may foe found in the "Handbook of Pharmaceutical Excipients, 2 nd Edition, (1994), Edited by A Wade and PJ Weller.

Acceptable! carriers or diluents for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington ! s Pharmaceutical Sciences, Mack Publishing Co. (A, R. Gennaro edit, 1985). Examples of suitable carriers include lactose, starch, glucose, methyl cellulose, magnesium stearate, mannitol, sorbitol and the like. Examples of suitable diluents include ethanol, glycerol and water. The choice of pharmaceutical carrier, excipient or diluent can be selected with regard to the intended route of administration and standard pharmaceutical practice. The pharmaceutical compositions may comprise as, or in addition to, the carrier, excipient or diluent any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s), solubilising agent(s), buffer(s), flavouring agent(s), surface active agent(s), thickener(s), preservative(s) (including anti-oxidants) and the like, and substances included for the purpose of rendering the formulation isotonic with the blood of the intended recipient.

Examples of suitable binders include starch, gelatin, natural sugars such as glucose, anhydrous lactose, free-flow lactose, beta-lactose, corn sweeteners, natural and synthetic gums, such as acacia, tragacanth or sodium alginate, carboxymethyl cellulose and polyethylene glycol.

Examples of suitable lubricants include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.

Preservatives, stabilizers, dyes and even flavoring agents may be provided in the pharmaceutical composition. Examples of preservatives include sodium benzoate, sorbic acid and esters of p-hydroxybenzoic acid. Antioxidants and suspending agents may be also used.

Pharmaceutical formulations include those suitable for oral, topical (including dermal, buccal and sublingual), rectal or parenteral (including subcutaneous, intradermal, intramuscular and intravenous), nasal and pulmonary administration e.g., by inhalation. The formulation may, where appropriate, be conveniently presented in discrete dosage units and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing into association an active compound with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired formulation. Pharmaceutical formulations suitable for oral .administration wherein the carrier is a solid are most preferably presented as unit dose formulations such as boluses, capsules or tablets each containing a predetermined amount of active compound. A tablet may be made by compression or moulding, optionally with one or more accessory ingredients. Compressed tablets may be prepared b compressing in a suitable machine an active compound in a free lowing form such as a powder or granules optionally mixed with a binder, lubricant, inert diluent, lubricating agent, surface-active agent or dispersing agent. Moulded tablets may be made by moulding an active compound ith a inert liquid diluent, Tablets may be optionally coated and, if uncoated, may optionally be scored. Capsules may be prepared by filling an active compound, either alone or in admixture with one or more accessory ingredients, into the capsule shells and then sealing them in the usual manner. Cachets are analogous to capsules wherein an active compound together with any accessory ingredients) is sealed in a ric paper envelope. An active compound ma also be formulated as dispersfbie granules, which may far example, be suspended in water before

administration, or sprinkled on food. The granules may be packaged, e.g., in a sachet. Formulations suitable for oral administration wherein the carrier is a liquid may be presented as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oiNn- vater liquid emulsion,

Formulations far oral administration include controlled release dosage forms, e.g., tablets wherein an active compound is formulated in an appropriate release.- controlling matrix, or is coated with a suitable release - controlling film. Such formulations may be particularly convenient for prophylactic use,

Pharmaceutical formulations suitable for rectal administration wherein the carrier is a solid are most preferably presented as unit dose suppositories. Suitable carriers include cocoa butter and other materials commonly used in the art. The suppositories ma be conveniently formed by admixtu e of an active compound with the softened or melted carrier(s) followed by chilling and shaping in moulds. Pharmaceutical

formulations suitable for parenteral administration include sterile solutions or suspensions of an active compound in aqueous or oleaginous vehicles.

Injectable preparations may be adapted fo bolus injection or continuous infusion. Such preparations are conveniently presented in unit dose o multi-dose containers which are sealed after introduction of the formulation until required for use. Alternatively, an active compound may be in powder form which is constituted with a suitable vehicle, such as sterile, pyrogen-free water, before use. An active compound may also be formulated as long-acting depot preparations, which may be administered by intramuscular injection or by implantation, e.g., subcutaneously or intramuscularly. Depot preparations may include, for example, suitable polymeric or hydrophobic materials, or ion-exchange resins. Such long-acting formulations are particularly convenient for prophylactic use.

Formulations suitable for pulmonary administration via the buccal cavity are presented such that particles containing an active compound and desirably having a diameter in the range of 0.5 to 7 microns are delivered in the bronchial tree of the recipient. As one possibility such formulations are in the form of finely comminuted powders which may conveniently be presented either in a pierceable capsule, suitably of, for example, gelatin, for use in an inhalation device, or alternatively as a self-propelling formulation comprising an active compound, a suitable liquid or gaseous propellant and optionally other ingredients such as a surfactant and/or a solid diluent. Suitable liquid propellants include propane and the chlorofluorocarbons, and suitable gaseous propellants include carbon dioxide. Self-propelling formulations may also be employed wherein an active compound is dispensed in the form of droplets of solution or suspension.

Such self-propelling formulations are analogous to those known in the art and may be prepared by established procedures. Suitably they are presented in a container provided with either a manually-operable or automatically functioning valve having the desired spray characteristics; advantageously the valve is of a metered type delivering a fixed volume, for example, 25 to 100 microlitres, upon each operation thereof. As a further possibility an active compound may be in the form of a solution or suspension for use in an atomizer or nebuiiser whereby an accelerated airstream or ultrasonic agitation is employed to produce a fine droplet mist for inhalation.

Formulations suitable for nasal administration include preparations generally similar to those described above for pulmonary administration. When dispensed such formulations should desirabiy have a particle diameter in the range 10 to 200 microns to enable retention in the nasal cavity; this may be achieved by, as appropriate, use of a powder of a suitable particle size o choice of ai appropriate valve. Other suitable formulations include coarse powders having a particle diameter in the range 20 to 500 microns, for administration by rapid inhalation through the nasal passage from a container held dose up to the nose, and nasal drops comprising 0.2 to §% w/v of an active compound in aqueous or oily solution or suspension.

Pharmaceuticali acceptable carriers are well known to those skilled in the art and include, but are not limited to, 0.1 and preferably 0.05 M phosphate buffer or 0.8% saline. Additionally, such pharmaceutically acceptable carriers may be aqueous or nonaqueous solutions, suspensions, and emulsions. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate, Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactafed Ringer's or fixed o ls; Preservatives and other additives may also be present, such as, for example, antimicrobials, antioxidants, chelating agents, inert gases and the like.

Formulations suitable for toxical formulation ma be provided for example as gels, creams or ointments. Such preparations may be applied e.g. to a wound or ulcer either directly spread upon the surface of the wound or ulcer or carried on a suitable support such as a bandage, gauze, mesh or the like which may be applied to and over the area to be treated.

Liquid or powder formulations may also be provided which can be sprayed or sprinkled directly onto the site to be treated, e.g. a wound or ulcer, Alternatively, a carrier such as a bandage, gauze, mesh or the like can foe sprayed or sprinkle with the formulation and the applied to the site to be treated.

According to a further aspect of the invention, there is provided a process for the preparation of a pharmaceutical or veterinary composition as described above, the process comprising bringing the active compoundfs) into association with the carrier, for example by admixture. In general, the formulations are prepared by uniformly and intimately bringing into association the active agent with liquid carriers or finely divided solid carriers or both, and then if necessary shaping the product. The invention extends to methods for preparing a pharmaceutical composition comprising bringing a compound of general formulae (I) or (II) in conjunction or association with a pharmaceutically or veterinarily acceptable carrier or vehicle.

SALTS/ESTERS

The compounds of the invention can be present as salts or esters, in particular pharmaceutically and veterinarily acceptable salts or esters.

Pharmaceutically acceptable salts of the compounds of the invention include suitable acid addition or base salts thereof. A review of suitable pharmaceutical salts may be found in Berge ef a/, J Pharm Sci, 66, 1-19 (1977). Salts are formed, for example with strong inorganic acids such as mineral acids, e.g. hydrohalic acids such as

hydrochloride, hydrobromide and hydroiodide, sulphuric acid, phosphoric acid sulphate, bisulphate, hemisulphate, thiocyanate, persulphate and sulphonic acids; with strong organic carboxylic acids, such as alkanecarboxylic acids of 1 to 4 carbon atoms which are unsubstituted or substituted (e.g., by halogen), such as acetic acid; with saturated or unsaturated dicarboxylic acids, for example oxalic, malonic, succinic, maleic, fumaric, phthalic or tetraphthalic; with hydroxycarboxylic acids, for example ascorbic, giycolic, lactic, malic, tartaric or citric acid; with aminoacids, for example aspartic or glutamic acid; with benzoic acid; or with organic sulfonic acids, such as (Ci-C 4 )-alkyl- or aryl- sulfonic acids which are unsubstituted or substituted (for example, by a halogen) such as methane- or p-toluene sulfonic acid. Salts which are not pharmaceutically or veterinarily acceptable may still be valuable as intermediates.

Preferred salts include, for example, acetate, trifluoroacetate, lactate, gluconate, citrate, tartrate, maleate, malate, pantothenate, adipate, alginate, aspartate, benzoate, butyrate, digluconate, cyclopentanate, glucoheptanate, glycerophosphate, oxalate, heptanoate, hexanoate, fumarate, nicotinate, palmoate, pectinate, 3-phenylpropionate, picrate, pivalate, proprionate, tartrate, lactobionate, pivolate, camphorate, undecanoate and succinate, organic sulphonic acids such as methanesu!phonate, ethanesufphonate, 2-hydroxyethane sulphonate, camphorsulphonate, 2-naphthalenesuiphonate, benzenesulphonate, p-chlorobenzenesulphonate and p-toluenesulphonate; and inorganic acids such as hydrochloride, . , hydrobromide, hydroiodide, sulphate, bisuiphafe, hemisu!phaie:, thiocyanate, persulphate, phosphoric and sulphonic acids.

Esters are formed either using organic acids or alcohols/hydroxides, depending on the functional group being esierified. Organic acids include carboxyiie acids, such as alkanecarboxylic acids of 1 to 12 carbon atoms which are unsubsistuied or substituted (e.g., by halogen), such as acetic acid; with saturated or unsaturated dicarpoxyiic acid, for exampie oxalic, maionie, succinic, maie ' ic, . fumaric, phthalic or ietraphtha!ie; with hydroxycarboxyiic acids, for example ascorbic, glycolic, lactic, malic, tartaric or citric acid; with amlnoacids, for example aspart!c or glutamic acid; with benzoic acid; or with organic sulfonic acids, or aryi-suifonic acids which are

unsubstituted or substituted (for example, b a halogen) such as methane- or p-toiuene; sulfonic acid. Suitable hydroxides include inorganic hydroxides, such as sodium hydroxide, potassium hydroxide, calcium hydroxide, aluminium hydroxide. Alcohols include alkahealcohols of 1-12 carbon atoms which may be unsubstituted or

subst i tuted, e.g. by a halogen).

EMA TIO^E S/TAUTOMERS

In all aspects of the present invention previousl discussed, the invention includes, where appropriate ail enantiomers, diastereoisomers and tautomers of the compounds of the invention. The person skilled in the art will recognise compounds that possess optical properties (one or more chiral carbon atoms) or tautomeric characteristics. The corresponding enantiomers and/or tautomers may be isolated/prepared by methods known in the art.

Enantiomers are characterised by the absolute configuration of their chira! centres and described by the R- and ' ^-sequencing rules of Cahn, Ingold and Preiog. Such conventions are well known in the art (e g, see 'Advanced Organic Chemistry', 3 fd edition, ed. March, X, John Wiley and Sons, New York, 1085).

Compounds of the invention containing a chiral centre may foe used as a raeemic mixture, an enantiomeriGally enriched mixture, or the raeemic mixture may be separated using well-known techniques and a individual enantiomer may be used alone. STEREO AND GEOMETRIC ISOMERS

Some of the compounds of the invention may exist as stereoisomers and/or geometric isomers - e.g. they may possess one or more asymmetric and/or geometric centres and so may exist in two or more stereoisomeric and/or geometric forms. The present invention contemplates the use of all the individual stereoisomers and geometric isomers of those inhibitor agents, and mixtures thereof. The terms used in the claims encompass these forms, provided said forms retain the appropriate functional activity (though not necessarily to the same degree). The present invention also includes all suitable isotopic variations of the agent or a pharmaceutically acceptable salt thereof. An isotopic variation of an agent of the present invention or a pharmaceutically acceptable salt thereof is defined as one in which at least one atom is replaced by an atom having the same atomic number but an atomic mass different from the atomic mass usually found in nature. Examples of isotopes that can be incorporated into the agent and pharmaceutically acceptable salts thereof include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulphur, fluorine and chlorine such as 2 H, 3 H, 3 C, 4 C, 15 N, 17 0, 18 0, 31 P, 32 P, 3S S, 8 F and 36 CI, respectively. Certain isotopic variations of the agent and pharmaceutically acceptable salts thereof, for example, those in which a radioactive isotope such as 3 H or 14 C is incorporated, are useful in drug and/or substrate tissue distribution studies. Tritiated, i.e., 3 H, and carbon-14, i.e., 1 C, isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with isotopes such as deuterium, i.e., 2 H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements and hence may be preferred in some circumstances. For example, the invention includes compounds of general formulae (I) or (if) where any hydrogen atom has been replaced by a deuterium atom. Isotopic variations of the agent of the present invention and pharmaceutically acceptable salts thereof of this invention can generally be prepared by conventional procedures using appropriate isotopic variations of suitable reagents.

PRODRUGS

The invention further includes the compounds of the present invention in prodrug form, i.e. covalently bonded compounds which release the active parent drug according to general formula (I) or (II) in vivo. Such prodrugs are generally compounds of the invention wherein one or more appropriate groups have been modified such that the modification may be reversed upon administration to a human or mamrttaiian subject Reversion is usually performed by an enzyme naturally present in such subject, though it is possible for a second agent to be administered together with such a prodrug in order to perform the reversion in vivo: Examples of such modifications Include ester (for example, any of those described above), wherein the reversion may be carried out be a esterase etc. Other such systems will be well known to those skilled in the art.

SOLVATES

The present invention also includes solvate forms of the compounds of the present Invention. The terms used in the claims encompass these forms.

POLYMORPHS

The invention further relates to the compounds of the present invention in their various crystalline forms, polymorphic forms and (an)hydrous forms. It is well established within the pharmaceutical industry that chemical compounds may be isolated in any of such forms b slightly varying the method of purification and or isolation form the solvents used in the synthetic preparation of such compounds.

ADMINISTRATION

The pharmaceutical compositions of the present invention may b adapted fo rectal, nasal, intrabronchial, topical (including buccal and sublingual), vaginal ' or parenteral (including subcutaneous, intramuscular, intravenous, Intraarterial and intradermal), intraperitoneal or intrathecal administration. Preferably the formulation is a orally administered formulation. The formulations may conveniently be presented in unit dosage form, i.e., in the form of discrete portions containing a unit dose, or a muitipie or sub-unit of a unit dose. Byway of example, the formulations ma be in the form of tablets and sustained release capsules, and may be prepared by .any method well known in the art of pharmacy. Formulations for oral administration in the present invention may be presented as:

discrete units such as capsuies, geSlules, drops, cachets, pills or tablets each containing a predetermined amount of the active agent; as a powder or granules; as a solution, emulsion or a suspension of the active agent in an aqueous liquid or a non-aqueous liquid; or as an oil-ih-water liquid emulsion or a vvater-in-oil liquid emulsion; or .as a bolus etc. Preferably, these compositions contain from 1 to 250 mg and more preferably from 10-100 mg, of active ingredient per dose.

For compositions for oral administration (e.g. tablets and capsules), the term

"acceptable carrier" includes vehicles such as common excipients e.g. binding agents, for example syrup, acacia, gelatin, sorbitol, tragacanth, polyvinylpyrrolidone (Povidone), methylceilulose, ethy!cellulose, sodium carboxymeihy!cellulose, hydroxypropyl- methylcellulose, sucrose and starch; fillers and carriers, for example corn starch, gelatin, lactose, sucrose, microcrystalline cellulose, kaolin, mannitol, dicalcium phosphate, sodium chloride and alginic acid; and lubricants such as magnesium stearate, sodium stearate and other metallic stearates, glycerol stearate stearic acid, silicone fluid, talc waxes, oils and colloidal silica. Flavouring agents such as peppermint, oil of wintergreen, cherry flavouring and the like can also be used. It may be desirable to add a colouring agent to make the dosage form readily identifiable. Tablets may also be coated by methods well known in the art.

A tablet may be made by compression or moulding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active agent in a free flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface-active or dispersing agent. Moulded tablets may be made by moulding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may be optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active agent.

Other formulations suitable for oral administration include lozenges comprising the active agent in a flavoured base, usually sucrose and acacia or tragacanth; pastilles comprising the active agent in an inert base such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active agent in a suitable liquid carrier.

Other forms of administration comprise solutions or emulsions which may be injected intravenously, intraarterially, intrathecally, subcutaneously, intradermally,

intraperitoneaily or intramuscularly, and which are prepared from sterile or sterilisable solutions. Injectable forms typically contain between 10 - 1000 mg, preferably between 10 - 250 mg, of active ingredient per dose. The pharmaceutical compositions of the present invention may also be in form of suppositories, pessaries, suspensions, emulsions, lotions, ointments, creams, gets, sprays, solutions or dusting powders. An alternative means of transdermal administration is by use of a skin patch. For example, the active ingredient can be incorporated into a cream consisting of an aqueous emulsion of polyethylene ' .glycols or liquid paraffin. The active ingredient can also be incorporated : , at a concentration of between 1 and 10% by weight, into an ointment consisting of a white wax or white soft paraffin base together with such stabilisers and preservatives as may foe required.

A person of ordinary skiil in the art can easily determine an appropriate dose of one of the instant compositions to administer to a subject without undue experimentation. Typically, a physician will determine the actual dosage which will be most suitable for an individual patient and it will depend on a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body eight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the individual undergoing therapy. The dosages disclosed herein are exemplary of the average case. There can of course be individual instances where higher or lower dosage ranges are merited, and suc are within the scope of this invention. I n accordance with this invention, an effective amount of .a compound of general formulae (I) or(ii) may be administered to inhibit the kinase implicated with a particular condition or disease. Of course, this dosage amount will further foe modified according to the type of administration of the compound. For example, to achieve an "effective amounf for acute therapy, parenteral administration of a compound of general formula (I) or (it) is preferred. An intravenous infusion of the compound in 5% dextrose in water or normal saline, or a similar formulation with suitable excipients, is most effective, although an intramuscular bolus injection is also useful. Typically, the parenteral dose will be about 0,01 to about iOO mg/kg; preferably between 0.1 and 20 mg/kg, in a manner to maintain the concentration of drug in the plasma at a concentratio effective to inhibit a kinase. The compounds may be administered one to four times dail at a level to achieve a total daily dose of about 0.4 to about 400 mg/kg/day. The precise amount of an inventive compound which is therapeutically effective, and the route by which such compound is best administered, is readily determined by one of ordinary skill in the art by comparing the blood level of the agent to the concentration required to have a therapeutic effect.

The compounds of this invention may also be administered orally to the patient, in a manner such that the concentration of drug is sufficient to achieve one or more of the therapeutic indications disclosed herein. Typically, a pharmaceutical composition containing the compound is administered at an oral dose of between about 0.1 to about 50 mg/kg in a manner consistent with the condition of the patient. Preferably the oral dose would be about 0.5 to about 20 mg/kg.

No unacceptable toxicoiogica! effects are expected when compounds of the present invention are administered in accordance with the present invention. The compounds of this invention, which may have good bioavailability, may be tested in one of several biological assays to determine the concentration of a compound which is required to have a given pharmacological effect. COMBINATIONS

In a particularly preferred embodiment, the one or more compounds of the invention are administered in combination with one or more other active agents, for example, existing drugs available on the market. In such cases, the compounds of the invention may be administered consecutively, simultaneously or sequentially with the one or more other active agents.

Drugs in general are more effective when used in combination. In particular, combination therapy is desirable in order to avoid an overlap of major toxicities, mechanism of action and resistance mechanism(s). Furthermore, it is also desirable to administer most drugs at their maximum tolerated doses with minimum time intervals between such doses. The major advantages of combining chemotherapeutic drugs are that it may promote additive or possible synergistic effects through biochemical interactions and also may decrease or delay the emergence of resistance. Beneficial combinations may be suggested by studying the inhibito ' r activity of the test compounds with agents known or suspected of being valuable in the ' treatment of a particular disorder, For example, the invention relates to the use of a compound as described above in an assay for identifying compounds that promote additive and synergistic activit Upon anti-cahcer activities when combined with the compound.

Preferabfy the assay is a ' high-throughput cell based phenotypic screen. This procedure can also be used to determine the order of administration of the agents, i.e> before, simultaneously, or after delivery. Suc scheduling may be a feature of all the active agents identified herein. ASSAY

A further aspect of the invention relates to the use of a compound as described above in an assay for identifying further candidate compounds capable of inhibiting or selectively inhibiting one or more tyrosine kinases, more preferably a Src family kinase. Preferably, the candidate compound is capable of selectively inhibiting c-Src kinase over Abi-klnase.

Preferably, the assay is a competitive binding assay.

!vlore preferably, the competitive binding assay comprises contacting a compound of the invention with the kinase, and a candidate compound and detecting any change in the interaction between the compound according to the invention and the kinase.

Preferably, the candidate compound is generated by conventional SAR modification of a compound of the invention,

As used herein, the term "conventional SAR modification" refers to standard methods known in the art for varying a give compound b way of chemical derivatisation.

Thus, in one aspect, the identified compound may act as a mode! (for exampie, a template) for the development of other compounds. The compounds employed in such a test may be free in solution, affixed to solid support, borne o a cell surface, or located intraceliuiarly. The abolition of activity or the formation of binding complexes between the compound and the agent being tested may be measured. The assay of the present invention may be a screen, whereby a number of agents are tested, in one aspect, the assay method of the present invention is a high throughput screen. This invention also contemplates the use of competitive drug screening assays in which neutralising antibodies capable of binding a compound specifically compete with a test compound for binding to a compound.

Another technique for screening provides for high throughput screening (HTS) of agents having suitable binding affinity to the substances and is based upon the method described in detail in WO 84/03564.

It is expected that the assay methods of the present invention will be suitable for both small and large-scale screening of test compounds as well as in quantitative assays.

Preferably, the competitive binding assay comprises contacting a compound of the invention with a kinase in the presence of a known substrate of said kinase and detecting any change in the interaction between said kinase and said known substrate. A further aspect of the invention provides a method of detecting the binding of a ligand to a kinase, said method comprising the steps of:

(i) contacting a ligand with a kinase in the presence of a known substrate of said kinase;

(ii) detecting any change in the interaction between said kinase and said known substrate;

and wherein said ligand is a compound of the invention.

One aspect of the invention relates to a process comprising the steps of:

(a) performing an assay method described hereinabove;

(b) identifying one or more ligands capable of binding to a ligand binding domain; and

(c) preparing a quantity of said one or more ligands. Another aspect of the invention provides a process comprising the steps of;

(a) performing an assay method described hereinabove;

•(b) identifying one or more ligands capabl of binding to a ligand binding domain; and

(c) preparing a pharmaceutical composition comprising said one or more ligands.

Another aspect of the invention provides a process com prising the steps of:

(a) performing an assay method described hereinabove;

(b) identifying one or more ligands capable of binding to a ligan binding domain;

(c) modifying said one or more ligands capable of binding to a ligand binding

domain;

(d) performing the assay method described hereinabove;

(e) optionally preparing a pharrnaoeutioal comfsosition comprising said one or more iigands.

The invention also relates to a ligand identified by the method described hereinabove.

Yet another aspect of the invention relates to a pharmaceutical composition comprising a ligand identified by the method described hereinabove.

Another aspect of the invention relates to the use of a ligand Identified by the method described hereinabove in the preparation of a pharmaceutical composition for use in the treatment of one or more disorders as described hereinabove.

The above methods may be used to screen for a ligand useful as an inhibitor of one or more kinases.

Compounds of general formulae (1) or (IS) are useful both as laboratory tools and as therapeutic agents, in the laboratory certain compounds of the invention are useful in establishing whether known or newly discovered kinase contributes a critical or at least significant biochemical function during the establishment or progression of a disease state, a process commonly referred to as 'target validation'. SYNTHESIS

Another aspect of the invention relates to a process for preparing a compound of formula I as defined above, said process comprising the steps of:

(i) converting a compound of formula (III) to a compound of formula (IV);

(ii) converting said compound of formula (IV) to a compound of formula (V);

(iii) converting said compound of formula (V) to a compound of formula (VI); and

(iv) converting said compound of formula (VI) to a compound of formula (I).

A preferred synthetic scheme for preparing compounds according to the invention is set out in Figure 2. Preferred reagents for steps (i) to (iv) are as shown in Figure 2.

By way of illustration 4-aminopyrazolopyrimidine was synthesized by microwave- assisted reaction of pyrazole in an excess of formamide. /V-lodosuccinimide (NIS) mediated iodination, W-alkylation with bromoacetaidehyde diethyl acetal, followed by Suzuki cross-coupiing with the corresponding arylboronic acid produced acetal- protected derivative in good overall yield (25-50 %, 3 steps). Quantitative acetal depratection in TFA:water (1 :1) gave rise the corresponding ' aldehyde derivative, which was used to generate the final compounds by reductive amination with cyclic secondar amines (piperidines, morphoiine and piperaanes), The invention is further described by way of the following non-limiting examples, and with reference to the following figures, wherein;

Figure 1 shows: Upper panel; ATP and PPi {neutral forms); Lower panel: general structure of the novei compounds and their reirosynthetic analysis.

Figure 2 shows a preferred synthetic scheme for preparing compounds according to the invention.

Figure 3 shows: a) Compounds PP20 [351, 506 and 503; b) ECso values calculated after incubation of MCF7 and MDA-MB-231 cells with compounds S08 and 503, Dasatinib was used as positive control; c) ECso values calculated after incubation of SYF cells with compounds 508, 503 and Dasatinib,

Figure 4 shows a full kinome screen dot plot of compound 506 at 1 pfVi. Enzymatic aciivity of most kinases tested (321) was not, or only Weakly, affected by compound 506 (activity >35%). Although 21 kinases were identified as potential hits (<35%), compound 506 was most active against 3 kinases (0-0,5% of activity) at the concentration tested: c-Src, Yes and PTK6. Calculation of iC 60 values for a. panel of kinases showed that e- Src and Yes were i fact inhibited at sub-nartoM range. However, PT 8 (also known as Brk, breast cancer kinase) showed SCg 0 ~ 2Qn!Vl. Other Srcfa ily kinases were also identified to have IG 60 in the low nanoH; range,

Figure 5 shows a Wester Blot on MDA-MB-231 cells comparing compound 506 with Dasatinib. Compound 506 induced strong inhibition of SRC auto-phosphorylation ( Y 18 phospho-S C) and downstream SRC-mediated phosphorylation of FAK

( ye61 phospho-FA ). While Dasatinib inhibited pSRG and p FAK at the same

concentrations, it also displayed additional effects in other pathways (strong

upregulation of pA T) and seemed to stabilize total SRC. Figure 6 shows the antagonistic effect of G F-2 (selective Abl inhibitor) over the EC 50 values of compound 506. As shown in the Figure, EC 50 values are incremented up to 50-fold at 10 μΜ of GNF-2. Figure 7 shows the migration of MDA-MB-231 treated cells across a scratch wound at 6h, 12h and 24h time points normalized to D SO. Drugs, compound 506 and

Dasatinib, were used at 10 nM and compared against DMSO.

Figure 8 shows a zebrafish tail regeneration assay. Procedure: 2 hour pre-treatment with dasatinib or compound 506 at 100μΜ -> tail cut- treatment for 2 more hours - wash out, image 2 days later. Note heart enlargement induced by dasatinib treatment.

Figure 9 shows phenotypic screening of SRC inhibitor 506 in zebrafish. (a,b) Neuromast migration assay. Fresh E3 media with DMSO or 506 (500 μΜ) was added to zebrafish embryos at 20 hpf, 36 hpf and 48 hpf, and imaged at 72 hpf. (a) Representative images of the tail of a 3 dpf zebrafish without (top) and with 506 treatment (bottom).

Neuromasts are identified by GFP expression (green) and the tip of the tail as a red line. Yellow arrow indicates shortest distance from tail tip to a neuromast. (b) Imaging analysis of the distance between the last neuromast and the tip of the tail (n=10) under treatment with DMSO (negative control) or 506 (500 μΜ). P value calculated from i-test. (c) Study of zebrafish heart development under short treatment with 506 (500 μΜ) and dasatinib (10 μΜ). Compounds were added to 2 dpf zebrafish embryos and incubated for 4 h (n=5). Subsequently, fresh media was added and fish imaged after 48 h incubation.

Figure 10 shows immunohistochemical analysis of phospho-SRC Y416 in human tumor xenografts, (a) Images of representative sections (low and high resolution) of HCT1 16 xenografts from: (left) untreated mice and (right) mice treated with 506 (n=4). (b) Histoscore analysis (6-7 sections analysed per experiment). Quantification of immunohistochemistry across tumor tissue sections from untreated animals (water) and 506 treated groups performed in blinded fashion. P value calculated from i-test.

EXAMPLES

Materials and Methods

General procedures for synthesis of compounds Chromatography

Column chromatography refers to silica gel chromatography and was carried out manually using conventional glass cclumnsand silica gel (pore size 60A, particle size 230-400 mesh, 40-63 μϊΤί) from Sigma-Aldrich.

Analytical Methods

fH Nuclear magnetic resonance (N ) spectroscopy was carried out using a 500 MHz Bruker Avance ill spectrometer in the stated solvent at around room temperature uniess otherwise stated, in a!i cases, NMR data were consistent with the proposed structures. Characteristic chemical shifts (δ) are given in parts-per-miliion using conventional abbreviations for designation of major peaks: e.g. s, singlet; d, doublet; t, triplet; q, quartet; dd, doublet of doublets; br, broad. Low Resolution Mass Spectra (LR S) were obtained using a iv icrosaic Systems 4000 MiD system under electron spray ionisation (ESI) conditions. High Resolution Mass Spectra (HR S) were obtained using a Bruker 3.0 T Apex ii Spectrometer. Thin layer chromatog aphy was run on Ivlerck TLC Silica gel 80 F254 plates; typically 5 cm x 10 cm. Detection was achieved using a 254 nm UV source or permanganate stain. Compound preparation

Where the preparation of starting materials is not described, these are commercially available, known in the literature, or readily obtainable by those skilled in the art using standard procedures. All commercially available chemicals used herein were obtained from either Fisher Scientific, Matri Scientific, Sigma-Aldrich or VWR International Ltd. Where it is stated that compounds were prepared analogously to earlier examples or intermediates, it will be appreciated by the skiiled person that the reaction time, number of equivalents of reagents and temperature can be modified for each specific reaction and thai it may be necessary or desirable to employ different work-up or purification techniques. Where reactions were carried out using microwave irradiation, the microwave used was an Initiator 80 supplied by Biotage. Non-microwave reactions were performed under an inert atmosphere of nitrogen using anhydrous solvents. The actual power supplied varies during the course of the reaction in order to maintain a constant temperature, Abbreviations CDCIg = Deuterated Chloroform

DCM = Dichloromethane

Et 2 0 = Diethyl ether

MgS0 4 = Magnesium sulfate

DMF = /V,A/-Dimethylformamide

eq. = Equivalents

mg - Milligram

mi = Millilitre

mmol = Millimoles

m/z = Mass to charge ratio

eOD = Deuterated Methanol

MHz = Mega Hertz

mw = Microwave

TLC = Thin Layer Chromatography

NEt 3 = Triethy!amine

THF = Tetrahydrofuran

MeOH = Methanol

TFA = Trifluoroacetic acid

r.t. = Room temperature

AcOH = Acetic acid

EtOH = Ethanol

EtOAc = Ethyl Acetate

UV = Ultraviolet

DMSO = Dimethylsulphoxide

The synthesis of selected compounds of the invention is described below.

1H-pyrazolo[3,4-d]pyrimidin-4-amine

5-amino-1 H-pyrazole-4-carbonitrile (3 g, 27.77 mmol) and formamide (15 ml) were added to a 20 ml microwave vial and the mixture heated at 180 °C for 2 hours using microwave radiation. The precipitate formed on cooling was filtered off and washed with water (50 ml) and allowed to dry giving the product as a cream solid (3.5 g, 25.92 mmol, 93 ). 1 H HMR (500 MHz, DMSO) δ ' 13.34 (s, 1.H), 8.13 (s : H). 8,07 (g, 1 H), 7,69 (br. m, 2H); n C HMR (126 MHz, DMSO) δ 158.19 (CH), 156,03 (C), 154.98 (C), 132,79 (CH),99.83 (C); SSS (ES +ve) [M+ f: 136.0, 157.9 (+Naj, (ES -ve) [ -ΗΓ: 133.9. 3-iodo~1 H~pyrazoSo[3,4^c 3pyrimidisi-

1.H<-pyrazoloI3.4-d]pyri ' rnidtn-4~amine- {1..5 g, 11.11 mmoi) was suspended in 15 m! of DMF and N-iodosuccinimide (1.2 eq„ 3.0 g, 13,34 mmoi) added. The mixture was heated at 180 °C in the microwave for an hour. EtOH (80 mi) was added to the reaction and a precipitate began to form, which was aided by sonicatibn, The precipitate was filtered and washed with EtOH (x3, 20 ' ml) and ' allowed to dry in an oven at 40 °C overnight to give a sand coloured solid (2.115 g, 8.105 mmoi, 73,0 %). Ή NNIR {500 MHZ, DMSO) δ 13,80 (s, 1 H), 8.16 (s, 1 H), 7 79 - 8.44 (m, 2H); C HMR (126 MHz, DMSO) δ 157.60 (C), 156.08 (CH), 155.04 (C), 102.50 (G), 89.82 (G); MS (ES +ve) [M+H]*: 283.9 (+Na), (ES -ve) f -Hf: 259.9, 287,8 (+Na).

1 -(2,2-diethoxyethyl)-3-iodo-pyrazolo|3 > 4-d]pyrimidin-4-amine:

To a solution of S-iodo-iH-pyrazotolS^-dipyrimidin^-amine (500 rag, 1 .92 mmoi) i

DMF (15 ml) was added sodium hydride (1.5 eq,, 2.88 mmoi, 80 % dispersion in mineral oil, 116.2 mg) and the solution allowed to stir for 30 mins until the gas evolution had subsided, Bromoacetaidehyde diethyl acetal (1 .5 eq. 2.88 mmo ' l, 0.435 mi) was then added dropwise and the mixture heated at 150 °C in the microwave for 40 mins. EtOAc and water (50 ml) were added to the mixture and the organics separated. The aqueous iayer was washed With EtOAc (50 ml, x3) and the organics combined and washed with water (x3 : 30 ml), dried over MgSQ 4 and concentrated in vacuo. The crude product was purified by column chromatography (vleGH/DG! (0-5 %) to give a light orange solid (461 mg, 1.22 mmol, 63.7 %). 1 H NMR (500 MHz, DMSO) δ 8.21 (s, 1 H), 7.90 - 6.30 (m, 2H), 4.93 (t, J = 5.7, 1 H), 4.33 (d, J = 5.8, 2H), 3.62 (dq, J = 9.4, 6.9, 2H), 3.40 (dq, J = 9.6, 7.0, 2H), 0.98 (t, J = 7.0, 6H); 3 C NMR (126 MHz, DMSO) δ 157.86 (C), 156.30 (CH), 154.03 (C), 103.18 (CH), 99.50 (C), 89.51 (C), 61.39 (CH 2 ), 48.76 (CH 2 ), 15.39 (CH 3 ); MS (ES +ve) [M+H] + : 377.8, 400.0 (+Na), (ES -ve) [M-H]-: 376.0.

1-(2,2^ϊθΙΗοχγ6ΐ!ινΙ)-3-(ρ-ίοΙγΙ)ρ/Γ3 θΙο[3,4^3ρνπηιί(ϋη-4-3ηιϊηβ:

To a solution of 1-(2,2-diethoxyethyl)-3-iodo-pyrazolo[3,4-d]pyrimidin-4-amin e (1.135 g, 3.01 mmol) in dioxane/waier (10 ml/1 ml) was added p-tolylboronic acid (1.5 eq., 614 mg, 4.52 mmol), potassium carbonate (1.5 eq., 624.7 mg, 4.52 mmol) followed by palladium acetate (5 mol %, 33.8 mg) and the mixture heated in the microwave at 20 °C for an hour. EtOAc and water (50 ml) were added to the mixture and the organic layer separated, dried over MgS0 4 and concentrated in vacuo. The crude product was purified by column chromatography, MeOH/DCM (0-5 %) to give a light brown solid (902 mg, 2.64 mmol, 87.8 %). 1 H NMR (500 MHz, CDCI 3 ) δ 8.42 (s, 1 H), 7.58 (d, J = 8.0, 2H), 7.34 (d, J = 7.8, 2H), 5.12 (t, J = 5.8, 1 H), 4.58 (d, J = 5.8, 2H), 3.78 (dq, J = 9.4, 7.0, 2H), 3.52 (dq, J = 9.4, 7.0, 2H), 2.44 (s, 3H), 1.12 (t, J = 7.0, 6H); 13 C NMR (126 MHz, CDCI 3 ) 6 158.28 (C), 156.43 (C), 155.55 (CH), 145.25 (C), 139.76 (C), 131.90 (CH), 128.93 (CH), 100.49 (CH), 62.46 (CH 2 ), 49.53 (CH 2 ), 21.09 (CH 3 ), 15.78 (CH 3 ); MS (ES +ve) [M+1] + : 341.19, (ES -ve) [M-1] " : 340.0.

2-[4-amino-3-(p-tolyI)pyrazolo[3,4-d]pyrimidin-1-yl]aceta Idehyde:

To a suspension of 1-(2,2~diethoxyethyi)-3-(p-to^^

(400 mg, 1.17 mmol) in 5 ml of water was added 3 ml of TFA and the mixture heated to 100 °C for 30 mins in the microwave. The mixture was transferred to a RBF, washed with DCM and concenirated in vacuo. The product was washed with DCM and B ? 0 and dried in vacuo to give a jight brown solid (assuming quantitative yield). N MR spectra were hot obtained for this compound as different salts of the product led to messy spectra. Compounds 106 & 109. To a suspension of 2-[4-am!no-3 p-tolyi}pyraZoioi3;4- d]pyrimidm~1-yl}aeetaidehyde (40 mg, 0,105 mmo|) in DCM (1 ml) was added either ,N- dime hy!--2-p!peraz!n--1-yi-ethanamihe or N, -dimethy]p!peridtn-4-amine (1 eq., 0.105 mmol), respectively, and a drop of AcOH and the mixture alio ed to stir for 10 mins. Sodium triacefoxyborohydride (22.2 mg, 0.105 mmol} was then added and the mixture stirred until complete (~ 1 hr). The reaction mixture was concentrated in vacuo and purified without any further work up due to the high solubility of the product in the aqueous layer,

1 -|2-(4-ϊΤί .ethyl p J perazi n*1 -y I ethy S] -3-{ p -toly ! }py raaol o[3 s 4-d] py rs kl I n-4-amt ne {103}:

Purified by column chromatography (MeOH/DCM 0-10% - 10% MeOH with 3 drops of NH 3 aq. per 20 ml) to give a pale orange solid (20 mg, 0.057 mmol, 54.2 %) H NMR (500 MHz, MeOD) δ 8.27 (s, 1 H), 7.60 (d, J = 8.1, 2H), 7.41 (d, J = 7.8, 2H), 4.57 (t, J = 6.5, 2H), 2.97 (t, J = 6.5, 2H), 2.53 (m, 8H), 2.47 (s, 3H), 2.37 (s, 3H).; 13 C NMR (126 MHz, CDCI 3 ) 6 158.02 (C), 155.87 (CH), 154.77 (C), 144.58 (C), 139.25 (C), 130.45 (C), 130.14 (CH), 128.50 (CH), 98.59 (C), 56.89 (CH 2 ), 54.91 (CH 2 ), 52.50 (CH 2 ), 45.59 (CH 3 ), 44.60 (CH 2 ), 21.47 (CH 3 ); MS (ES +ve) [ +1]*: 352.0, 374.2 (+Na), (ES -ve) [M- 1] " : 350.2; HRMS (ES +ve), C 19 H 26 N 7 [M+H] + : calculated 352.22442, found 352.224816. 1-[2-[4-(2-dimethyEaminoethyl)piperazin-1-yl]ethyl]-3-(p-tol yl)pyrazolo[3,4- d]pyrimidin-4-amine (105):

Purified by column chromatography (MeOH/DCM 5-10% - 10% MeOH with 25 drops of NH 3 aq. per 100 ml) to give a pale orange solid (5 mg, 0.0312 mmol, 11.7). 1 H NMR (500 MHz, MeOD) δ 8.27 (s, 1 H), 7.59 (d, J = 8.0, 2H), 7.41 (d, J = 8.0, 2H), 4.56 (t, J = 6.6, 2H), 2.96 (t, J = 6.6, 2H), 2.89 (t, J = 6.6, 2H), 2.76 - 2.60 (m, 6H), 2.60 (s, 6H), 2.51 (m, 4H), 2.46 (s, 3H); 13 C NMR (126 MHz, MeOD) δ 158.50 (C), 155.39 (CH), 154.14 (C), 145.16 (C), 139.21 (C), 129.83 (C), 129.60 (CH), 128.10 (CH), 97.76 (C), 56.40 (CH 2 ), 54.64 (CH 2 ), 53.51 (CH 2 ), 52.56 (CH 2 ), 52.24 (CH 2 ), 43.79 (CH 2 ), 43.36 (CH 3 ), 19.97 (CH 3 ); MS (ES +ve) [M+1]*: 409.3, 431.2 (+Na); HRMS (ES +ve), C 22 H 33 N 8 [M+H] + : calculated 409.28227, found 409.282102.

1-[2-[4-(dimethylamino)-1-piperidyl]ethyl]-3-(p-tolyl)pyr azolo[3,4-d]pyramidin amine (109):

Purified by column cnromatography (Sv1eOH/PCy 10% - 10% MeOH with 20 drops of Hs aq, per 50 ml) to give a light brown solid (13 mg, 0,034 mmol, 32,7 %). 1 H MWIR (500 MHz, CDCia) S 8.35 (s, 1 H) ? 7.56 (d, J = 8.0, 2H>, 7.33 (d, J - 7.8, 2H), 5.68 (s, 2H), 4.54 (t, J - 7.0, 2H), 3,08 d, J = 11 ,8, 2H), 2.93 (t, J =7.0, 2H), 2.43 (s, 3.H), 2.41 (m, 1 H), 2.40 (s, 6H), 2.10 (del J = 11.8, 9.9, 2H), 1.84 (d, J = 12.3, 2H), 1.52 qd, J ? 12.1 , 3.7, 2H); ,3 C M (126 MHz, CDCij) 6 158.16 (C), 156.04 (CH), 154.8 (C), 144.69 (C), 139.38 (C), 130,60 (C), 130.32 (CH), 128.57 (CH), 98.70 (C), 62,72 (CH), 56.96 (GH 2 ), 52.86 (2x CH ? ), 45.13 (CH a ), 41.01 (2x CH 3 ), 27.71 (CH 2 ). 21.61 (CH 3 ); MS (ES -t-vej [M+1f : 380.2, 402.1 (+Na); HRM$ (ES +ve), C 2 ,H 30 7 [M* †l calculated 380.25572, found 380,255345,

1 2-I -(d iniet SamiiTsomethyS)-1 -pip© r!dyIlethyI]-3-<ip->to!yS)pyra2oi©[3,4- dJpyrimiilsr(-4-am!rie (112):

To a suspension of 2-[4~am!no-3-(p oly )pyrazGiQ[3,4-d]pyrimidin~1-y (100 mg, 0.374 mmol) in DCM (2 ml) was added N,N-Dimethyk1-pfp^ (1 eq. 53.2 mg, 0.374 mmol) and a drop of AcOH and the mixture allowed to stir for 10 mins. Sodium triacetoxyborohydride (79.3 mg, 0.374 mmol) was then added and the mixture ailowed to stir for 18 hours. The mixture was reduced in vacuo and purified, without a work up, by column chromatography MeOH/DCM (0-10 % - 10 % with 10 drops NH3 aq. per 100 ml) to give a tight orange solid (48 mg, 0.122 mmol, 32.6 %). 1H NMR (500 MHz, CDCI3) δ 8.30 (s, 1 H), 7.55 (d, J = 8.1, 2H), 7.32 (d, J = 7.8, 2H), 4.54 (t, J = 6.8, 2H), 3.04 (d, J = 11.6, 2H), 2.95 (t, J = 6.8, 2H), 2.57 (s, 6H), 2.41 (s, 3H), 2.11 (t, J = 10.9, 2H), 1.74 (d, J = 12.6, 2H), 1.63 (s, 2H), 1.26 (m, 3H); 13C NMR (126 MHz, CDC13) δ 158.06 (C), 155.40 (CH), 154.56 (C), 144.79 (C), 139.23 (C), 130.36 (C), 130.16 (CH), 128.40 (CH), 98.53 (C), 64.32 (CH2), 56.97 (CH2), 53.15 (CH2),

44.52 (CH2), 44.42 (CH3), 32.62 (CH), 30.14 (CH2), 21.44 (CH3); MS (ES +ve) [M+1]+: 394.3, 416.2 (+Na); HRMS (ES +ve), C22H32N7 [M+H]+: calculated 394.27137, found 394.271595. 1 -[2-[4-(2-dimethylaminoethyl)-1 -piperidyl]ethyl]-3-(p-tolyl)pyrazolo[3,4- djpyrimidin-4-amine (113):

To a suspension of 2-[4-amino-3-(p"tolyl)pyrazolo[3,4-d]pyrimidin-1-yl]acealdeh yde (100 mg, 0.374 mmol) in DCM (2 ml) was added dimethy!-(2-piperidin-4-yl-ethy!)-amine (1 eq. 58.4 mg, 0.374 mmol) and a drop of AcOH and the mixture allowed to stir for 10 mins. Sodium triacetoxyborohydride (79.3 mg, 0.374 mmol) was then added and the mixture allowed to stir for 18 hours. The mixture was reduced in vacuo and purified, without a work up, by column chromatography MeOH/DCM (0- 0 % - 10 % with 0-10 drops NH 3 aq per 100 ml) to give a light yellow solid (69.8 mg, 0.171 mmol, 45.8). 1 H NMR (500 MHz, MeOD) δ 8.25 (s, 1 H), 7.59 - 7.55 (m, 2H), 7.38 (d, J = 7.8, 2H), 4.58 (t, J 6.6, 2H), 3.14 (d, J = 11.8, 2H), 3.09 - 2.99 (m, 4H), 2.79 (s, 6H), 2.43 (s, 3H), 2,23 (t, J ^ 11.0, .2H), ,74 (d, J - 12.9, 2H), .66 - 1.57 (m, 2H), 1.44 - 1.36 (τη, 1 H), 1.27 (dd, = 21.1 11.8, 2H); 13 C NMR (126 MHz, ΜβΌΌ) δ 158.50 {C}, 155.45 (CH), 154.14 (C), 145,30 (C), 139.23 (C), 129.79 (C) r 129.60 (CH), 128.09 {CH}, 97.83 (C), 56.50 (CH 2 ) ! 55.70 {CH 2 ), 52.99 (2x CH 2 J : , 43.45 (Cl¼ 42.14 (2x CH,), 32.84 .(CH), 30.88 (2x QH 2 ), 30,64 (CH 2 ), 19,97 (GH 3 ); MS (ES +ve) [Μ+ψ. 408,1 ; H MS (ES +ve), C 23 H ¾ N ? [M+Hf: caScUiated 408,28702,. found 408,288812.

Compound fnterroecfiates for 021-030: To a solution of 1-i2,2 iieth0xyethyi}-3-sodo~ pyrazoSo 3 : 4-d]pyr!iT idin-4-afT!ine (100 rrig, 0,285 frirnol) in dfoxane/water (4,5 rn!/0,5 mi) was added either 1 H-pyrrolo ^-blpyridine-S-boronic acid pinscoi ester, 5- isoquinoiineboronlcacid, 3^4-dimeihoXyphenyiboronicacid, S-hydroxyphenylboiOnicacid or furan~3~boronicacid (1.5 eq., 0.397 mmol), potassium carbonate (1.5 eq., 54.8 mg, 0.397 mmol), triphen !phosphine (20 mol %, 20.8 mg) and palladium acetate (6 moi %, 4.5 mg) and the mixture heated in the microwave at. 120 °G for an hour. EtOAc (50 mi) and water (50 mi) were added to the mixture and the organic layer separated, washed with brine (50 mi), dried over gSO^ and concentrated in vacuo.

1 2,2-dt@th© ethy1)"3- 1H-^

amine;

Purified by column chromatography, eOH DCM (0-8 %) to give a white solid (93 mg, 0.258 mmol, 95.8 %). 1 H UMR (500 MHz, CDCI3) δ 9.53 (s, 1 H), 8.62 (d, J = 1.9, 1 hi), 8,41 (s, 1H), 8.24 (d, J = 2.0, 1 H), 7.45 (d, J 3.4, H), 8.62 (d, J = 3.5, 1 H), 6.29™ 5.86 (br. s, 2H), 5.14 (t, J = 5.7, 1 H ) , 4.62 (d, J = 5,7, 2H), 3,79 (dq, J - 9.4, 7,0, 2H), 3.55 (dq, J = 9.4, 7.0, 2H), 1.14 (t, J = 7.0, 6H); 13 C NMR (126 MHz, CDC!3) δ 156.75 (C), 154.58 (C), 153.4:2 (CH), 148.87 (C), 143..78 (C), 142.83 (CH), 128.83 (CH), 126.85 (CH), 121.30 (C), 120.46 (C), 101.80 (CH), 99.93 (CH), 98,47 (C), 02.1 (2.x CH 2 ), 49.33 (CH 2 ), 15.34 (2x CH 3 ); MS (ES +ve) [M+H] + : 368.2, 390.2 (+Na), (ES -ve) [M-H] " : 366.2; HRMS (ES +ve), C 18 H 22 N 7 02 (M+H) + : calculated 368.18295, found 368.18090.

1-(2,2-diet oxyethyl)-3-(6-quinolyl) razolo[3,4-dJpyrimidin-4-amine:

Purified by column chromatography, MeOH/DCM (0-5 %) to give a pale orange solid (86 mg, 0.227 mmol, 85.8 %). 1 H NMR (400 MHz, CDCI3) δ 9.39 (s, 1H), 8.59 (d, J = 6.0, 1Η), 8.47 (s, 1 H), 8.16 (d, J = 8.2, 1 H), 7.93 (dd, J = 7.1 , 1.2, 1 H), 7.84 (d, J = 6.0, 1H), 7.79 (dd, J = 8.2, 7.2, 1 H), 5.18 (t, J = 5.7, 1 H), 4.68 (d, J = 5.8, 2H), 3.83 (dq, J = 9.4, 7.0, 2H), 3.58 (dq, J = 9.4, 7.0, 2H), 1.21 - 1.13 (m, 6H); 13 C NMR (126 MHz,

CDCI3) δ 157.36 (C), 155.87 (CH), 154.77 (C), 152.99 (CH), 144.34 (CH), 141.21 (C), 134.32 (C), 132.29 (CH), 129.43 (CH), 129.32 (C), 128.98 (C), 127.01 (CH), 118.25 (CH), 99.92 (C), 99.80 (CH), 61.94 (2x CH 2 ), 49.18 (CH 2 ), 15.23 (2x CH 3 ); MS (ES +ve) [M+H] + : 379.2, 401.2 (+Na), (ES -ve) [M-H] ' : 377.2; HRMS (ES +ve), C 20 H 23 N 6 O 2 (M+H) + : calculated 379.18770, found 379.18660.

1-(2,2-diethoxyethyl)-3-(3,4-dimethox henyl)pyrazolo[3,4-d]pyrimidin-4-amine:

Purified by column chromatography, MeOH/DCM (0-2%) to give a pale yellow solid (97 mg, 0.251 mmol, 94.5%). 1 H NMR (500 MHz, CDCI3) δ 8.38 (s, 1H), 7.23 - 7.19 (m, 2H), 7.02 (d, J = 8.7, 1 H), 5.94 (s, 2H), 5.12 (t, J = 5.7, 1H), 4.57 (d, J = 5.8, 2H), 3.97 (d, J = 9.8, 6H), 3.78 (dq, J = 9.4, 7.0, 2H), 3.53 (dq, J = 9.4, 7.0, 2H), 1.13 (t, J = 7.0, 6H); ' X MR (126 MHz, CDCI3) δ 157,59 (C), 155.28 (CH), 154.78 .(C), 149.94 (G), 149.89 -(C), 144.70 (C), 125.68 (G), 120.81 (CH), 111.81 (CH), 111.56 (CH), 99.82 (CH) 98.32 (C). 61.81 (2x GH 2 ), 58.06 (2x CHa), 48,88 (CH 2 ), 15,19 (2x CH 3 ); MS (ES ÷ve) IM+Hf 388.2, (ES -ve [S I-Hf: 368.2; RMS (ES +y©), C^NeO* (M+H.) * : calculated 388.19793, found 388. 9820.

3 -am!no - 2, -diethaxy^

Purified by coiumn chromatography, MeQH/DGM (0-3%) to give a cream solid (8Q mg, 0.233 mmo!, 88.0 %). 5 H N» (500 MHz, CDC!3) δ 8,45 (s. 1 H) t 7.38 (t, J = 7.9, 1 H), 7.20 (d, J - 7.5, 1..H , 7.12 (s, 1 H), 8.95 (d, J - 8.1 , 1 H], 5,89 (s, 2H), 5,10 (t, J = 5,7, 1 H) i .57 (d 1 J ~ 5.7, 2H), 3.76 (it, J -■ 14.1 , 7.0, 2H), 3,52 (tt, J. = 14.1 , 7.0, 2H), 1.11 (t, J « 7,0, 6H); i5 C HBR (126 MHz, CDGS3) δ 157.10 (C), 155.86 (CH), 153.86 (C),

152.23 (C), 145,53 (C), 133.41 (C). 131.04 (GH), 120.31 (CH), 117.10 (CH), 115.15 (CH), 99.75 (GH), 97.78 (C), 62.16 (2x CH a ), 49.22 (CH 2 ), 5.17 (2x CH 3 ); IVIS (ES +ve) [M+Hj*: 344.2, 366.2 (+Ua), (ES -ve) [ -H] " : 342.2; HRMS (ES +ve), 0 !7 Η 22 5 0 3

(M*H)*: calculated 344/171 2, found 344.17000.

Purified by coiumn chromatography, MeOH/PGW (0-2 % ' ) to give a cream coloured solid, (66,8 mg, 0,211 mmol, 79.5 %), 1 H HMR (500 MHz, CDGI3) δ 8.38 <s, 1 H), 7.83 (dd, J - · 1.4, 0.9, . 1 H), 7.81 (t, J = 1.7, I N), 8.77 (dd, J = 1.8, 0.8, 1 H), 5.82 (s, 2H), 5.09 (t, J = 5.8, 1 H), 4.55 (d, J- 5.8, 2H), 3,76 (dq, J - 9.4, 7,0, 2H), 3,52 (dq, J = 9.4, 7.0, 2H), 1.11 (t, J = 7.0, 6H); 1J C NMR (126 MHz, CDCI3) δ 156.93 (C), 154.38 (C), 154.12 (CH), 144.66 (CH), 141.11 (CH), 136.98 (C), 118.52 (C), 110.29 (CH), 99.73 (CH), 98.61 (C), 61.92 (2x CH2), 49.04 (CH2), 15.17 (2x CH3); WIS (ES +ve) [M+H| + : 318.2, 340.2(+Na), 657.2 (2M+Na), (ES -ve) [M-H] " : 317.2; HRMS (ES +ve), C 15 H 2 oN 5 0 3 (M+H) + : calculated 318.15607, found 318. 5400.

1-(2,2-diethoxyethyl)-3-(2-phenylethyny])pyrazolo[3,4-d]p yrrmidin-4-amine:

To a solution of 1-(2,2-diethoxyethyl)-3-iodo-pyrazolo[3,4-d]pyrimidin-4-amin e (100 mg, 0.265 mmoi) in THF (5 ml) was added phenylacetylene (1.5 eq., 0.397mmol, 40.5 mg, 37.7 μΙ), triethylamine (1.5 eq., 0.397 mmol, 29.1 μΙ), palladium acetate (5 mol %, 4.5 mg), triphenylphosphine (20 mol %, 20.8 mg) and copper iodide (5 mol%, 2.5 mg). The mixture was heated conventially at 70°C for 2 hours. EtOAc and water were added to the mixture and the organic layer separated, dried over MgS0 4 and concentrated in vacuo. The crude product was purified by column chromatography, MeOH/DCM (0-2%) to give a light yellow solid (52 mg, 0.148 mmol, 55.9 %). H NMR (500 MHz, CDCI3) δ 8.90 - 8.35 (m, 1 H), 7.59 (dd, J = 7.7, 1.7, 2H), 7.45 - 7.36 (m, 3H), 6.20 (s, 2H), 5.09 (t, J = 5.6, 1 H), 4.53 (d, J = 5.7, 2H), 3.74 (dq, J = 9.3, 7.0, 2H), 3.50 (dq, J = 14.4, 7.2, 2H), 1.10 (t, J = 7.0, 6H); 3 C NMR (126 MHz, CDCI3) δ 153.88 (CH), 131.82 (2x CH), 129.58 (CH), 128.67 (2x CH), 121.45 (C), 99.81 (CH), 94.33 (C), 80.63 (C), 62.07 (2x CH 2 ), 49.41 (CH 2 ), 15.16 (2x CH 3 ); MS (ES +ve) [M+H]*: 352.2, 725.2 (2M +Na), (ES - ve) [M-H]-: 350.2; HRMS (ES +ve), C 19 H 22 N 5 0 2 (M+H) + : calculated 352.17680, found 352.17680. 1 -[2-[4-(dimethyiaminomethyl)-1 -piperidyl]ethyl]-3-(1 H-pyrrolo[2,3-b]pyridin-5- yl)pyrazolo[3,4-d]pyrimidin-4-amine (221 ):

80 mg, 0.183 mmol, of 1-(2,2-dsethQ yeihy!)-3-(1H-pyrrQlo[2,3--b]pyrid!n"5~

yl)pyf azolo[3,4-d pyrimidin~4~amine was added to a 20 ml microwave vial . 5 ra! of water was added foilowed by 5 ml of TFA and the mixture heated conventiaiiy at 100°C for an hour. The mixture was concentrated in vacuo to leave a light brown oil which was used without further purification 0.183 mmol of 2-[4-ammo~3~{1 H-pyrrolo|2,3-b3pyridSn-S~ yl)pyraTOlo 3,4-d]pyrimidin -yl]acetaldehycle was dissolved in 2 ml of DCM, N,M~ dimethyi-1-(4-p:iperidyi}mefhanamine (1.5 eq., 0.245 mmoi, 34.8 mg) was added followed by a drop of acetic acid. The mixture was allowed to stir for 10 rnins then sodium triaceto)fybQfOhydride (1 ,5 eq,, 0.245 mmol, 51.9 mg) added and the mixture allowed to stir for 2 hours. The mixture was concentrated in vacuo and the product purified by column chromatography, MeQ IDCM (5-10% then 10% with 5-20 drops of NHs aq. Per 100 ml) to give a light orange solid, (15.3 mg, 0.0385 mmol, 14.9 %). H HMR (500 MHz, MeOD) δ 8.S2 (s, 1Ή), 8,29 (d, J - 2.0, 1H), 8.28 (s, 1 H), 7.52 (d, J = 3.5, 1 H), 6.62 (d, J = 3.5, 1 H), 4.84 (t, J = 6,4, 2H), 3.25 (d, J = 1 .7, 2H), 3.13 (i, J - 6.3, 2H), 2.98 .{«_, J = 7.2, 2H) f 2.84 (s, 6H), 2.37 (t, J = 11 ,4, 2H), 1.88 (m, 1 H), 1.80 (d, J = 13,1 , 2H), 1.36 - 1.31 (m, 2H); 13 C UWR (126 MHz, UeOD) δ 161.79 (C). 158.65 (G), 155.54 (CH), 154.33 (G), 148.16 (G), 143.72 (C), 141.84 (GH), 128.87 (CH), 127.12 (CH), 120.73 (C) : 100.63 (CH), 98.19 (C), 62.67 (CH 2 ), 56.31 .(CH 2 ), 52.22 (2x CH 2 ), 43.41 (CH 2 , 42.72 (2x CH 3 ), 30.93 (CH), 28.39 (2x CH 2 ); MS (ES +ve) { +Hf: 420.2;

S (ES +ve), G 22 H 2S N 9 [M+Hf: calculated 420,25404, found 420.254249.

1~[2-[4-{cf l ffiethylarn s nome hy -pipe rid SJet Sj-S-ie- issiioiyllp razoioP^- djpyfimidin-4-aiTiIne (223):

60 mg, 0.159 mmol of 1-(2,2-diethoxyeihyl)-3-(6-quinolyl)pyrazolo[3,4-d]pyrimidin -4- amine was added to a 20 ml microwave vial. 5 ml of water was added followed by 5 ml of TFA and the mixture heated conventially at 100°C for an hour. The mixture was concentrated in vacuo to give a brown oil which was used without further purification. 0.159 mmo! of 2-[4-amino-3-(6-quinoiyl)pyrazoio[3,4-d]pyrimidin-1-yi]aceta ldehyde was dissolved in 2 m! of DCM. N,N-dimethyl-1-{4-piperidyl)methanamine (1.5 eq., 0.238 mmo!, 33.9 mg) was added followed by a drop of acetic acid and the mixture allowed to stir for 10 mins. Sodium triacetoxyborohydride (1.5 eq., 0.238 mmol, 50.4 mg) was added and the mixture allowed to stir for 17 hours. The mixture was concentrated in vacuo and purified by column chromatography, MeOH/DCM (5-10% then 20 drops NH 3 aq per 100 m!) to give a fluorescent yellow coloured solid, 48.9 mg, 0.122 mmol, 70.6 %. 1 H NWIR (500 MHz, MeOD) δ 9.37 (s, 1 H), 8.47 (d, J = 6.0, 1 H), 8.31 (m, 2H), 7.99 {dd, J = 7.1 , 1.2, 1 H), 7.90 - 7.84 (m, 2H), 4.65 (t, J = 6.7, 2H), 3.09 (d, J = 11.7, 2H), 2.99 (t, J = 6.7, 2H), 2.28 (s, 6H), 2.27 (m, 2H), 2.18 - 2.11 (m, 2H), 1.75 (d, J = 12.4, 2H), 1.57 (ddd, J = 11.3, 7.4, 3.9, 1 H), 1.24 - 1.12 (m, 2H); 3 C N R (126 MHz, MeOD) δ 158.26 (C), 155.67 (CH), 154.06 (C), 152.47 (CH), 142.47 (CH), 141.58 (C), 134.67 (C), 132.89 (CH), 129.35 (CH), 129.21 (c), 129.1 1 (C), 127.33 (CH), 118.82 (CH), 99.55 (C), 65.35 (CH 2 ), 56.79 (CH 2 ), 53.22 (2x CH 2 ), 44.34 (2x CH 3 ), 44.05 (CH 2 ), 33.06 (CH), 30.04 (2x CH 2 ); MS (ES +ve) [M+H] + : 431.2; HRWIS (ES +ve), C 24 H 3 oN e [M+H] + :

calculated 431.25879, found 431.258695.

3-(3,4-dimethoxyphenyi)-1-[2-E4-(dimethylaminomethyI)-1-p iperidyi]ethyI] pyrazolo[3,4-d]pyrimidin-4-amine (224):

70 mg, 0.181 mmq! of 1-(2,2-dieihoxyethyi)-3-{3 -dimethoxyphen

d]pyrimidin-4-amlne was added to a 20 mi microwave vial. 5 mi of water was added followed by 5 ml of TFA and the mixture heated conventialiy at 100°C for an hour. The mixture was concentrated In vacuo to leav a Sight brown oil which was used without further purification. 0.181 mrhol of ^-^-amino-S-fS^-dimethoxyphenylJpyrazoloiS^- d]pyrimidin-1-yi]acetald:ehyde was dissolved in 4 mi of DCM. j N-dimefhyl- - - piperidyijmeihanamine (1 ,5 eq., 0.271 minoi, 38,5 mg) was added followed b a drop of acetic acid and the mixture allowed to stir for 10 mins. Sodium triacetoxyborohydride (1.5 eq., 0.271 mmol, 57.4 mg) was added and the mixture allowed to stir for 17 hours. The mixture was concentrated in vacuo and the product purified by column

chromatography, eOH/DC (5-10 % then 0 drops of NH 3 a per 100 ml) to give a light yellow solid (14.5 mg. 0.033 mmol, 20.8 %). f H NMR {500 MHz, MeOD) δ 8.26 (s, 1 H), 7,2.7 (s, 1 H), 7,25 (d, J ~ 8.2, ΪΗ), 7, 14 (d, J ~ 8.2, 1H), 4.63 {% J = 6.3, 2H), 3.91 (s, 6H), 3.18 (s, 2H), 2.98 (d, J = 7.2, 2H), 2.85 (s, 6H), 2.45 (m, 2H), 1 ,9.2 (m, 1 H), 1 ,83 (d, J = 13.2, 2H), 1.35 (dd, J = 22.2, 11.5, 2H>; 15 C M R (1 . 26 MHz, MeOD) δ 158.58 (C), 155.51 (CH). 154.22 (G), 150.22 (C), 149.67 (C), 145.44 (C), 125.19 (C), 120.91 (CH), 11 .94 (CH), 111.73 (CH), 97,82 (C), 62.49 (CH ? ), 55.18 (CH 2 ). 55.13 (2x CH 3 ), 52.16 (2x CH 2 ), 43.08 (GH £ ), 42.69 (2x CH 3 ), 30.70 (CH), 28.13 (2x CH 2 ); MS (ES. +ve) [M ' +Hf- 440.2; HRIVSS (ES +ve), C ¾ H 33 N 7 Q 2 [M+Hf : calculated 440.26902, found 440.268379.

d] py r i m idl n P henoi (22S :

60 mg, 0.175 mmol of 3-[4-amino-1-(2,2-diethoxyethyl)pyrazolo[3,4-d]pyrimidin-3- yl]phenol was added to a 20 ml microwave vial. 5 ml of water was added followed by 5 ml of TFA and the mixture heated conventially at 100°C for an hour. The mixture was concentrated in vacuo to leave a light brown oil which was used without further purification. 0.175 mmol of 2-[4-amino-3-(3-hydroxypheny!)pyrazolo[3,4-d]pyrimidin-1- yl]acetaldehyde was dissolved in 4 ml of DCM. N,N-dimethy!-1-(4- piperidyl)methanamine (1.5 eq., 0.262 mmol, 37.2 mg) was added followed by a drop of acetic acid and the mixture allowed to stir for 10 mins. Sodium triacetoxyborohydride (1.5 eq., 0.262 mmol, 55.5 mg) was added and the mixture allowed to stir for 20 hours. The mixture was concentrated in vacuo and the product purified by column

chromatography, MeOH/DCM (10 % then 0-20 drops of NH 3 aq per 100 ml) to give a dark orange solid (5.5 mg, 0.0139 mmol, 8.0 %). 1 H N R (500 MHz, MeOD) δ 8.26 (s, 1 H), 7.39 (t, J = 7.9, 1 H), 7.17 - 7.09 (m, 2H), 6.95 (dd, J = 7.8, 2.1 , 1 H), 4.57 (t, J = 6.7, 2H), 3.08 (d, J = 11.7, 2H), 2.96 (t, J = 6.7, 2H), 2.47 (m, 8H), 2.16 (t, J = 1 1.0, 2H), 1.74 (d, J = 12.9, 2H), 1.64 (m, 1 H), 1.21 (dd, J = 21.1 , 12.0, 2H); 13 C NMR (126 MHz, MeOD) δ 158.44 (C), 158.06 (C), 155.42 (CH), 154.06 (C), 145.17 (C), 133.97 (C), 130.16 (CH), 119.09 (CH), 1 15.96 (CH), 114.92 (CH), 97.73 (C), 64.57 (CH 2 ), 56.66 (CH 2 ), 52.78 (2x CH 2 ), 43.83 (2x CH 3 ), 43.77 (CH 2 ), 32.46 (CH), 29.58 (2x CH 2 ). MS (ES +ve) [M+H] + ; 396.4; HRMS (ES +ve), C 21 H 29 N 7 0 [M+H] + : calculated 396.24281 , found 396.242971.

1-[2-[4-(dimethylaminomethyl)"1-piperidyl]ethyI]-3-(3-fur yi)pyrazolo[3,4- d]pyrimidin-4-amine (226):

52 mg, 0.16 mmoS of i-iS^-diethox^

was added to a 20 mi microwave ial. 5 mi of water was added followed by 5 ml of TFA and the mixture heated conventjaliy at 1 D0 Q G for an hour. The mixture was

concentrated Jn vacuoiQ leave a light brown oil which was used without further purification. 0.164 mmo of .2~[4-ΒΓπτηο~3-{3 -furyf )pyraz0io[3,4-d]py rimidin-t- yljacetaldehyde was dissolved In 4 ml of DOM. , -dirn8ihyl-1-{4- piperidy methananTine (1.5 eq,, 0.246 mmol, 35.0 mg) was added followed by a drop of acetic acid and the mixture allowed, to stir for 0 mins. Sodium triacetoxy orohydride (1 ,5 eq,, 0,246 mmol, 52.1 mg) was added and the mixture allowed to stir over the weekend, The mixture was concentrated in vacuo and the product purified by colum chromatography, SvleOH/DCy (5-10 % then 10-20 drops of Ha a per 1Q0 mi) to give a dark golden brown solid (45.7 mg, 0.124 fnmoJ, 75.5 %). i HMR (500 MHz, MeQD) δ 8.23 (s, 1 H), 7,96 (dd, J = 1.4, 0.9, 1*1), 7.71 <t, J - 1 ,7, 1H), 6.82 (dd, J ~ 1.8, 0.8, 1H), 4.53 (t, J = S.7, 2H), 3.08· (d, J = 11,7, 2H), 2.94 (t, J = 6,7, 2H), 2.62 (d, J - 6,6, 2H), 2,57 (s, 6H), 2,16 (dd, J = 11.8, 10 -0, 2H), ; 172 (d, J = 12,9, 2H), 1.68 (m, 1 H), 1.26 - 1.18 (m, 2H); ¾ NM.R (126 MHz, eOD) δ 158.57 (O), 115.45 (CH), 1 S3.98 (G), 144.34 (CH), 141.47 (CH), 137.22 (C), 18.30 (C), 109.73 (CH), 98.17 (C), 63.97 (C ¼ 56,59 <CH 2 ), 52.62 (2x CH 2 ), 43.73 (CH 2 ), 43.43 (2x CH 3 ), 32.02 (CH), 29,27 (2x CH 2 ); MS (ES +V8) [SVl÷Hf: 370.2; MMS (ES +ve), C 1S H 27 M7© [ +Hf : caicuiated 370.22716, found 370.227G49.

1 - 2- 4-(dsrrs t ySaminQmethy!)-1 -pipe

J]pyrim!dim<-4"amifie (230):

50 mg, 0.142 mmol of 1-(2,2-diethoxyethyl)-3-(2-phenyleihynyl)pyrazoIo[3,4-d]pyri midtn- 4-amine was added to a 20 ml microwave vial. 5 ml of water was added followed by 5 ml of TFA and the mixture heated conventially at 100°C for an hour. The mixture was concentrated in vacuo to leave a dark brown oil which was used without further purification. 0.142 mmol of 2-[4-amino-3-(2-phenylethynyl)pyrazoio[3,4-d]pyrimidin-1- yl]acetaldehyde was suspended in 4 ml of DCM. N,N-dimethyl-1 -(4- piperidyl)methanamine (1.5 eq., 0.213 mmol, 30.3 mg) was added followed by a drop of acetic acid and the mixture allowed to stir for 10 mins. Sodium triacetoxyborohydride (1.5 eq., 0.213 mmol, 45.1 mg) was added and the mixture allowed to stir for an hour. The mixture was concentrated in vacuo and the product purified by column

chromatography, MeOH/DCM (5-10 % then 5-10 drops of NH 3 aq per 100 ml) to give a dark orange solid, (23.4 mg, 0.058 mmol, 40.7 %). 1 H NMR (500 MHz, MeOD) δ 8.23 (s, 1 H), 7.69 - 7.61 (m, 2H), 7.46 - 7.40 (m, 3H), 4.51 (t, J = 6.7, 2H), 3.02 (d, J = 1 1.7, 2H), 2.90 (t, J = 6.7, 2H), 2.29 (s, 6H), 2.28 - 2.25 (m, 2H), 2.10 (td, J = 11.8, 2.3, 2H), 1.71 (d, J = 12.8, 2H), 1.54 (dtd, J = 14.6, 7.5, 3.7, 1 H), 1.15 (qd, J = 12.5, 3.7, 2H); 3 C NMR (126 MHz, MeOD) δ 158.22 (C), 156.07 (CH), 153.13 (C), 131.48 (CH x2), 29.25 (CH), 128.38 (CH x2), 126.72 (C), 121.47 (C), 100.98 (C), 93.76 (C), 79.88 (C), 65.21 (CH 2 ), 56.68 (CH 2 ), 52.96 (CH Z x2), 44.27 (CH 3 x2), 44.22 (CH 2 ), 32.94 (CH), 29.92 (CH 2 x2); MS (ES +ve) [M+Hj + : 404.3; HRMS (ES +ve), C 23 H 29 N 7 [M+H] + : calculated 404.24790, found 404.247888.

1 -[2-[4-(dimethylaminomethyl)-1-piperidyt]ethyl]-3-iodo-pyraz olo[3,4-d]pyrim 4-amine (232):

75 rtfg, 0.199 mmol of 1-(2,2-dieihoxyethyl}-3-iGdo~pyrazolo^

was added to a 10 m! microwave tube. 2.5 mi of water and 2,5 ml of TFA were added and the mixture heated to 100°C for an hour, The mixture was concentrated in vacuo to give a white solid which was used without further purification. .0,199 rnmol of 2-(4- amino-3-!odo-pyrazolo[3 l 4-d]pyrim}d!n-1~y!)a was suspended in 3 mi of

DCM. N,N-dimethyi-1-(4-p!peridyl)methanarnine {1.5 eq,, 0.290 mmol, 42.2 nig} was added foiiowed b a drop of acetic acid and the mixture allowed to stir for 10 mins. Sodium triacetoxyborohydride (1 ,5 eq., 0.299 mmol, 63,4 mg) was added and the mixture allowed to stir for 17 hours. The mixture was concentrated in vacuo and purified by column chromaiography, MeOH/DCM (0-10 % then 5-15 drops of NH 3 aq, per 100 ml) to give a Sight yello coloured solid {63,8 mg, 0.148 mmol, 74.5 %), 1 H MS¾f (600 MHz, M&OD) δ 8.20 (s, 1 H), 4,49 (t, J ~ 6.7, 2H), 3,01 (d, J - 11.7, 2H), 2.87 (t, J = 6.7, 2H), 2,28 (s, 6H), 2,27 (d, 2H), 2,10 (Id, J = 1 .8, 2,3, 2H), 1.72 (d, 13,0, 2B) ( 1.55 (ddt, J = 15.0, 7.6, 3.8. 1H), 1.15 (qd, J = 12.3, 3.7, 2H); % UMR (128 MHz, MeOD) δ 158,05 (C), 155.63 (CH), 153.58 .(C), 103,66 (C), 86.95 (G), 65.27 (GH 2 ), 58,73 (CHs), 52.96 (2x CH 2 ), 44,30 (2x CH 3 } , 44.15 {0H 3 ), 32,97 (GH>, 30.09 (2x CH 2 >; MS (ES +ve) [M+H : 430.2, 1~[2H.4 ^ (dim&t y.amirio3^

yS)pyrazofo{3,4"d]pyrimidsri-4"ani!ne (402);

40 mg, 0.109 mmoi of 1-(2,2-diethoxyethyl)-3-(1 H-pyrro[o[2,3-b]pyridin-5- yl)pyrazolo[3,4-d]pyrimidin-4-amine was added to a 10 ml microwave tube. 2.5 ml of water and 2.5 ml of TFA were then added and the mixture heated to 100 °C for 30 mins in the microwave. The solvents were removed in vacuo to give a dark brown oil which was used without further purification. 0.109 mmol of 2-[4-amino-3-(1 H-pyrrolo[2,3- b]pyridin-5-yl)pyrazolo[3,4-d]pyrimidin-1-yl]acetaidehyde was dissolved in 3 ml of THF. N,N-dimethy!piperidrne-4~amine (1.5 eq., 0.1635 mmol, 20.9 mg) was added and the mixture was allowed to stir for 5 mins. Sodium triacetoxyborohydride (1.5 eq. ( 0.1635 mmol, 34.7 mg) was then added and the mixture allowed to stir for an hour. The reaction mixture was concentrated in vacuo and purified by column chromatography, MeOH/DCM (10 % then 5-40 drops of NH 3 aq. per 50 ml) to give a light yellow solid (26.9 mg, 0.0664 mmol, 60.9 %). H N R (500 MHz, MeOD) δ 8.50 (d, J = 1.9, 1 H), 8.28 (d, J = 2.0, 1 H), 8.27 (s, 1 H), 7.51 (d, J = 3.5, H), 6.62 (d, J = 3.5, 1 H), 4.56 (t, J = 6.4, 2H), 3.19 (d, J = 12.1 , 2H), 3.06 (tt, J = 12.0, 3.9, 1 H), 2.97 (t, J = 6.4, 2H), 2.77 (s, 6H), 2.18 (t, J = 1 1.1 , 2H), 2.00 (d, J - 12.4, 2H), 1.58 (qd, J = 12.3, 3.9, 2H); 13 C NMR (126 MHz, MeOD) δ 158.64 (C), 155.49 (CH), 154.32 (C), 148.16 (C), 143.50 (C), 141.84 (CH), 128.66 (CH), 127.12 (CH), 120.78 (C), 120.73 (C), 100.63 (CH), 98.08 (C), 63.37 (CH), 55.82 (CH 2 ), 51.54 (2xCH 2 ), 44.1 (CH 2 ), 39.16 (2xCH 3 ), 26.18 (2xCH 2 ); MS (ES +ve) (M+H) + : 406.6; HR S (ES +ve), C 21 H 28 N9 (M+H) + : calculated 406.24622, found 406.24490.

1-[2-[4-(dimethylamino)-1 -piperidyl]ethyl]-3-iodo-pyrazolo[3,4-d]pyrimidin-4- amine:

150 mg, 0,398 mmol, of 1-{2,2-diethQxyethyi)-3-iQdo-pyra

was added to a 10 rn! microwave tube. 2.5 ml of water and 2.5 mi of TFA were added and the mixture heated to 100°C for an hour. The mixture was CQncentrated in vacuo to give a white solid which was used without further purification. 0.398 mmol of 2~{4- amino-3-iodo-pyraEolQi3,4-dlpyrimidin~1-yf}acet was suspended in 3 mi of

DG . ! J,N-dimethylpiperidine-4-amine (1.5 eq,, 0,598 mmoi, 76.8 mg) was added followed, by a drop of acetic acid and the mixture allowed to stir fo 10 mins. Sodium tnacetoxyborohydnde (1.5 eq.., 0.598 mmol, 126.8 mg)-was added and the mixture allowed to stir for 17 hours overnight. The mixture was concentrated in vacuo and the product purified b column chromatography, MeOH/DGM (0-10. % then 5-20 drops of NH 3 acf. per 100 ml) to give a light orange/brown solid (163.8 mg, 0,405 nimol, 99.1 %). ¼N¾S . (500 MHz, MeOD) 5 8.22 (s, 1H), 4.49 (t, J ~ 6,4, 2H), 3.32 (s, 3H), 3,15 (d, .J * 12.1 , 2H), 2,96 ddd, J ~ 16.0, 8.0, 4.0, 1 H), 2.91 (t, J ' = 8.4, 2H), 2.72 (s, 8H), 2.15 (td, J = 12.0, 2.0, 2H), 2.04 - 1.96 (m, 2H), 1.54 (qd, = 12.2, 3.9, 2H); i G HMR (126 MHz, MeOD) δ 158,07 (C), 155.87 (CH), 153.70 (C), 103.59 (C), 86,97 (C), 63.18 (CH), 55.86 (CH 2 ), 51 ,38 (2x CH*), 44.37 (CH 2 ), 39.31 (2x.CH 3 ), 28.42 (2 CH 2 ); MS (ES +ve

[M+Hf: 418,2. ier -butyi N-|4-[4-am!no-1 -i2-[4-(climetl ylaminomethyl)-1 - plperl dy ] ©tbyljpy ra^olo py rim M \ n-3 -y I] ~2~methoxy~pheny carbamate (S03) :

To a solution of l -^-^dimethylaminomethylJ-l -piperidylJethylj-S-iodo-pyrazolo ^- djpyrimidin-4-amine (50 mg, 0.1 165 mmol) in dioxane/water (4.5 ml/0.5 ml) was added [4-(tert-butoxycarbony!amino)~3-methoxy-phenyl]boronic acid (1 .5 eq. , 46.7 mg, 0.175 mmol), potassium carbonate (1 .5 eq., 24.2 mg, 0.175 mmol) and triphenylphosphine (20 mol %, 9.2 mg) followed by palladium acetate (5 mol %) and the mixture heated in the microwave at 120 °C for 45 mins. EtOAc (50 ml) and water (50 ml) were added to the mixture and the organic layer separated. The aqueous layer was washed with EtOAc (20 ml, x3) and the organics combined dried over MgS0 4 and concentrated in vacuo. The crude product was purified by column chromatography, MeOH/DCM (0-10 % then 5-20 drops of NH 3 aq. per 100 ml) to give a dark brown solid (36.5 mg, 0.0696 mmol, 59.7 %). H N R (500 MHz, MeOD) δ 8.27 (s, 1 H), 8.08 (d, J = 8.2, 1 H), 7.30 (d, J = 1 .8, 1 H), 7.26 (dd, J = 8.2, 1.8, 1 H), 4.58 (t, J = 6.8, 2H), 3.98 (s, 3H), 3.08 (d, J - 1 1 .7, 2H), 2.96 (t, J = 6.8, 2H), 2.34 (s, 6H), 2.32 (d, J = 7.2, 2H), 2.16 (dd, J = 1 1.8, 9.6, 2H), 1 .76 (d, J = 12.3, 2H), 1.63 - 1 .58 (m, 1 H), 1.57 (s, 9H), 1 .24 - 1 .16 (m, 2H); 13 C NMR (126 MHz, MeOD) δ 158.52 (C), 155.40 (CH), 154.07 (C), 153.46 (C), 149.24 (C), 145.04 (C), 128.77 (C), 127.39 (C), 120.43 (CH), 1 19.56 (CH), 1 10.29 (CH), 97.78 (C), 80.18 (C), 65.19 (CH 2 ), 56.76 (CH 2 ), 55.08 (CH 3 ), 52.96 (2x CH 2 ), 44.24 (2x CH 3 ), 43.79 (CH 2 ), 32.92 (CH), 29.89 (2x CH 2 ), 27.22 (3x CH 3 ); MS (ES +ve) [M+H] + : 525.4, (ES - ve) [M-H] " : 523.3; HRMS (ES +ve), C 27 H 4 i N 8 0 3 (M+H) + : calculated 525.32961 , found 525.32890. tert-butyl N-[4-[4-amino-1 -[2-[4-(dimethylamino)-1 -piperidyi]ethyi]pyrazolo[3,4- d] pyri m id in-3-y l]-2-methoxy-p henyl]ca rbamate (506) :

To a solution of 1 ~[2-[4-{dimei yiamino} » 1 ^ piper!dyi]eihyi]-3-iodo-pyrazoio[3,4- d]pyr!midin~4~amine (50 mg, 0.1205 mmoi) in dioxane/water (4.5 m!/0.5 ml) was added [4~(terH3utoxycarbonylamino}-3-^ acid (1.5 eq., 48.3 mg, 0.181 mmoi). potassium carbonate (1.5 eq., 25.0 mg, 0.181 mmoi) and triphenyfpbosphine (20 mol %, 9.5 mg) foflowed by palladium acetate (5 mol %) and the mixture heated in the microwave at 120 °C for an hour. EtOAc (50 mi) and water (50 mi) were added to the mixture and the organic layer separated. The aqueous layer was washed with EiOAc (20 mi, x2) and the organics combined dried over MgS0 4 and concentrated in vacuo, The crude product was purified by column chromatography, MeOH/DCM (0-10 % then 5-20 drops of NH 3 aq. per 100 mi) to give a light brown solid (23.1 mg. 0.0453 mmoi, 37.8 %},. *.H HMR (500 MHz, MeOD) 5 8.27 (s, 1 H), 8.08 d, J ~ 8.2, 1 H), 7.30 (d, J ~ 1 ,8, 1 ' HJ, 7.26 (dd, ~ 8.2, 1 ,9, 1 H), 4.56 ( - 6J, 2 . H), 3.98 (s, 3H), 3,14 (d, J~ 1 .9, 2H), 2.94 (t, J ~ 6.7, 2H), 2.39 (m, 7H), 2.14 (dd, J ~ 12.0, 0.0, 2H . ), 1.90 (d, J " 12.5, 2.H), 1.57 (s, 9H). 1,49 (qd, J - 12.1 , . 3.6, 2H); 13 C NJIR (126 fVIH , MeOD) 5 158.53 (C), 155.40 (CH), 154.12 (C), 153,46 (C), 149,24 (C), 145.02 (C), 128.78 (C), 127,38 (C), 120.43 (CH), 119.58 (CH), 10.28 (CH), 97.73 (C), 80.18 (C), 62.2 (CH), 58.22 (GH 2 ) f 55,07 (CH 3 ), 52,20 (2x CH 2 ), 44.00 (CH 2 ), 40.06 (2x,CH 3 ), 27.24 (2 GH 2 ), 27.21 (3x CH 3 ); MS (ES - ve) [iVH-H ; 51 .3; HRM& (ES ÷ve), G^HssNsQa [fVl+Hf: calculated 511.31396, found 511 ,3151,

3-iodo-1 -[2-(4-pyrrol ln-1 -yi-t- iperldy

300 mg, 0.8646 mmol of 1-(1 ,3-dioxo!an-2-ylmethyl)-3-iodo-pyrazolo[3,4-d]pyrimidin-4- amine was added to a 10 ml microwave tube. 2.5 ml of water and 2.5 ml of TFA were added and the mixture heated to 100°C for 3 hours. The product was concentrated in vacuo and used without further purification. 0.432 mmol of 2-(4-amino-3-iodo- pyrazoio[3,4-d]pyrimidin-1-yl)acetaldehyde was suspended in 3 ml of DCM. 4-(1- pyrrolidinyl)piperidine (1.5 eq., 0.648 mmol, 99.9 mg) was added followed by a drop of acetic acid and the mixture aiiowed to stir for 10 mins. Sodium triacetoxyborohydride (1.5 eq., 0.648 mmoi, 137.3 mg) was added and the mixture allowed to stir for 17 hours The mixture was concentrated in vacuo and purified by column chromatography,

MeOH/DCM (0-10 %) to give a green/brown solid, (183.1 mg, 0.415 mmol, 96.1 %). 1 H NMR (500 MHz, MeOD) δ 8.22 (s, 1 H), 4.50 (t, J = 6.3, 2H), 3.34 (m, 4H), 3.12 (d, J = 12.0, 2H), 3.06 (dd, J = 13.9, 9.7, 1 H), 2.90 (t, J = 6.3, 2H), 2.21 - 2.00 (m, 8H), .52 (td, J = 12.1 , 8.1 , 2H); MS (ES +ve) (M+H) + : 442.2

3-iodo-1-[2-[4-(1^iperidyl)-1-piperidyl]ethyl3pyrazolo[3, 4-d3pyrimidin-4-amine:

300 mg, 0.8646 mmol of l-dS-dioxolan^-ylmeth -S-iodo-pyrazolotS^-dlpyrimidin^- amine was added to a 10 ml microwave tube. 2.5 ml of water and 2.5 ml of TFA were added and the mixture heated to 100°C for 3 hours. The product was concentrated in vacuo and used without further purification. 0.432 mmoi of 2-(4-amino-3-iodo- pyrazolop.^cJlpyFimidin-l-y acetaSdehyde was suspended in 3 mi of. DCM. 1 ,4- blpiperidine (1.5 eq., 0.848 mmol, 108.9 mg) was added followed by a drop of acetic acid and the mixture allowed to stir for 10 rains. Sodium triacetoxybOro hydride (1 ,5 eq., 0,648 mmol, 137.3 mg) was added and th mixture allowed to stir for 1 hours. The mixture was concentrated in vacuo and purified by colum chromatography,

MeOH/DCM (0-10 %) to give a dark green/brown i hick oil, (191 ,6 rag, 0.421 mmol, 97.5%). Ή UMR (601 MHz, DivTSO) 5 8.21 (s, 1 H). 4.33 (t, J - 6,6, 2H), 2.85 (broad m, ), 1.98 (m, 2K), 1.54 (broad m, 0H>; S (ES +ve) ( +H} + : 456.2 tert-butyl

d3py¾'i^Win~3~ S]-2-meihoxy- !ie 18):

To a solution of 3-iodo-1-[2-(4-pyrrGHdin-1-y1-1-piperi^

4-amine £50 mg, 0.11:3 mmol) in dioxane/water (4,5 ml/0.5 ml) was added |4*(tert- butoxycarbonylamino)-3-methoxy-phenyl]boronic acid (1.5 eq., 45.4 mg, 0.170 rnmoi), potassium carbonate (1.5 eq., 23,5 mg, 0.170 mmol) and iriphenyiphosphine (20 rnoi %, 8.9 mg) followed by palladium acetate (5 mo! %) and the mixture heated in the microwave at 120 °C for an hour. EtQAc (50 mi) and water (50 ml) were added to the mixture and the organic layer separated. The aqueous layer was washed ith EtQAc (20 ml, x2) and the organics combined and washed with brine then dried over IVlgS0 and concentrated in vacuo. The crude product was purified by column chromatography, MeQH/DCM (0-10 % then 5-25 drops of NH 3 aq. per 100 mi) to give a light orange solid (18.69 mg, 31.12 pmoi, 27,5 %). NM (500 IvlHz, MeOD) δ 8,27 (s, 1 H), 8,08 (d, J = 8.2, 1 H), 7,30 (d, J = 1.8, 1 H), 7.25 (dd, J = 8.2, 1.8, 1H), 4.58 (t, J = 8.8, 2H), 3.98 (s, 3H), 3,12 (d, J - 12.0, 2H) :( 2.94 (m, 8H), 2.53 (m, IK), 2.15 (t, J = 11.1 , 2H), 2.01 (d, J = 12.2, 2H), 1.93 (dd, J - 8.3, 5.0, 4H), 1.57 (s, 9H), 1.55 - 1.48 (m, 2H); 3 C NMR (126 MHz, MeOD) δ 158.52 (C), 155.40 (CH), 154.15 (C), 153.45 (C), 149.24 (C), 145.00 (C), 128.80 (C), 127.36 (C), 120.43 (CH), 1 19.55 (CH), 1 10.27 (CH), 97.69 (C), 80.19 (C), 62.00 (CH), 56.16 (CH 2 ), 55.08 (CH 3 ), 51.62 (2x CH 2 ), 51.22 (2x CH 2 ), 44.01 (CH 2 ), 29.65 (2x CH 2 ), 27.22 (3x CH 3 ), 22.49 (2x CH 2 ); MS (ES +ve) [M+H] + : 537.4, (ES -ve) [M-H] " : 535.3; HRMS (ES +ve), C 28 H 4 oN 8 0 3 [M+H] + : calculated 537.32961 , found 537.3281. tert-butyl N-[4-[4-amino-1 -[2-[4-(1 -piperidyi)-1 -piperidyl]ethyl]pyrazolo[3,4- d]pyrimidin-3-yl]-2-methoxy-phe 19):

To a solution of 3-iodo-1-[2-[4-(1-piperidyl)-1-piperidyl]ethyl]pyrazolo[3,4- d]pyrimidin-4- amine (50 mg, 0. 10 mmol) in dioxane/water (4.5 ml/0.5 ml) was added [4-(tert- butoxycarbonylamino)-3-methoxy-phenyl]boronic acid (1.5 eq., 44.0 mg, 0.165 mmol), potassium carbonate (1.5 eq., 22.8 mg, 0.165 mmol) and triphenylphosphine (20 mol %, 5.8 mg) followed by palladium acetate (5 mol %) and the mixture heated in the microwave at 120 °C for an hour. EtOAc (50 ml) and water (50 ml) were added to the mixture and the organic layer separated. The aqueous layer was washed with EtOAc (20 ml, x2) and the organics combined, dried over MgS0 4 and concentrated in vacuo. The crude product was purified by column chromatography, MeOH/DCM (0-10 % then 5-15 drops of NH 3 aq. per 100 ml) to give a light brown solid (12.3 mg, 22.35 Mmol, 20.3 %). 1 H NMR (500 MHz, MeOD) δ 8.27 (s, 1 H), 8.08 (d, J = 8.2, 1 H), 7.30 (d, J = 1.7,

1 H), 7.26 (dd, J = 8.2, 1.8, 1 H), 4.55 (t, J = 6.6, 2H), 3.96 (s, 3H), 3.15 (d, J = 1 1.8, 2H), 2.93 (t, J = 6.6, 2H), 2.74 (br. s, 4H), 2.51 (br. s, 1 H), 2.13 (t, J = 11.1 , 2H), 1.91 (d, J = 12,0, 2H), 1.72 - 1.62 (m, 4.H), S§ .(m, 13H); 3 C NM (†26 MHz, MeGD S 158,52 (C), 155.40 (01-0, 154.13 (C), 153,45 (0), 149.24 (C), 145.02 (0), 128,79 (G), 127.37 (C), 120,43 (CH), 118,56 (CH), 110.28 (CH), 97.73 (C) : 80.19 (C), 62.69 (OH}, 56.21 (CH 2 ), 55.08 (CHsJ, 52.43 (2x CH 2 ), 49,83 (2x CH 2 ), 44.00 (CH?), 27.22 (3x CH 3 ) ; 26 78 (2x GH 2 ), 24..8S (2x : CH 2 ), 23.34 (CH ¾ );; MS (ES +ye) +H| * : 65 .2; HRMS (ES -f-vej, C S9 H4 2 B Q 3 [M+Hf: calculated 551.34526, found 551.3469.

Phenyl 2 m©th©xy~ -( , ,5,5-i^

carbamate:

To a solution of 4-Amino-3-methoxybenzeneboronic aeid, pinaco! esier (150 mg, 0.602 mmol) in DCM (2 ml) was added triethy!amine (1.2 eq,, 0,722 mmol, 73,1 mg, 100,76 μ{) followed by phenyl chloroformaie ( ,2 eq., 0.722 mmol, 12.6 mg, 90.25 pi) and the mixture allowed to stir for 20 hours. Water was added to the mixture and the organic layer separated, dried over MgSQ 4 and concentrated in vacuo. The product was purified by column chromatography, (100% DCM) to give a clear white solid (190.8 mg t 0,517 mmol, 85.9 %) *H HMR (500 MHz, CDCiS) δ 8,18 (d, J= 7.6, 1H), 7.74 (s, 1H), 7.49 (dd, J = 8 0. 1.1. 1H} : 7.46 - 7.40 (m, 2H), 7.33 (d, J = 1.0, 1H), 7.29 - 7,20 (m, 3H), 3,99 (s, 3H), 138 (s, 12H); 13 C UMR (126 MHz, CDCI3) 5 151 ,36 (C), 150.61 (G), 146.98 (C), 129.97 (C), 129.41 (2x GH), 128.61 (CH), 125.88 (CH), 125.05 (C), 121 ,74 (2x CH), 117.30 (CH), 1 15,41 (CH), 83.80 (2x C), 55,96 (CH 3 ), 24.92 {4x CH 3 ); MS (ES ÷ve) pVHHf: 370,5, 392 5 (+Na).

H~[2-methoxy-4^4,4,S J S4etramethy!-1

foutarsamSde:

To a solution of 4-Amino-3-methoxybenzeneboronic acid, pinacol ester (150 mg, 0.602 mmol) in DCM (2 ml) was added triethylamine (1.2 eq., 0.722 mmol, 73.1 mg, 100.76 μί) followed by t-butyiacetal chloride (1.2 eq., 0.722 mmol, 96.8 mg, 99.9 μΙ) and the mixture allowed to stir for 20 hours. Water was added to the mixture and the organic layer separated, dried over MgS0 4 and concentrated in vacuo. The product was purified by column chromatography, MeOH/DCM (0-3%) to give a brown solid (195.7 mg, 0.564 mmol, 93.6 %). 1 H NIUIR (500 MHz, CDCI3) δ 8.44 (d, J = 8.0, 1 H), 7.83 (s, 1 H), 7.47 (dd, J = 8.0, 1.0, 1 H), 7.32 - 7.29 (m, 1 H), 3.95 (s, 3H), 2.31 - 2.27 (m, 2H), 1.36 (s, 12H), 1.13 (s, 9H); 3 C NMR (126 MHz, CDCI3) 6 170.06 (C), 169.99 (C), 146.89 (C), 130.52 (C), 128.54 (CH), 1 18.61 (CH), 115.22 (CH), 83.75 (2x C), 55.86 (CH 3 ), 52.08 (CH 2 ), 31.22 (C), 29.81 (3x CH 3 ), 24.86 (4x CH 3 ); MS (ES +ve) [M+H] + : 348.6, 370.6 (+Na). N-[2-methoxy-4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaboroIan-2-yl)phenyl]-3-phenyl- propanamide:

To a solution of 4-Amino-3-methoxybenzeneboronic acid, pinacol ester (150 mg, 0.602 mmol) in DCM (2 ml) was added triethylamine (1.2 eq., 0.722 mmol, 73.1 mg, 100.76 μΙ) followed by hydrocinnamoy! chloride (1.2 eq., 0.722 mmol, 121.3 mg, 106.9 μΙ) and the mixture allowed to stir for 23 hours. The mixture was concentrated in vacuo and purified by column chromatography, MeOH/DCM (0-2 %) to give a brown solid, (240.0 mg, 0.629 mmol, 100 %). 1 H NMR (500 MHz, CDCI3) δ 8.44 (d, J = 8.0, 1 H), 7.83 (s, 1 H), 7.47 (d, J = 8.0, 1 H), 7.35 - 7.30 (m, 2H), 7.28 - 7.21 (m, 4H), 3.90 (s, 3H), 3.14 - 3.04 :(m, 2B), 2,79 - 2,72 (m, 2H) S 1.37 (s, 12H); C NMR (126 MHz, COCI3) δ " 170.25 (C), 170.17 (C), 148.82 (0}, 140.70 (C), 130,40 (C), 128,56 (OH), 128.52 (CH), 1.28.40 (CH), 126,34 (CH} » 118,68 (CH), 115,21 (CH), 83.78 (2x C), 55.84 (CH 3 ), 39.72 (CH 2 ), 31,42 (CH 2 ), 24.87 (4x CH 3 ); MS (ES +ve) P+Hf ; 382.7, 390.6 (+Na)

' N - 2~meihoxy-4-f ,4 f Sj S~tetra rnethy H ,3-d i oxota rs~2»y I ) p eny Sj~2~ p he rvy l-acetam Id© ;

To a solution of ^-Amino-S-meihoxybenzeneboron c acid, pinacorester (150 mg, 0.602 mmo!) in DCM (2 ml) was added tneihy!amine (1.2 eq., 0,722 mmoi, 73.1 mg, 100.78 ί) followed by pheftylaceiyl chloride (1.2 eq., 0.722 mmoi, 11 1.2 rng. 95.1 μί). and the mixture allowed to stir for 18 hours. The mixture was concentrated in vacuo and purified by column chromatography, !¾1e H/DCM (0-1.5 %} to give a cream solid, (200.9 mg, 0.547 mmoi, 80.9 %). 1 H UMR (5G0 MHz, CDCiS) δ 8.40 (d, J = 8.0, 1 H), 7.95 (s, !H), 7.43 (ddd, J = 11.3, 6,6, 1 ,6, 3H), 7.39 - 7.33 (m, 3H), 7,22 (d, J = 1.0, 1 H>, 3.78 (s, 5H) . , 1.35 (s, 12H}: n C NMR (126 MHz, CDC 13) δ 68.92 C), 168,84 (C), 147,04 (C), 134.50 (G), 130.31 . (G), 129.61 (2x GH), 29.05 (2x CH), 128.51 (CH), 127.47 (CH), 118.43 (CH), 1 15.31 (CH), 83.76 (C), 55.88 (CHa), 45.24 (ΟΗ ? ), 24.86 (4.x CH 3 ); SVIS (ES +ve) Μ+Η] + : 368.7, 390,6 (+Na)

N-beiizy^2-methoxy-4-(4,4,S,5¾ 0xafooro!aii- -yl)ai i!lne:

To solution of 4~Amino-3-methoxyfoenzeneboronie acid, pinaeol ester (150 mg, 0.602 mmoi) in DCM (2 ml) was added triethylamine (1.2 eq., 0.722 mmoi, 73,1 mg, 100.76 μί) followed by benzyl ch!oraformaie (1.2 eq., 0.722 mmoi, 123.2 mg, 103.07 μί) and the mixture allowed to stir for 18 hours. The product was concentrated in vacuo and purified by coiumn chromatography (100% DCM), to give a light brown thick oil, (65.9 mg, 0.172 mmol, 28.6 %). Only after characterisation was it discovered that the alkylated product (shown) had been produced instead. 1 H NMR (600 MHz, CDCI3) δ 7.43 - 7.26 (m, 8H), 6.68 (d, J = 7,6, 1 H), 4.41 (s, 2H), 3.91 (s, 3H), 1.35 (s, 12H); 13 C NMR (126 MHz, MeOD) δ 158.39 (C), 146.09 (C), 141.18 (C), 139.78 (C), 128.82 (CH), 128.08 (CH), 126.76 (CH), 126.52 (CH), 114.34 (CH), 108.93 (CH), 83.13 (CH 2 ), 54.55 (CH 3 ), 46.58 (C), 23.73 (4x CH 3 ); MS (ES +ve) [M+H] + : 340.6.

1-tert-butyl-3-[2-methoxy-4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)phenyl]urea:

To a solution of 4-Amino-3-methoxybenzeneboronic acid, pinacol ester (150 mg, 0.602 mmol) in DCM (3 ml) was added t-butylisocyanate (20 eq., 12.04 mmol, 1.19 g) and the mixture left to stir at for 72 hours. DCM and water were added to the mixture and the organic layer separated, dried over MgS0 4 and concentrated in vacuo. The product was purified by column chromatography, MeOH/DCM (0-2%) to give a dark brown solid, (40.9 mg, 0.117 mmol, 19.5 %). 1 H NMR (500 MHz, CDCI3) δ 8.09 (d, J = 8.0, 1 H), 7.44 (dd, J = 8.0, 1.2, 1 H), 7.27 (d, J = 1.0, 1 H), 6.88 (s, 1 H), 3.92 (s, 3H), 1.42 (s, 9H), 1.37 (d, J = 3.9, 12H); 13 C NMR (126 MHz, CDCI3) δ 153.97 (C), 146.81 (C), 131.78 (C), 128.70 (CH), 1 17.77 (CH), 1 15.36 (CH), 83.65 (CH), 55.74 (C), 50.97 (CH 3 ), 29.36 (C), 29.09 (3x CH 3 ), 24.87 (4x CH 3 ); MS (ES +ve) [M+H] + : 349.7, 371.6 (+Na) tert-butyi 2-[4-(4,4,5,5-tetramethyl lan-2-yl)phenyl]acetate:

4-{CarbQ yiriethyl)p enyiborGnic acid pinacoi ester (150 mg, 0.572 mrno!) was dissolved in 5 ml of dry toluene. Thionyi ohioride (1 ,2 eq,, 0.686 mmol, 81.6 mg, 49,8 u!) was added followed by a drop of DMF and the reaction heated to reflux (12G°C) for 2 hours. The reaction was cooled to r,t. then t-butylalcohoi (5 eq,, 2,86 mmoi, 21 ,8 mg, 0.273 ml) and trteth iamine (2.5 eq., 1.43 mmol, 1 9.3 ul) added and the mixture allowed to stir for 18 hours. Water and DCM were added to the mixture and the organic layer separated, dried over gSC and concentrated in vacuo. The crude product was purified by column chromatography, (100 % DCM), b give a light brown oil, (60 mg, 0.189 mraoi, 33.0 %}, MS (ES +ve) (jvl+H}*; 340.8 (+ a)

|3~(tert~butoxycarb©nylamino)- -meth acid:

(3~Am!nQ-4-methQxypherry!)boranic acid (150 mg, 0.898 mnioi) was suspended in DCM (5 ml). Di -tertbutyfdicarbonate (1.5 eq,, 1.347 mmol, 294,0 mg) was added followed by a small spatula of DMAP and the reaction allowed to stir fo 18 hours. Water was added to the mixture and the organic layer separated, dried over MgSCv, and concentrated in vacuo. The product was purified by column chromatograpny, MeOH/DCM (0-4 %) to give a dark brown solid (189,9 mg, 0.711 mmoi, 79,2 %). 1 H (500 MHz, MeOD) 5 8.12 (s, 1 H), 7,34 (dd, J = 8.2, 15, 1 H), 8.98 (d, J = 8.2, 1Ή), 3.88 (s, J = 4.9, 3H), 1 ,53 (s, 9H); ¾ UMR (128 MHz, MeOD) δ 153,77 (C), 150.S2 (C), 129.82 (CH), 129.49 (CH), 126.76 (C), 125.18 (C), 109.48 (CH), 79.78 (C), S4,74 (CH 3 ) 1 27,26 (3x CH 3 ); MS (ES +ve) [M+H]' : 268.5. tert-huty! 2~[2-mei oiX|i~4- 4,4,S J S etr methy » 1 ! 3^

acet . *

2-{4-Bromo-2-methoxyphenyl)acetic acid (2 g, 8.198 mmol) was dissolved in 20 ml of dry THF. Thionyl chloride (1.2 eq., 9.837 mmol, 1.17 g, 0.714 ml) was added followed by a drop of DMF and the reaction heated to reflux (80°C) for 2 hours. The reaction was cooled to r.t. then t-butylalcohof (5 eq., 40.99 mmol, 3.04 g, 3.92 ml) and triethylamine (2.5 eq., 20.49 mmol, 2.86 ml) added and the mixture left to stir for 20 hours. Water and EtOAc were added to the mixture and the organic layer separated, dried over MgS0 4 and concentrated in vacuo. The product was purified by column chromatography, EtOAC/Hexane (0-4%) to give a light orange liquid, (694.8 mg, 2.3 6 mmol, 28.2 %). 1 H NMR (500 MHz, MeOD) δ 7.10 (s, 1 H), 7.07 - 7.02 (m, 2H), 3.81 s, 3H), 3.47 (s, 2H), 1.42 (s, 9H); 13 C NMR (126 MHz, MeOD) 6 171.39 (C), 158.43 (C), 131.77 (CH), 122.99 (C), 122.98 (CH), 121.02 (C), 113.67 (CH), 80.51 (C), 54.84 (CH 3 ), 36.42 (CH 2 ), 26.85 (3x CH 3 ); MS (ES +ve) (M+H) + : 323.2/324.8 (+Na). tert-butyl 2-[2-methoxy-4-(4,4,5 J 5-tetramethyl-1 J 3,2-dioxaboroian-2-yl)phenyl] acetate:

To a solution of tert-butyl 2-[2-methoxy-4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yi)phenyl]acetate (600 mg, 2.0 mmol) in dioxane/water (18/2 ml) was added

bis(pinacolato)diboron (1.5 eq., 762.6 mg, 3.0 mmol), potassium carbonate (1.5 eq., 414.6 mg, 3.0 mmol) and triphenylphosphine (20 mol %, 104.9 mg) followed by palladium acetate (5 mol %, 22.5 mg) and the mixture heated in the microwave at 120 °C for an hour. MS showed a little SM so the mixture was heated for another hour. MS and TLC showed the reaction was complete. EtOAc and water were added to the mixture and the organic layer separated, dried over MgS0 4 and concentrated in vacuo. The product was purified by column chromatography, EtOAc/Hexane (0-10%) to give a colourless oil, (322.4 mg, 0.926 mmol, 46.3 %). 1 H NMR (500 MHz, CDCI3) δ 7.37 (d, J = 8.0, 1 H), 7.26 (d, J = 3.0, 1 H), 7.18 (d, J = 7.3, 1 H), 3.86 (s, 3H), 3.54 (s, 2H), 1.42 (s, 9H), 1.34 (s, 12H); 13 C NMR (126 MHz, CDCI3) δ 170.96 (C), 157.02 (C), 130.25 (CH), 127.29 (C), 127.28 (CH), 115.89 (CH), 109.97 (C), 83.75 (C), 80.34 (C), 55.46 (CH 3 ), 37.58 (CH 2 ), 28.03 (3x CH 3 ), 24.85 (4x CH 3 ); MS (ES +ve) [M+H] + : 349.7. 2~(4-br0mo-2~methoxy^ hen l)-N-pro '2^nyl"a0et rnsde:

2-{4~Bromo-2~methoxyphenyl)acetic acid (500 mg, 2.049 mmol) was dissolved in 5 ml of dry toluene. Thionyi chloride (1.2 eq,, 2.48 mmol, 292.7 mg, 178,3 ul) was added followed by a drop of D F and the reaction heated to reflux (1-20°C) for 3 hours, The reaction was cooled to r.t. then propargyiamine {2.5 eq., 5.12 mrnol, 282.2 mg, 0.328 mi) and trieihylamine (2.5 eq., 5.12 mmol, 0.714 ml) added and the reaction left to stir for 20 hours. DCM and water were added to the mixture and the organic layer separated, dried over MgS0 4 and concentrated in vacuo. The product was purified by column chromatography, MeOH/pCivl {0-3 %) to give a cream coloured solid, (458.2 mg, 1.631 mmol, 79.6 %). 1 H HMR (500 MHz, CDC!3) 5 7.13 - 7.07 (m, 2H), 7.04 (s. 1 H), 5,78 (s, 1 H), 3 99 (dd, J = 5,2, 2.6, 2H), 3.86 (s, 3H), 3.51 (s, 2H), 2,21 - 2.18 (ffi, 1 H); M C NMR (126 MHz,. CDCI3) δ 170,1 (C), 57.79 .(C), . 132.32 (CH), 124.16 {CB), 122.35 (C), 122,07 (C), 114.50 (CH), 79.53 {C}, 71.45 (CH), 55.85 (CH 3 ), 38.02 (GH 2 , 29.23 (Chi;,); MS (ES +ve) (M+H : 281 ,6/283,6, 3Q3.8/3Q6.Q (÷Na)

2-{4-bromo~2~methoxy-ph mg, 0.713 mmol) was dissolved in 2 mi of 1 ,2-d!chiGroethane in a microwave vial, FeCI 3 (0,5 eq., 57.8 mg, 0.356 mmol) was added and the mixture heated to 15G°G for 90 mins. Water and DCM were added to the mixture and the organic iayer separated, dried over MgS0 4 and concentrated in vacuo. The product was purified by column chromatography,

MeQH/DCM (1 %), to give a light yellow/orange oil, (108.7 mg, 0.389 mmol, 54.3 %). ¾f N R (500 MHz, CDCi3) δ 7.09 - 7.03 (m, 2H), 7.01 (s, 1 H), 6,65 (d, - 1.1 , 1 H), 4,04 (s, 2H), 3,82 (s, 3H), 2,28 (d, J = 1.2, 3H); % HMR (126 MHz, CDCI3) 5 161.68 (C), 157.87 (C), 148,90 (G) s 131.42 (CH), 123,68 (CH), 123 09 (C), 122.03 (CH), 121.65 (C), 1 14.31 (CH), 55.83 (CH 3 ), 28.30 (CH 2 ), 10.88 (CH 3 ); MS (ES +ve) (l\/I+H) + : 281.6/283.6, 303.8/306.0 (+Na).

2-[[2-methoxy-4-(4,4,5,5-tetramethyf-1,3,2-dioxaborolan-2 -yl)phenyl]methyl]-5- methyl-oxazole:

To a solution of 2-[(4-bromo-2-methoxy-phenyi)methyl]-5-methyl-oxazole (80 mg, 0.285 mmol) in dioxane/water (4.5/0.5 ml) was added bis(pinacolato)diboron (1.5 eq., 108.5 mg, 0.427 mmol), potassium carbonate (1.5 eq., 59.0 mg, 0.427 mmol) and

triphenylphosphine (20 mol %, 15.0 mg) followed by palladium acetate (5 mol %) and the mixture heated in the microwave at 120 °C for 30 mins. The mixture was

concentrated in vacuo and purified by column chromatography, EtOAc/hexane (30-40 %) to give a clear oil, (37.0 mg, 0.1124 mmol, 39.4 %). 1 H NMR (500 MHz, MeOD) δ 7.35 - 7.30 (m, 1 H), 7.20 - 7.12 (m, 1 H), 6.69 - 6.65 (m, 1 H), 5.97 (s, 1 H), 4.51 (s, 2H), 4.08 (s, 3H), 2.28 (s, 3H), 1.37 (s, 12H); MS (ES +ve) (M+H) + : 329.18

N-[4-[4-amino-1-[2-[4-(dimethylamino)-1-piperidyl]ethyl]p yrazolo[3,4~d]pyrimidin- 3-yl3-2-methoxy-phenyl]-3,3-dim (526):

To a. solution of i -[2^f4-{dimethyiamino}-1 -piperidy!]ethy!]-3Hodo~pyrazoio[3,4- d]pyrimidin-4-amine (50 rag, 0.1205 mrnoi) in dioxane/water (4,5 ml/0.5 mi) was added

[2-meth oxy~4~( , 4 , 5 ^

butanarrwde (1.5 eq., 83.1 mg, 0.181 mrrtoi), potassium carbonate (1.5 eq., 25.0 mg, 0.181 mmoi) and triphenylprrasphine (20 mol %, 9.5 mg) followed by palladium acetate (5 moi %) and the mixture heated in the microwave at 120 °G for 30 rn ins. The product was concentrated .in vacuo and purified by column chromatography, MeOH/DG (5~ 10% then 0-30 drops of trtethylamine per 100 ml) to give a sand coloured solid, (34,3 mg, 0.0675 mmoi, 56.0 %). 1 H N MR, {500 MHz, e.OO) δ 3,28 (s, 1 H), 8.10 (d, J = 8.1 , N), 7,35 (d, . J = 1.8, 1 H), 7,28 (dd, J = 8.1 , 1.8, 1 H), 4.57 (t, 6.7, 2H), 3.99 (s, 3H), 3. 5 (m, 2H), 2.95 (t, J = 6.7, 2H), 2.39 (m, 9H), 2 15 (i, J = 11 0, 2H), 1.91 (d, J = 16.8, 2H), 1.49 (ra, 2H), 1.14 (s, 9H); 13 C UMR (126 MHz, MeOD) δ 172.20 (C), 158.53 (C), 155,44 (GH), 154.22 (C), 151 ,00 (C), 144.8Θ (C), 123.52 (C). 127,69 (C), 123.12 (OH), 120.22 (CH), 1 10,74 (GH), 97.79 (C), 62,24 (CH), 56.24 (CHg), 55 08 (GH 3 ), 52.23 (2x CH 2 ), 49,79 (GH 2 ), 44,04 (GH 2 ), 40, 11 (2x CH 3 ), 30.67 (C), 28 81 (3x GH 3 ), 27.31 (2x CH 2 ); MS (ES +ve) [ +Hf: 509.8; HRM8 (ES +ve), G / H 1 N 8 0 2 -s-Hf ; calculated 509.33470, found 509.3363.

3-{4*am in o-S -methoxy-p e ny -[2- [4-(dimethy lam s np)-1 -p ί peri dyS] ethy ] pyrazol o

To a solution of 1 -[2-[4-{diraethyiamiriG)"1 -piperidyi]et y!]-3-iodo-pyrazolo[3,4- d]pyrimidin~4-amine (50 mg, 0.1205 mmoi) in dioxane/water (4.5 mi/0.5 mi) was added phenyl N^[2,-methoxy-4~(4,4,5,5- etramethyl-1 ,3

(1.5 eq., 66.8 mg, 0.181 mmoi), potassium carbonate (1.5 eq., 25,0 mg, 0.181 mmoi) and triphenyiphosphine (20 mol %, 9.5 mg) followed by palladium acetate (5 mo! %) and the mixture heated in the microwave at 120 °C for 30 mins. The mixture was concentrated in vacuo and the product purified by column chromatography, MeOH/DCM (10% then 0-30 drops of NEt 3 per 100 mi) to give a brown solid, ( 9.4 mg, 0.0473 mmol, 39.2 %). 1 H NMR (600 MHz, MeOD) δ 8.25 (s, 1 H), 7.16 (d, J = 1.8, 1 H), 7.09 (dd, J = 7.9, 1.8, 1 H), 6.91 (d, J = 7.9, 1 H), 4.54 (t, J = 6.7, 2H), 3.95 (s, 3H), 3.14 (d, J = 12.0, 2H), 2.93 (t, J = 6.7, 2H), 2.40 (m, 7H), 2.14 (t, J = 11.0, 2H), 1.90 (d, J = 12.3, 2H), 1.49 (d, J = 12.1 , 2H); 13 C NMR (126 MHz, MeOD) δ 158.62 (C), 155.35 (CH), 153.98 (C), 147.74 (C), 146.01 (C), 138.32 (C), 121.70 (CH), 121.07 (C), 1 14.69 (CH), 1 10.25 (CH), 109.97 (C), 62.19 (CH 2 ), 56.24 (CH 3 ), 54.74 (2x CH 2 ), 52.29 (CH), 43.91 (CH 2 ), 40.14 (2x CH 3 ), 27.35 (2x CH 2 ); MS (ES +ve) (M+H) + : 41 1.6; HRMS (ES +ve),

(M+H) + : calculated 4 1.26208, found 41 1.26270.

N-[4-[4-amino-1-[2-[4-(dimethyiamm

3-yl]-2-methoxy-phenyl]-2-phen -acetamide (530):

To a solution of 1-[2-[4-(dimethylamino)-1-piperidyl]ethyl]-3-iodo-pyrazolo[3 ,4- d]pyrimidin-4-amine (50 mg, 0.1205 mmol) in dioxane/water (4.5 ml/0.5 ml) was added N-[2-methoxy-4-(4,4,5,5-tetramethyl-1 ,3-dioxolan-2-yl)phenyl]-2-phenyl-acetamide (1.5 eq., 66.5 mg, 0.181 mmol), potassium carbonate (1.5 eq., 25.0 mg, 0.181 mmol) and triphenylphosphine (20 mol %, 9.5 mg) followed by palladium acetate (5 moi %) and the mixture heated in the microwave at 120 °C for 30 mins. The product was concentrated in vacuo and purified by column chromatography, MeOH/DCM (10% then 0-30 drops of triethylamine per 100 ml) to give a light brown solid, (37.9 mg, 0.07 7 mmol, 59.5 %). 1 H NMR (600 MHz, MeOD) δ 8.27 (s, 1 H), 8.20 (d, J = 8.2, 1 H), 7.40 (dt, J = 15.2, 7.5, 4H), 7.32 (dd, J = 11.0, 4.4, 2H), 7.26 (dd, J - 8.2, 1.8, 1 H), 4.56 (t, J = 6.7, 2H), 3.95 (s, 3H), 3.83 (s, 2H), 3.13 (d, J = 11.8, 2H), 2.93 (t, J = 6.7, 2H), 2.34 (d, J = 13.6, 7H), 2.13 (t,. J - 1 1.0, 2H), 1.88 {¾ J s 12.7, 2H), 1.47 (dt, J - 12.2, 8.6, 2H); 1 ¾ UMR (126 MHz, MeQD) δ 171.20 (C), 158-51 (C). 155.42 (CH), 154.17 (C), 150.35 (C). 144.81 (G), 135.19 (G), 129.30 (C), 128.98 (2x OH), 128.38 (2x CH) , 127.83 (6), 126.78 (CH), 122.00.(CH), 120,30 (CH), 110,62 (CH), 97.77 (C), 62.25 (CH), 58.21 (CH^, 55, 12 (CH 3 ), §2.24 (2x CH 2 ), 44.05 (CH 2 ), 43.30 (CHa), 40.09 (2x CH 3 ), 27.29 (2x CH 2 ); MS (ES +ve) [M+Hf : 529.6; RMS (ES ÷ve}, [M+Hf calculated 529.30340, found 529,3051 ,

3-yl]-2-methoxy-pfta^ }:

TQ a solution of 1-[2~[4-(diraethy!a ino^^

d3pyrimidin~4-amine (50 mg, 0. 205 mmol) in dioxane/water (4,5 ml/0.6 ml) was added N-p-mefbo ^

propanamide (1.5 eq, f 69.0 mg, 0.181 mmol), potassium carbonate ( .5 eq„ 25.0 mg, 0.181 mmol) and triphenyiphosphjne (20 mol %, 9.5 mg) followed by palladium acetate (5 mot %) and the mixture heated in the microwave at 120 °G for 30 roins. The product was concentrated in vacuo and purifred toy column chromatography, fvSeGH/DGM (10% then 0-30 drops of iriethyiaraine per 100 ml) to give a tight brown solid, (36.0 mg, 0.0664 mmol, 55.1 %). Ή R (600 MHz, MeOD) δ 8,27 (s, 1 H), 8.13 (d, J - 8.1 , 1 H), 7.27 (d, J« 1 ,7, 5H), 7.26 (d , J ^ 8.2, 1.8. 1 H), 7.21 (dd, . J = 8.6, 4.4, 1 H), 4.57 (t, * 6.7, 2H), 3,95 (s, 3H), 3.15 (d, J - 11 ,8, 2H), 3.05 (m, 3.H), 2,95 (t, J - 6,7, 2H), 2.81 (t, J = 7,7, 2H), 2.41 (s, 8H), 2, 14 {t, J - 1 1.0, 2H), 1 ,91 (d, J - 17.2, 3H), .49 (d, = 8.7, 2H); 13 C WR (126 MHz, MeOD) δ 172.65 (C), 158,53 (CH), 155.44 (G), 154.18 (C), 150,84 (C), 144.88 (C), 140,73 (C), 129,32 (C), 128,15 (CH), 127.70 (C), 125,85 (CH), 122.70 (CH), 120.20 (CH), 110.65 (CH), 97.77 (CH), 62.38 (C), 56.18 (CH 2 ), 55.06 (CH 3 ), 52.14 (2x CH 2 ), 46.33 (CH 2 ), 44.04 (CH 2 ), 39.99 (2x CH 3 ), 38.18 (CH 2 ), 31.38 (CH 2 ), 27.17 (2x CH 2 ); WIS (ES +ve) [M+H] + : 543.6; HRMS (ES +ve), C 30 H 39 N 8 O 2

[M+H] + : calculated 543.31905, found 543.3204. 3-[4-(benzyIamino)-3-methoxy-phenyl]-1 -[2-[4-{dimethylamino)-1 -piperidyf]ethyl] pyrazo[o[3,4-d]pyrimidin-4-amin

To a solution of 1-[2-[4-(dimethylamino)-1-piperidyl]ethyl]-3-iodo-pyrazoio[3 ( 4- d]pyrimidin-4-amine (50 mg, 0.1205 mmol) in dioxane/water (4.5 ml/0.5 ml) was added N-benzy[-2-methoxy-4-(4,4,5,5~tetramethyl~1 ,3,2-dioxaborolan-2-yl)aniline (1.5 eq., 6 .4 mg, 0.181 mmol), potassium carbonate (1.5 eq., 25.0 mg, 0.181 mmol) and

triphenylphosphine (20 mol %, 9.5 mg) followed by palladium acetate (5 mol %) and the mixture heated in the microwave at 120 °C for 30 mins. The mixture was concentrated in vacuo without and purified by column chromatography, MeOH/DCM (10% then 5-20 drops of NEt 3 per 100 ml) to give a light brown coloured solid, (31.8 mg, 0.0584, 48.5 %). H NWIR (400 MHz, MeOD) δ 8.24 (s, 1 H), 7.40 (d, J = 7.5, 2H), 7.33 (t, J = 7.5, 2H), 7.24 (t, J = 7.3, 1 H), 7.15 (d, J = 1.8, 1 H), 7.08 (dd, J = 8.1 , 1.8, 1 H), 6.67 (d, J = 8.1 , 1 H), 4.52 (t, J = 6.7, 2H), 4.47 (s, 2H), 3.98 (s, 3H), 3.13 (d, J = 11.9, 2H), 2.92 (t, J = 6.7, 2H), 2.37 (s, 7H), 2.13 (t, J = 11.0, 2H), 1.88 (d, J = 12.3, 2H), 1.47 (d, J = 8.5, 2H); 13 C NMR (126 MHz, MeOD) δ 166.23 (C), 158.62 (C), 155.34 (CH), 153.97 (C), 147.34 (C), 139.75 (C), 139.27 (C), 128.13 (2x CH), 126.78 (2x CH), 126.58 (CH), 121.15 (CH), 1 19.94 (C), 109.78 (CH), 109.27 (CH), 97.66 (C), 62.17 (CH), 56.20 (CH 2 ), 54.81 (CH 3 ), 52.22 (2x CH 2 ), 46.71 (CH 2 ), 43.89 (CH 2 ), 40.08 (2x CH 3 ), 27.28 (2x CH 2 ); MS (ES +ve) [M+H] + : 501.4; HRMS (ES +ve), C^H^N t [M+H] * : calculated 501.30848, found 501.3087.

ylj~2-methoxy-pheri y!] ~3~tert-but :

To a solution of H2-[4-{diniethyiamino)

d]pynrnidin-4-amine (50 mg, 0.1 05 mmof in diGxane/water (4,5/O.S ml) was added 1- tert-butyi-3-[2-methoxy-4-(4,4 ! 5 ! 5-tetramethyl-1 ^-dioxaboro!an-SI-yljphenyiliirea (1 ,5 eq,, 62.9 mg, 0.181 mrnof), potassium carbonate (1.5 eq„ 25.0 mg, 0,181 mmo!) and triphenylpbosphine (20 moi %, 9.5 mg) followed by palladium acetate (§ mol %) and the mixture heated in the microwave at 120 C for 30 mins. The mixture was concentratsd tn vacua and purified by coiurnn chromatograpny, MeOH/DC!Vl (10% then 0-30 drops of NEt 3 per 100 mi) to give a light brown solid, (30.4 mg, 0,0697 mmoi, 49,5 % }. *H MMH (500 MHz, MeOD) δ 8.24 (s, H), 8. 6 (d, J = 8.3, 1 H), 7.25 (d, J = .8, 1H), 7.19 (dd, J = 8,3, 1.9, 1 H), 4,53 (t, J ~ 6.7, 2H), 3.96 {s, 3H), 3.12 (d, J - 1 1.9, 2H), 2,92 ( J = 6.7, 2H), 2.36 (m, 7H), 2.12 (t, J = 1 1.1 , 2H), 1 ,87 (d, J = 12.5, 2H), 1.51 - 1.42 (m, 2H), 1.38 (s, SH); 13 0 HUH (126 MHz, MeOD) δ 155.74 (C), 155.40 (CH), 154.09 (C), 148,52 (C), 145,33 (C), 130.34 (C), 125;68 (C), 120,49 (CH), 118 66 (CH), 110.0 (CH), 97,73 (C), 62,27 (CH), 56.23 (GH 2 ), 55.02 (CH 3 ), 52.22 (2x CH 2 ), 49.71 (C), 43,97 (CH 2 ), 40.07 (2x GHg), 28.15 (Sx CHa), 27.20 (2x CH,); MS (ES +ve) [M+H : 510,8; RUB calculated 510,32995, found 510.32830, tert-buiyl N 3-[4-amino ^ 1-P-[4-(dimet ylamsno)»1-p|peridyS]ethyi]

d] p si mid i n-3 -y I] henyljcarbamate (542):

To a solution of 1-[2-[4-(dimethylamino)-1-pipendyl]ethyl]-3-iodo-pyrazolo[3, 4- d]pyrimidin-4-amine (50 mg, 0.1205 mmol) in dioxane/water (4.5/0.5 ml) was added 3- (boc-amino)benzeneboronic acid (1.5 eq., 42.9 mg, 0.181 mmol), potassium carbonate (1.5 eq., 25.0 mg, 0. 81 mmol) and triphenylphosphine (20 mol %, 9.5 mg) followed by palladium acetate (5 mol %) and the mixture heated in the microwave at 120 °C for 30 mins. The mixture was concentrated in vacuo and purified by column chromatography, MeOH/DCM (10% then 0-30 drops NEt 3 per 100 ml) to give a cream solid, (28.0 mg, 0.0583 mmol, 48.4 %). 1 H NMR (500 MHz, MeOD) δ 8.27 (s, 1 H), 7.88 (s, 1 H), 7.50 - 7.44 (m, 2H), 7.37 (d, J = 7.0, 1 H), 4.57 (t, J = 6.7, 2H), 3.16 - 3. 0 (m, 2H), 2.95 (t, J = 6.7, 2H), 2.35 (m, 7H), 2.14 (t, J = 1 1.0, 2H), 1.88 (d, J = 12.7, 2H), 1.56 (s, 9H), 1.47 (m, 2H); 13 C NMR (126 MHz, MeOD) δ 158.42 (C), 155.37 (CH), 154.22 (C), 154.00 (C), 144.97 (C), 140.14 (C), 133.30 (C), 129.46 (CH), 122.27 (CH), 1 19.16 (CH), 118.54 (CH), 97.74 (C), 79.72 (C), 62.16 (CH), 56.21 (CH 2 ), 52.29 (2x CH 2 ), 44.06 (CH 2 ), 40.16 (2x CH 3 ), 27.38 (2x CH 2 ), 27.28 (3x CH 3 ); MS (ES +ve) [M+H]*: 481.4; HRMS (ES +ve), C 25 H 37 N 8 0 2 [M+H] + : calculated 480.30340, found 481.3054. tert-butyl N-[5-[4-amino-1 -[2-[4-(dimethy!amino)-1 -piperidyl]ethyi]pyrazolo[3,4- d]pyrimidin-3-yl3-2-pyridyl]carbamate (549):

To a solution of 1-2~[4~{din etb iamino)-1-pipendyl]ethy!3-^

d]pynniidin-4-aroine (50 mg, 0.1205 mmoi) in d xane/water (4,5/0.6 mi) was added (6- ((tert " ButoxyGarbonyl)amino)pyridin-3-yi)boronic add (1.5 eq. s 43, mg, 0.181 mmoi), potassium carbonate (1 ,5 eq., 25.0 mg, 0,181 mmoi) and triphenyiphospHine (20 mo! %, 9,5 mg) followed by palladum acetate (5 mol %) and the mixture heated in the microwave at 120 °C for 30 mins. The mixture Was concentrated in vacuo and purified by column chromatography, MeOH/DCfVI ( 0 % then 10-30 drops of NE¾ per od ml) to give a white solid, (23.8 mg, 0,0495 mmoi, 41.0 %). 1 H H R (500 MH^ ' MeOD) ' δ 8.55 -8.51 (m, IN), 8.26 (s, N), 8.06-8,01 (m, 2H), 4.55 (t, J « 6.5, 2H, 3.14 (d, J= 12.0, 2H), 2.94 (t, J ~ 6,5, 2H), 2.88 (m, 1 M), 2.55 (s f 6H), 2.14 (t, J ~ 11.0·, 2H), 1 ,92 (d, J = 12,7, 2H), 1.55 (s, 9H), 1,53- 1.48 (m, 2H); 1, C H (126 MHz, MeOD) δ 158.58 (C), 1S5.47 (CH), 154.40 (C), 153.05 (C), 152,96 (G), 147.13 (CH), 141.83(C), 137.78 (CH), 123.46 (C), 112.28 (CH), 97,92 (C), 80.49 (C), 62.79 (CH), 56.00 (CH¾), 51.78 (2x CH¾), 44.12 (CH 2 ), 39.84 (2x CH-,}, 27.16 (3x GH S ), 26.76 (2x GH 2 ); M (ES ÷ve) M*H .: 481.8; HR!ViS (ES +ve), ΡΛβΝι [M÷Hf: calculated 482.29865, found 482,3004. tert-buty! 2-[ -[ -aiBSno-1-2-l4-|dimeh iamin0-1~pi^

€lJpyrimicin-3-yI] heny acetate (553):

To a solution of 1-[2-[4-(dimethylamino)-1 -piperidyi]ethyl]-3-iodo-pyrazolot3,4- d]pyrimidin-4-amine (50 mg, 0.1205 mmol) in dioxane/water (4.5/0.5 ml) was added tert-butyl 2-[4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaboro!an-2-yl)phenyljacetate (1.5 eq., 57.6 mg, 0.181 mmol), potassium carbonate (1.5 eq., 25.0 mg, 0.181 mmol) and

triphenylphosphine (20 mol %, 9.5 mg) followed by palladium acetate (5 mol %) and the mixture heated in the microwave at 120 °C for 30 mins. The mixture was concentrated in vacuo and purified by column chromatography, MeOH/DCM (10 % then 0-30 drops of NEt 3 per 100 ml) to give a light brown thick oil, (40.5 mg, 0.0845 mmol, 70.1 %). H NMR (500 MHz, MeOD) 5 8.26 (s, 1 H), 7.68 - 7.63 (m, 2H), 7.47 (d, J = 8.2, 2H), 4.55 (t, J = 6.6, 2H), 3.68 (s, 2H), 3.12 (d, J = 1.9, 2H), 2.93 (t, J = 6.6, 2H), 2.43 (m, 7H), 2.13 (t, J = 11.0, 2H), 1.87 (m, 2H), 1.47 (m, 1 1 H); 3 C NMR (126 MHz, MeOD) δ 171.52 (C), 158.52 (C), 155.44 (CH), 154.21 (C), 144.85 (C), 136.00 (C), 131.41 (C), 129.95 (2x CH), 128.45 (2x CH), 97.81 (C), 81.01 (C), 62.40 (CH), 56.15 (CH 2 ), 52.10 (2x CH 2 ), 44.04 (CH 2 ), 41.58 (CH 2 ), 39.95 (2x CH 3 ), 27.13 (2x CH 2 ), 26.88 (3x CH 3 ); MS (ES +ve) [M+H] + : 480.0; HRMS (ES +ve), C 26 H 38 N 7 0 2 [M+Hf: calculated 480.30815, found 480.3121.

2-[4-[4-amino-1-[2-[4-(dimethylamino)-1^^

yl]phenyl]acetic acid (556):

To a solution of 1-[2-[4-(dimeihylamjnQ)-1~pip^

d]pynmidin-4-amine (50 mg, 0.1205 mm pi) In dsoxane/water (4,5/0.5 mi) was added 4-

potassium carbonate (1.5 eq., 25,0 mg, 0,181 mmol) and triphenyiphosphine (20 mol %, 9.5 mg) followed b palladium acetate (5 moi %) and the mixture heated in the microwave at 120 °G for 30 mins. The mixture was concentrated in vacuo and purified by column chromatography, IVteQH/DCM (10 % then 0-50 drops of NEt 3 per 00 mt) to give a cream solid, (25.0 mg, 0.0591 mmol, 49.0 %). 1 H HMR (500 MHz, eQD) δ 8.26 (s, 1 H), 7.59 (d, J " 8,2, 2H), 7.49 (d, J = 8.2, :2H), 4.55 (t, J = 6.3, 2M), 3.62 (s, 2H),

3.20 - 3.15 (m, 2H), 2.98 (t, J - 8.3, 3H), 2.74 (s, 6H), 2.18 (t, J = 11.0, 2H), 2.03 - 1.96 (m, 2H), 1.56 (dd, J - 12.1 , ¾9, 2H); i3 G HMR (128 MHz, MeOD) β 177.8 (G), 158,57 '■ (C) , 155.42 (CH), 154.16 (C), 145.31 (C), 139.11 (C), 130.29 (G), 129.88 (2x CH), 128.10 (2x CH), 97.82 (C), 63.29 (CH 2 ), 55.78 (GH 2 ), 51.37 (2x CH Z ), 44.08 (GH 3 ), 39.26 (2x CH§)> 29.53 (CH), 26,34 (2.x CH¾); MS (ES +ve) [M+Hf: 424.4: HRftHS (ES +ve), C 22 H 30 N ? O £ +ΗΓ: calculated 42424555, found 424.2475. tert-buiyl - » 4-amino-1~[2-[4-(dimethylamsnoH-P^P

dJpyrimWin-S-ylJ-S-methQxy- henylJcarbamate (550):

To a solution of 1-[2-[4-(dimethylamino)-1-piperidyl]ethyl]-3-iodo-pyrazoio[3 ,4- djpyrimidin-4-amine (50 mg ( 0.1205 mmol) in dioxane/water (4.5/0.5 ml) was added [3- (tert-butoxycarbonylamino)-4-methoxy-phenyljboronic acid (1.5 eq., 48.4 mg, 0.181 mmol), potassium carbonate (1.5 eq., 25.0 mg, 0.181 mmol) and triphenylphosphine (20 mol %, 9.5 mg) followed by palladium acetate (5 mol %) and the mixture heated in the microwave at 120 °C for 30 mins. The mixture was concentrated in vacuo and purified by column chromatography, MeOH/DCM (0- 0 % then 0-20 drops of NEt 3 per 100 ml) to give a light brown solid, (32.9 mg, 0.0645 mmol, 53.5 %). H NMR (500 MHz, MeOD) 6 8.26 (d, J = 5.2, 1 H), 8.19 (d, J = 2.1 , 1 H), 7.41 (dd, J = 8.4, 2.2, 1 H), 7.19 (d, J = 8.5, 1 H), 4.56 (t, J = 6.6, 2H), 3.99 (d, J = 5.1 , 3H), 3.15 (d, J = 12.1 , 2H), 2.96 (t, J = 6.6, 2H), 2.54 (m, 1 H), 2.48 (s, 6H), 2.16 (t, J = 10.9, 2H), 1.92 (d, J = 10.5, 2H), 1.56 (d, J = 5.1 , 9H), 1.55 - 1.46 (m, 2H); 3 C NMR (126 MHz, MeOD) δ 158.48 (C), 155.35 (CH), 154.20 (C), 153.82 (C), 149.93 (C), 145.03 (C), 128.25 (C), 124.95 (C), 123.27 (CH), 119.69 (CH), 1 1 1.00 (CH), 97.67 (C), 80.15 (C), 62.52 (CH), 56.07 (CH 2 ), 55.11 (CH 3 ), 51.99 (2x CH 2 ), 44.01 (CH 2 ), 39.86 (2x CH 3 ), 27.22 (3x CH 3 ), 27.03 (2x CH 2 ); MS (ES +ve) [M+Hj + : 511.4; HRMS (ES +ve), C 26 H 39 N 8 0 3 [M+H] + : calculated 511.31396, found 51 1.3166. tert-butyl 2-[4-[4-amino-1 -[2-[4-(dimethylamino)-1 -piperidyl]ethyl]pyrazolo[3,4- d]pyritnidin-3-yl]-2-methoxy-phenyl]acetate (565):

To a solution ' of 1 ~f2-[4-(climethylamino)"1 -piperidy!JethyiJ-S-iodo-pyrazoSop^- d]pyriinidin-4-amfne (34.7 mg, 0,0838 mmoi) in dioxane/water (4,5/0.5 ml) was added tert-butyl 2-[2-m8ihQxy-4»{4,4,5, 5-tetram (1 eq,, 29.1 mg, 0,0836 mmoi), potassium carbonate (1.5 eq., 17.3 mg, 0.125 mmoi} and triphenyfphosphine (20 mo! %, 4,4 mg) Followed b palladium acetate (5 mo! ) and the mixture heated in the microwave at 120 °G for 30 mins. The mixture was concentrated in vacuo and purified by column chromatography, iVleOH/DCSyi (0-10 % then 0-30 drops of NEt 3 per 00 mi) to give a cream solid, (19 mg, 0,0373 mmoi, 44.6 %). 1 H MR (500 MHz, eOD) 5 8.26 (s, H), 7,35 (d, J =7.6, 1 HJ, 7.28 (d, J - 1.4, 1 H), 7.22 {dd, J «

7.5, 1.6, 1 H), 4.56 (t, J = 6.7, 2H), 3,91 (s, 3H), 3.84 (s, 2H), 3.13 J - 11.9, 2H), 2.94 (t, J = 8.7. 2H), 2.37 (m, 7H), 2.13 (t, J = 1 .0, 2H), 1.91 - 1.85 (m. 2H), 148 (m, 1 1 H); 13 C N ' MR (126 MHz, SVfeOD) 5 172.07 (C), 158,55 (C), 158.18 (C), 155.46 (OH), 154,14 (C), 145.09 (C), 133,02 (C), 131.48 (CH), 124.76 (C), 120.21 (CH), 110.46 (CH), 97.82 (C), 80.76 (C), 62.28 (CH), 56.22 (CH 2 ), 54.70 (GH 3 ), 52.24 (2 CH 2 , 44.04 (CH 2 ), 40,09 (2x CH 3 ), 38.61 (CHs), 27.28 (2x CH¾), 28,00 (3x GH 3 ); MS (ES +ve) [Μ+Η : 510.2; HRWS (ES +ve), C 27 H4 0 N 7 O 3 [ +Hf ; calculated 510.31872, found 510.3185,

3 odo*1 »[2-{4-metiiySpi eraziit-1 -yl}ethy pyrazoSo[3,4-d]pyrimldln-4»amirie:

100 mg, 0.265 mmol of 1-(2,2-diethoxyethyi)-3-iodo-pyrazolo[3,4-d]pyrimidin-4-amin e was added to a 5 ml microwave tube. 2.5 ml of water and 2.5 ml of TFA were then added and the mixture heated to 100 °C for 30 mins in the microwave. The solvents were removed in vacuo to give a dark brown oil. 0.265 mmol of 2-(4-amino-3-iodo- pyrazoio[3,4-d]pyrimidin-1-yl)acetaldehyde was suspended in 3 ml of DCM. 1- methylpiperizine (1.5 eq., 0.399 mmol, 39.8 mg, 44.1 ul) was added foilowed by a drop of acetic acid and the mixture allowed to stir for 10 mins. Sodium triacetoxyborohydride (1.5 eq., 0.399 mmol, 84.6 mg) was added and the mixture allowed to sttr for 17 hours overnight. The mixture was concentrated in vacuo and purified by column

chromatography, MeOH/DCM (5-10 % then 0-15 drops of NEt 3 per 100 ml) to give a light orange thick oil, (89.1 mg, 0.2302 mmol, 86.9 %). 1 H NMR (500 MHz, MeOD) δ 8.22 (s, 1 H), 4.51 (t, J = 6.0, 2H), 3.33 (d, J = 1.7, 4H), 3.20 - 3.04 (m, 4H), 2.98 (t, J = 6.0, 2H), 2.83 (s, 3H); 3 C NMR (126 MHz, MeOD) δ 158.05 (C), 155.67 (CH), 153.80 (C), 103.55 (C), 87.02 (C), 55.51 (CH 2 ) r 53.44 (CH 2 ), 49.45 (CH 2 ), 44.18 (CH 2 ), 43.07 (CH 2 ), 42.03 (CH 3 ); MS (ES +ve) (M+H) + : 388.4.

1-[2-[4-(dimethylamino)-1-pipericlyl]ethyl]-3-[3-methoxy- 4-[(5-methyloxazol-2- yl)methyl]phenyl]pyrazolo[3,4-d]pyrimidin-4-amine (584):

To a solution of 1÷[2- 4-(climethylamino)-1 -piper!dyi]ethyl]~3-iodo-pyrazQ!of3 1 4~ d]pyrimidjn-4-amine (50 mg, 0.1205 mmol.) in: dioxaiie/Wafer (4.5/0.5 mi) was added 2- f[2-methoxy-4-(4,4,5,5-tettamethy^

oxazoie (1 e ., 39.7 mg, 0,1205 mmol), potassium .carbonate (1.5 eq., 25,0 rag, 0.181 mmol) and triphenyiphosphine (20 mol %, 9.5 mg) followed by palladium acetate (5 mol %) and the mixture heated in the microwave at 120 °C for 30 mins. The mixture was concentrated /? vacuo and purified by column chromatography, MeOB/DGIVl (10% then 0-30 drops of NEt 3 per 100 ml) to give a pale yellow solid, (36.8 mg, 0,0751 mmol, 62,3 %). 1 H N¾R (5O0 MHz, e ' O.D) δ 8,28 (s, 1H) S 7.38 (d, J ~ 7.8, 1H), 7.29 (s, 1H), 7.28 (dd, J - 7.6, 1.6, 1H), 6.69 (d, J ~ 1.2, 1 H), 4.57 .(t, J = 6,6. 2H), 4.15 (s. 2.H), 3.91 (s, 3H), 3.17 (d, J * 7,4, 2H), 2.96 (t, J - 6,6, 2H), 2.69 (n 1H), 2.56 (s, 6H), 2.31 {s, 3H), 2.17 (t, J = 1.10, 2H), 1.94 (m, 2H), 1.54 (m, 2H); 1S C ig (126 MHz, eOD) δ 162.54 (C), 158.55 (C), 157.95 (C), 155,47 (CH), 154.21 (C)„ 149.33 (C), 145.01 (G), 133,18 (C), 130.96 (CH), 124 ^ 85 (C), 121.58 (CH), 120.34 (CH), 110,70 (CH), 97.78 (C), 62.77 (CH), 56.03 (CH 2 ), 54.78 (CHs), 51 ,82 (2x CH 2 ), 44.08 (CH 2 ), 39.66 (2x GH 3 ), 28.17 (CH?), 26.79 (2x CH 2 ), 9.16 (CH 3 ); MS (ES ÷ve) P+Hf : 491.8; HRMS (ES +ve), CseHas sOi P+Hf: calculated 491.28775, found 491.2862, 34©dQ 1~[2~(4~met i©xy-1-piperM^ ^

100 mg, 0.287 mmo! of 1-(2,2-dimethoxyethyl)-3-iodo-pyrazolo[3,4-d]pyrimidin--4-am ine was added to a 5 ml microwave tube. The mixture was then concentrated in vacuo. 0.287 mmol of 2-(4-amino-3-iodo-pyrazo!o[3,4-d]pyrimidin-1-yl)acetaldehyde was suspended in 3 ml of DCM. 4-methoxypiperidine (1.5 eq., 0.430 mmol, 49.5 mg) was added followed by a drop of acetic acid and the mixture allowed to stir for 10 mins. Sodium triacetoxyborohydride (1.5 eq., 0.430 mmol, 91.1 mg) was then added and the mixture allowed to stir for 72 hours. The product was concentrated in vacuo and the product purified by column chromatography, MeOH/DCM (0-8 %) to give a pale yellow solid, ( 12 mg, 0.279 mmol, 97.1 %). 1 H NMR (500 MHz, MeOD) δ 8.23 (s, 1 H), 4.53 (t, J = 6.6, 2H), 3.29 (s, 1 H), 2.98 (t, J = 6.5, 2H), 2.91 (s, 2H), 2.45 (s, 2H), 1.89 (s, 2H), 1.57 (s, 2H); 13 C NMR (126 MHz, MeOD) δ 158.09 (C), 155.71 (CH), 153.68 (C), 103.71 (C), 87.16 (C), 75.13 (CH), 56.19 (CH 2 ), 54.43 (CH 3 ), 50.24 (2x CH 2 ), 43.96 (CH 2 ), 29.68 (2x CH 2 ); MS (ES +ve) (M+H) + : 403.0. tert-butyl N-E4-[4-amino-1-[2-(4-methoxy~1-piperidyl)ethyl]pyrazolo[3,4 - d]pyrimidin-3-yl]-2-methoxy-pheny!]carbamate (593):

To a solution of 34odo-1 -[2-(4-methoxy-1 - iperidyl)ethyt]pyrazotpi 3,:4-d]pyrsm!din-4- amine (50 mg, 0,124 mmot) in dioxane/w ter (4.5 rn!/0.5 ml) was added |4~(feri- bistoxycarbonyl:amino)-3-methoxy-pheny!]boron acid (1 ,5 eq., 49.8 mg, 0.187 mmof), potassium carbonate (1.5 eq., 25.8 mg, 0.18 mmol) and triphenyiphosphine (20 mol %, 6.5 mg) followed by palladium acetate (5 mol %) and the mixture heated in the microwave at 120 °C for 30 mins. The mixture was concentrated in vacuo and purified by column chromatography, MeOH/DCIV! (0-10 %) ■■ to give a cream coloured solid, (60,0 mg, 0.1:21 mmol, 97.3 %). % HM R (500 MHz, SVteQD) 5 8.2® (s, 1Ή), 8,08 (d ( J = 8,2, 1 H), 7,31 (d, J ~ 1.8, 1 H), 7.27 (dd, J = 8.2, 1.8, 1 H), 4,83 (t, J = 6.6, 2H), 3.98 (s, 3H), 3.34 ( 3H), 3.12 (m ( 2H), 3,02 (m, 2H), 2,60 (m, 2H), 1.93 (m, 2H), 1 ,65 (m, 2H), 1 ,57 (s, 9H); 13 C HMR ( 6 MHz-, MeGD) δ 158,53 (C), 55.46 (CH), 154.22 (C), 153,42 (C), 149.28 (C), 145.17 (C), 128.73 (C), 27.38 (C), 120.44 (CH), 1 19,60 (CH), 1 10.33 (CH), 97.94 (C), 80.15 (C), 75. 2 (CH), 58.21 (CH 2 ) 1: 55,10 (CH 3 ), 54,79 (CH 3 ), 50.02 (2x

CH j ), 43,80 (CHa). 29.89 (2x CH,), 27.21 (3x CH 3 ) : ; MS (ES +ve) +H ; 498.2; HRftfiS (ES +ve), C 2 sH 3 sN 7 0 4 [M+Hf ; calculated 498,28233, found 498,2850.

1.-(2, 2-ct fmet hoxyeth !)-34 οά o-py razol e :

To a solution of 3-iodo-1 H-pyrazole (500 ig, 2.578 mmol) in DMF (10 ml) was added sodium hydride (1.5 eq,, 3,867 mraoi, 60% dispersion in. mineral oil, 154,7 tog) and the suspension allowed to stir for 30 mins until the gas evolution had subsided.

Bromoaceialdehyde dimethyl acetai (1.5 eq. 3.857 mmol, 649,6 mg, 0.454 ml) was then added dropwise and the mixture heated at 150 C C i the microwave for an hour. The mixture was concentrated in vacuo in order to remove as much DMF as possible, EtOAc and water were then added to the mixture and the organic layer separated. The aqueous layer was washed twice with EtOAc and organics combined, dried over MgSQ and concentrated in vacuo. The product was purified by column

chromatography, SvteQH/DOM (0-1 %) to give a light orange solid, (519.2 mg, 1.841 mmol, 71.4 %). H HMR (500 MHz, CDCiS) δ 7.28 (d, J ~ 2.3, 1H), 6.40 (d, J ~ 2.3, M), 4.62 (t, J ^ 5.4, 1 H), 4.20 (d, J ~ 5.4, 2H), 3.37 (d, J = 2,4, 6H); 13 C HM (126 z, GDCI3) δ 132.61 (CH), 114.84 (CH), 103.29 (CHa), 94.49 (C), 55.20 {2X.CH 3 ), 54,55 (CH); m (ES +ve) ( +Η) ·; 304.6 (+Na). 1-[2-(3-iodopyrazol-1-yl)ethyl]-N,N-dimethyl^iperidin-4-amin e:

1-(2,2-dimethoxyethyi)-3-iodo-pyrazole (250 mg, 0.887 mmol) was added to a microwave vial followed by water (0.25 ml) and TFA (0.25 mi) and the mixture heated to 00 °C in the microwave for an hour. The product was concentrated in vacuo and used without further purification. 0.887 mmol of 2-(3-iodopyrazol-1-yl)acetaidehyde was suspended in 5 ml of DCM. N,N-dimethylpiperidine-4-amine (1.5 eq., 1.33 mmol, 170.5 mg) was added followed by a drop of acetic acid and the mixture allowed to stir for 10 mins. Sodium triacetoxyborohydride (1.5 eq., 1.33 mmol, 281.9 mg) was added and the mixture aliowed to stir for 72 hours. The mixture was concentrated in vacuo and purified by column chromatography, MeOH/DCM (5-10 % then 10-20 drops of NEt 3 per 100 ml) to give a thick light orange oil, (306.3 mg, 0.880 mimoi, 99.2 %). 1 H N R (500 MHz, MeOD) δ 7.54 (d, J = 2.3, 1 H), 6.43 (d, J = 2.3, 1 H), 4.27 (t, J = 6.4, 2H), 3.10 - 3.05 (m, 1 H), 3.05 - 2.99 (m, 2H), 2.85 - 2.76 (m, 8H), 2.17 (td, J = 12.0, 2.3, 2H), 2.01 (d, J = 13.2, 2H), 1.67 (tt, J = 12.1 , 6.1 , 2H); 13 C NMR (126 MHz, MeOD) δ 132.70 (CH), 114.34 (CH), 93.40 (C), 63.39 (CH), 56.57 (CH 2 ), 51.40 (2x CH 2 ), 49.59 (CH 2 ), 39.17 (2xCH 2 ), 26.24 (CH 2 ); MS (ES +ve) (M+H) + : 348.8. tert-butyl N-[4-[1 -[2-[4-(dimethylamino)-1 -piperidyl]ethyi]pyrazoi-3-yl]-2-methoxy- phenyfjcarbamate (597):

To a solution of 1~[2-(3-iodopyrazoi-1~y^ (50 mg,

0.1436 mmoi) in dloxane/water (4,5 mi/0.5 ml) was added [4-{tert- butoxycarfao^ (1.6 eq. t 57,6 mg, 0,215 mmoi), potassium carbonate (1 ,5 eq. : 297 mg, 0.21 S mmoi) and trip eny!pbos s e (20 rriol %, 7.5 mg) foil owed by palladium acetate (5 mol } and the mixture heated in the microwave at 120 °C for an hour. The reaction was con centrated ' in vacuo and purified by CQiurnft cHrGmatagraphy, MeGH/DC (5-10 %) to give a dark orange solid, (42.7 mg, 0,0963 mmoi, 67.1 %). 1 H NNtR (500 MHz, MeQDJ δ 7.88 (s, 1H), 7.69.d,J-2.3 f H), 7.42 (d, J - 1.7, H), 7.3 (dd, J =8.3, 1.8, 1H), 6.62 (d, J= 2.3, 1H), 4.33 (t J = 6,4, 2H), 3,96 s, 3H), 3.22 -3.15 (m, 1H), 3.11 (s, 2H), 2.94 (s, 2H), 2,85 (d, = 14.6, 6H), 2.28 (s, 2H), 2.05 (s, :2H), 1.73 (d, J = 8.3, 2H), 1.55 (d, J = 4.2, 9H); ¾ NM (126 MHz, MeOD) 6153.56 (C), 151,55 (C), 149.10 (C), 131.77 (CH), 128.50 (C), 127.36 (C), 123.00 (C), 1 9.24 (GH), 117.75 (CH), 107.31 (GH), 102.26 (CH), 63.48 (CH), 56.69 (GH 2 ), 54.95 (CH ? ), 51 ,21 (2x GH 2 ), 49.11 (CH 2 ), 39. 9 (2x CH S ), 27,23 (3x.CH 3 ), 26,22 (2x CH 2 ); MS (ES +ve) (M+ : 444.2; HR S (ES +ve), C 24 H 38 g0 3 ( +H) + : calculated 444,29692, found 444.29650.

2-£4-[4-amSsio H2H[ »dira^

y!l heriYSacetooitrie f 5-100):

To a solution of 1-[2-[4-(dimethylamino)-1-piperidyI]ethyl]-3-iodo-pyrazolo[3 ,4- d]pyrimidin-4-amine (50 mg, 0.1205 mmol) in dioxane/water (4.5/0.5 ml) was added 4- (Cyanomethyl)benzeneboronic acid, (1 eq., 29.1 mg, 0.181 mmol), potassium carbonate {1.5 eq., 25.0 mg, 0.181 mmol) and triphenylphosphine (20 mol %, 9.5 mg) followed by palladium acetate (5 mol %) and the mixture heated in the microwave at 120 °C for 30 mins. The mixture was concentrated in vacuo and purified by column chromatography, MeOH/DCM (5-10 % then 20 drops of NEt 3 per 100 ml) to give a cream solid, (41.8 mg, 0.1034 mmol, 85.8 %). 1 H NMR (500 MHz, MeOD) δ 8.27 (s, 1H), 7.79 - 7.70 (m, 2H), 7.59 (d, J = 8.3, 2H), 4.56 (t, J = 6.4, 2H), 4.03 (s, 2H), 3.18 (d, J = 5.8, 2H), 3.04 - 2.93 (m, 3H), 2.74 (s, 6H), 2.17 (dd, J = 15.1 , 6.9, 2H), 1.99 (d, J = 12.8, 2H), 1.61 - 1.51 (m, 2H); 13 C NMR (126 MHz, MeOD) δ 158.49 (C), 155.47 (CH), 154.36 (C), 144.35 (C), 132.22 (C), 128.84 (2x CH), 128.70 (2x CH), 117.95 (C), 1 0.00 (C), 97.74 (C), 63.27 (CH), 55.84 (CH 2 ), 51.39 (2x CH 2 ), 44.11 (CH 2 ), 39.27 (2x CH 3 ), 26.32 (2x CH 2 ), 21.98 (CH 2 ); MS (ES +ve) [M+H] * : 405.0; HRMS (ES +ve), C 22 H 2e N 8 (M+H) + : calculated 405.25097, found 405.24950.

1-[2-[3-(dimethylamino)-1 -piperidy[]ethyl]-3-iodo-pyrazoio[3,4-d]pyrimidin-4- amine:

100 mg, 0.287 mmo! of 1-(2,2-diffiet oxyethy!)-3^

was added to a 5 mi microwave tube. 2.5 ml of water and 2 5 mi of TFA were then added and the mixture heated to 100 °C for 30 mins in the microwave. The product was then concentrated in vacuo. 0.287 mmoi of 2- 4-amino-3-iodo-pyrazoloi3\.4Hjjpyrimidir - 1-yi}aeetaldehyde was suspended in 3 mi of PGM. S-dimefhyiaminopiperidine (1.5 eq. ; 0.430 mmoi, 55.1 mg) was added followed by a drop of acetic acid and the mixture allowed to stir for 10 mins. Sodium triacetoxyborohydride (1.5 eq., 0.430 mmoi, 91.1 mg) was added and the mixture allowed to stir for 16 hours. The mixture was concentrated in. vacuo and purified by column . ■chromatography, eOH/DCM (5-10 } to give a cream solid, ( 17.5 mg, 0.283 mmoi, 98.6 %). ' ¾ NMR (500 MHz, MeOD) δ 8.22 (s, 1H), AM - 4.43 (m, 2H), 3.08 (m, 1 H), 2.9Θ - 2,90 (m, 3H), 2.84 (s, 6H), 2.65 (m, 2Ή), 2,38 (m, H), 1.88 (m, 1 H), 1 ,75 - 1.65 (m, 2H), 1.55 - 1.47 (m, 1 H); 13 C MMR (126 MHz, MeOD) 5 58.13 (C), 155.80 (CH), 153.62 (C), 103.65 (C), 87,01 (C), 62.48 (CH), 56.37 (CH 2 ), 52.78 (CH 2 ), 52.75 (CH 2 ), 44.50 (CH 2 ), 40.40 (2x CH 3 ), 24.67 (CH 2 ). 21.95 (CHa); MB (ES +ve) (M+H) + 415.8 tert-buty! N~[ H^amino-1^2-[3 ^ (dimet^

pyrImid-ii-3--y{]-2^ (6 » 103):

To a solution of 1-[2-[3-{dimethylamino)-1-piperidyl]ethyl]-3-iodo-pyrazolo[3 ,4- d]pyrimidin-4-amine (50 mg, 0.1205 mmoi) in dioxane/water (4.5 ml/0.5 mi) was added [4-(tert~butoxycarbonylamino)-3-methoxy-pheny!]boronic acid (1.5 eq., 48.3 mg, 0.181 mmol), potassium carbonate (1.5 eq., 25.0 mg, 0.181 mmol) and triphenylphosphine (20 mol %, 9.5 mg) followed by palladium acetate (5 mol %) and the mixture heated in the microwave at 120 °C for 30 mins. The mixture was concentrated in vacuo and purified by column chromatography, MeOH/DCM (5-10 %) to give a dark red solid, (60.5 mg, 0.119 mmol, 98.4 %). H N R (500 MHz, MeOD) δ 8.25 (s, 1 H), 8.05 (d, J = 8.2, 1 H), 7.26 (d, J = 1.8, 1 H), 7.22 (dd, J = 8.2, 1.8, 1 H), 4.58 - 4.48 (m, 2H), 3.94 (s, 3H), 3.13 (m, 1 H), 2.97 (m, 3H), 2.82 (s, 6H), 2.66 (m, 2H), 2.42 (m, 1 H), 1.86 (m, 1 H), 1.71 (m, 2H), 1.52 (m, 10H); 13 C NMR (126 MHz, MeOD) δ 158.61 (C), 155.59 (CH), 154.1 1 (C), 153.49 (C), 149.33 (C), 145.10 (C), 128.93 (C), 127.24 (C), 120.44 (CH), 119.66 (CH), 1 10.28 (CH), 97.76 (C), 80.24 (C), 62.58 (CH), 56.39 (CH 2 ), 55.12 (CH 3 ), 52.86 (CH 2 ), 52.73 (CH 2 ), 44.28 (CH 2 ), 40.32 (2x CH 3 ), 27.20 (3x CH 3 ), 24.59 (CH 2 ), 21.84 (CH 2 ); MS (ES +ve) [Μ+ΗΓ: 51 .0; HRMS (ES +ve), C 26 H 39 N 6 0 3 (M+H) + : calculated

511.31396, found 511.31140.

Bilogical Methods & Materials

Cell Culture General

Cells were grown in Dulbecco's Modified Eagle Medium (DMEM) or Roswell Park Memorial Institute (RPMI) medium supplemented with serum (10% fetal bovine serum) and L-glutamine (2 mM) and incubated in a Heracell 240i tissue culture incubator at 37 °C and 5 % C0 2 . Cell Viability Assay

Cells were plated in 96-weli plates at 2000 -cells/well in 100 μΙ of DMEM or RPMI medium containing 10% FBS and 2 mM L-giutamine and incubated for 48 h in an incubator at 37 °G and 5% C0 2 , After 48 hours, the media was aspirated from each well and replaced with 95 μ! of fresh medium. Compounds, inciudirig DMSO, were prepared at 20X in DM EM medium in a separate 96-weiT intermediate plate. 5 pi from the intermediate plate was then added to each well containing^ ceils. Untreated ceils were incubated with DMSO (0.1% v/v). After 5 days, Presto Blue™ cell viability reagent (10 μ!) was added to each well and the piafes incubated for 60 - 90 min. Fluorescence emission was detected using a fluorescence plate reader (excitation 540 nm, emission 590 nm). Ail conditions were normalised to the untreated cells (100%) and dose- response curves were fitted using GraphPad Prism software.

Apoptosis Assay - Using the IncuCyte-ZOO ® System from Essen Bioscience Celts were plated in 96-wei! Nunc™ black optical-bottom plates (Thermo Scientific) at 3000 ceils/well in 100 μ! of DMEM or RPMI medium containing 10% FBS and 2 mM L~ glutamine and: incubated for 48h in an incubator at 37 °C and 5% G0 2 . The media was replaced with 95 μί of fresh media containing NueView™ 488 at 1 μΜ concentration and drugs or DMSO added along a concentration gradient, as described in the cell viability assay, and the plates placed in the IncuCyte for incubation. Ceil growth and apoptosls was monitored over 5 days using brighffie!d and green fluorescenc channels

(excitation 460 nm, emission 524 nm) microscopy. Cell confluence (brighfieid) and apoptotlc (green) count was performed by the incuGyie software. Cell Cycle Assay

Ceils were plated at 5000 cell/well in 100 μΙ of DMEM or RPMi medium containing 10% FBS and 2 mM L-giutamine in 98-well Nunc™ black optical-bottom plates and incubated at 37 °C with 5% CO a , After 48 hours, the media was aspirated off and replaced with 95 μΙ of fresh media. Compounds, including DMSO, were prepared at 20X in DMEM or RMP! medium in a separate plate and 5 μ ' Ι then added to each well containing ceils. Untreated cells were incubated with DMSO (0.1% v/v). The ceils were incubated for 24 hours at 37 °C then 100 μ! .of 8% PFA in PBS was added to each well and left to incubate at room temperature for 20 mins. The media/PF was removed and wei!s washed with 100 μΙ of PBS ( 3). 100 μ| of blocking buffer (PBS containing 1.1% BS and 0,2% Trixton XI 00) was added to each well and left for 30 minutes. 30 μί of a primary antibody solution containing anti-Cyclin B1 mixed mouse monoclonal antibody (1:300) and anti-pHH3 rabbit polyclonal antibody (1:800) was added to each well and plates incubated for one hour at room temperature. The solution was then removed and wells washed with 100 μΙ of blocking buffer (x3). 100 μ[ of blocking buffer was added to each well and plates incubated for 30 minutes at room temperature. 30 μΙ of a secondary antibody solution containing 4 pg/ml of DAPI, AlexaFluor® 488 Donkey anti- mouse antibody (1 :500) and AlexaFluor® 594 Goat anti-rabbit antibody (1 :500) was added to each well and left to incubate for 45 mins at room temperature in the dark. The solution was then removed and plates washed with 100 μΙ of PBS (x3) and stored in 100 μΙ PBS in the dark until imaged. Images were acquired using the scan A R fluorescence microscope from Olympus or ImageXpress System from Molecular Devices and analysed using scan A R or ImageXpress software. Cells were sorted according to their cell cycle state by DNA content and intensity. Western Blotting Protocol

Cells were plated at 1x10 Θ cells/weil in 2 ml of DMEM or RPMI medium containing 10% FBS and 2 mM L-glutamine in 6-well plates and incubated at 37 °C with 5% C0 2 . After 24 hours, the media was aspirated and replaced with 2 ml of DMEM medium containing 0.1 % FBS and 2 mM L-glutamine and the cells incubated for a further 24 hours. 2 μΙ of compounds dissolved in DMSO at appropriate concentration was then added to each well and plates incubated for 30 mins. 222 μΙ of FBS was then added to each well (giving a final concentration of 10%) and cells incubated for one hour. Cell lysates were then prepared using 100 μΙ of MD Anderson lysate buffer per well. The total cell protein concentration in each lysate was determined using Precision Red Advanced Protein Reagent #2 from Cytoskeleton. 25 μΙ of SDS-PAGE sample loading buffer, 10 μΙ of 1 M DDT, lysate and water up to 100 μΙ to give solutions of 2-3 mg/ml were boiled at 100 °C for 3 mins. Samples were subjected to SDS-PAGE on BioRad 4-15% precast gels over 60 mins at 140 V and transferred to PVDF membranes over 150 mins at 210 mA.

Membranes were blocked for an hour at room temperature using Roche's blocking buffer then primary antibodies added in 0.5 % blocking buffer at 4°C overnight.

Membranes were washed with TBS T (x3, 5 mins) then secondary antibody linked to horseradish peroxidase (HRP) added for an hour at room temperature. Following further washing with TBS T (x3, 5 mins) and TBS (x2, 5 mins) HRP was detected by peroxidase enhanced chemiluminescence (POD ECL from Roche) and bands visualised using X-ray film or the ChemiDoc™ MP Imaging System from BioRad. Cell migration Assay

Ceils were plated at 50000 cells/well in 100 μ! of D ' EM or ' RPMI medium containing 10% PBS and 2 mM L-gfuiamine in a Q6~we ! imagetock plate from Essen Bioscience and left overnight to adhere in an incubator at 37 °C and 5% CO ? . Scratch wounds were created ' in each well using the oundMaker™ supplied by Essen Bioscience and each e(l washed wit media (100 pi, x2) to remove floating cells. 95 μΙ of fresh media was added in each well. Compounds, including DMSO, were prepared at 20X in DivlEiVI medium in a separate plate and 5 μ| then added to each well coniainirig cells. Untreated cells Were incubated with DMSO (0.1% v/v). images were recorded every 30 mins using the IncuGyte-ZQOlvl™ for 24 hours. Analysis of wound width to monito cell migration was performed using the IncuCyte software.

Zeforaf ish Assay

Wild-type zebrafish embryos were collected from AB-TPL breeding pairs and reared at 28 °C in E3 embryo media. The embryos, 2 days post fertilisation (dpf), were treated with compound S06 or Dasatinib at iOOpM, and D!vlSO (0,1 % v/v) as negative control, for 2 hours prior to tali amputation. The tails were then clipped from the embryos. The embryos were incubated with drug for a further 2 hours before being washed off and replaced with fresh E3 media. The embryos were left to develop in E3 media at 28 °C for 2 days, after which, they were imaged by phas contrast microscopy.

Kinase Screening Assay

Compound IC 5 g values were determined from 10-pcint, 1 :3 dilution curves starting at either 100 or 10 M with 10pM ATP, by Reaction Biolog Corp. For the whole kinome screen compounds were screened against 340 wild type kinases at a single dos of 1 μ , in duplicate, with 10 pSVI of ATP by Reaction Biology Corp. The data was averaged and plotted as percentage enzyme activity relative to DMSQ, as negative control, using DiscoverRX TREEspot™ software. Zebrafish PD toxicology assay

Transgenic eldnb:EGFP zebrafish embryos were collected from breeding pairs and reared at 28 °C in E3 embryo media. 1 dpf embryos were treated with 506 or dasatinib at different doses (10-7S0 μ ) at 20 hpf, 36 hpf and 48 hpf, or DMSQ (0.1. % v/v), Zebrafish embryos were imaged by fluorescent microscopy at 72 hpf. Safety assays. Wild-type zebrafish embryos were collected from AB-TPL breeding pairs and reared at 28 °C in E3 embryo media. 1 dpf embryos were treated with 506 or dasatinib at 100μΜ, and DMSO {0.1 % v/v) as negative control, for 4 h before being washed off and replaced with fresh E3 media. For PP20 treatment, the fish were incubated for 2 h post- amputation then replaced with fresh E3 media. The embryos were left to develop in E3 media at 28 °C for 2 d, after which, they were imaged by light microscopy. Zebrafish husbandry was performed under Home Office License in compliance with the Animals (Scientific Procedures) Act 1986 and approved by the University of Edinburgh Ethics Committee. In vivo PD Study

Tumor xenografts were generated in mice by injection of 2 million HCT116 cells subcutaneously. Tumors were allowed to grow until 3-4mm in diameter. A daily dose of 50 mg/kg of 506 in pure water was administered by oral gavage. Mice were sacrificed 3 h after the last dose and tumors excised, fixed in 4% formaldehyde in 0.1 M phosphate buffer (pH 7.2), and embedded in paraffin. Sections were cut using a Reichert-Jung 1150/Autocut microtome to perform phospho-SRC immunochemistry. Antigen retrieval was performed using heat treatment under pressure in a microwave oven for 10 min in 0 mM citrate buffer pH=6. Sections were blocked for endogenous peroxidase followed by incubation with anti-phospho-SRC antibody (Cell Signaling Technology) (1:200 dilution) at 4°C overnight. Staining was developed using EnVision (Dako) and diaminobenzidene (Dako) before slides were counterstained in haematoxylin, dehydrated and mounted in DPX. Slices were imaged on a NanoZoomer digital slide scanner, Hammamatsu. Staining was scored by a single experienced observer, blinded to treatment, using a weighted histoscore method.

Various modifications and variations of the described aspects of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes of carrying out the invention which are obvious to those skilled in the relevant fields are intended to be within the scope of the following claims. Table 1 : Activities of Dasatsnib and inhibitors with high SRC-over-ABL selectivity

ECso = ~ > 10 pM > + 1 Μ > ÷+ > 0.1 μΜ > +++

IC.;,o = - > 1.0 > -f 100 nfvl ++ 10 rM > ÷++

75

TABLE 2: Activities of Dasat niSb and structures related to the novel inhibitors- but exhibiting low antiproliferative properties or low SftG-over- L selectivity

EC50 = - > 0 μΜ ' > + > 1 μίνΐ ++ > 0.1 μΜ +++

!CsG - - > 0 μΜ > + > 100 n ++ > 1 nU > +++

Table 3: IC 50 (nM) values calculated for Dasatinib, compound 506, and compound 503 with a selection of recombinant kinases.

. Table 4: Ceil Screening fEC 5(5 *)

* EC½: 8-point half-log dose response profiling against cell viabiliiy endpoint

Table S: Kinase Screenin (I( ) {*} Sre Family

IC5G = - > 1.0 μΜ > + > 00 nM > +*· > 10 nM > - -÷

TafoSe 6: PhysicGchernicaS & ADS$E Properties

* % of unmodified drug after 30 min incubation with liver microsomes. Controls:

Lidocaine 92.0 %; Propanolol 68.1 %; Verapamil 8.1 %.

Table 7: hERG Channel Inhibition, Stability in Plasma and Oral Bioavailability

* % of unmodified drug after 120 min in plasma. Controls: Eucatropine 10.5 % (human) and 20.3 (mouse); Diitiazem 21.5 % (rat).

Table 8: CYP P450 inhibition values at 10 μΜ. Ail the compounds led to less than

50% inhibition of metabolic activity at the concentration tested.

Properties 506 518 519 533 553 565

CYP450

1A2 (% 24.6 % 6.9 % -1.6 % 8.3 % N/D 4.3 % inhibition)

CYP450

3A4 (% -74.6 % -29.8 % -12.1 % 3.1 % N/D 5.7 % inhibition)

CYP450

2C9 (% -23.1 % -92.3 % -70.3 % -35.4 % N/D -35.8 % inhibition)

CYP450

2D6 (% 31.8 % 6.7 % 3.0 % 7.4 % N/D 9.4 % inhibition)

CYP450

2C19 (% -5.4 % -6.5 % -3.1 % 26.8 % N/D -0.4 % inhibition) References

1. Hughes, J. P Re.es, S., Kalindjian, S. B. & Philpott, K. L. Principles of early drug discovery. Sr. J, Pharmacol. 162, 1239-1249 (2011 ).

2. Smith, A. Screening for drug discovery. Nature 418, 451-483 (2002).

3. Holdgaie, G. ©I al. Biophysical Methods in Drug Discovery from Smalt Molecule to Pharmaceutical Methods Mai. Βίοί 008, 327-355 (2013).

4. Mcinnes, C. Virtual screening strategies in drug discovery. Curr. Qpin. Ghem, Biol. 1 1 , 494-502 (2007).

5. Visi h, G,, Pedretti. A. & Testa, B, Assessing drug-!lkeness-what are we missing? Drug Discov, Today 13, 285-29 (2008).

6. Kamb, A., Wee, S. & Lengauer, C, Why is cancer drug discovery so difficult? Nat. Rev, Drug Diseov. e, 115-20 (2007),

7. Chin, L. & Gray, J, W. Translating insights from the cancer genome into clinical practice. Nature 452, 553-563 (2008).

8. Stomme!, J. M, et al. Coactivafion of receptor tyrosine kinases affects the response of tumor ceils to targeted therapies. Science 318, 287-280 (2007),

9. Garragher, N., Unciti-Broceia, A. & Cameron, D. Advancing cancer drug discovery towards more agile development of targeted combination therapies. Put Med. CI mm, 4, 87- 05 (2012),

10. Knight, Z. A. Lin, H. & Shokat, K. M, Targeting the cancer kinome through poiypharmacology< Nat Rev. Cancer., 10. 30- 37 (2010).

11. Kola, I, & Lahdis, J. Can the pharmaceutical industry reduce attrition rates? Nat Rev. Drug Discov, 3, 7 1-716 (2G04).

12. Sassoon, I. & Blanc, V. Antibody-drug conjugate (ADC) clinical pipeline: a review. Methods Mol. Biol. 1045, 1 -27 (2013).

13. Velema, W, A,, Szymanski, W. & Feringa, B, L. PhQtopharmacology: beyond proof of principle. J. Am, Chem. Soc. 136, 2 78-219 (2014),

14. Versieegen, R, M., Rossin, R., ten Hoeve, ,, jaossen H, M, & Robil!ard, . S. Click to Release: Instantaneous Doxorubicin Elimination upon Tetrazine Ligation,

Angew. Chem. !rit Ed. Engl. 52, 141 12-14 18 (2013).

15. Clave!, C, , et at. Thermoresponsive Chlorambucil Derivatives for Tumour

Targeting. Angew, Ch&m,, Int. Ed. 50, 7124-7127 (2Q1 1),

18. Weiss, J, T. et ai. Extracellular palladium-catalyzed dealkylatien of S-fiuoro- - propargyl-uraci! as a bioorthogonally-aetivafed prodrug approach. Nat, Commun. &, 3277 (2014). 17. Weiss, J. T. et al. Development and Bioorthogonal Activation of Palladium-Labile Prodrugs of Gemcitabine. J. Med. Chem, 57, 5395-5404 (2014).

18. Lee, J. A.; Uhlik, M. T.; oxham, C. M.; Tomandl, D. & Sail, D. J. Modern phenotypic drug discovery is a viable, neoclassic pharma strategy. J, Med. Chem. 55, 4527-4538 (2012).

19. Eder, J., Sedrani, R. & Wiesmann, C. The discovery of first-in-class drugs: origins and evolution Nat. Rev. Drug Discov. 13, 577-587 (2014).

20. Carragher, N. O., Brunton, V. G. & Frame, M. C. Combining imaging and pathway profiling: an alternative approach to cancer drug discovery. Drug Discov. Today 17, 203-214 (2012).

21. Cohen, P. Protein kinases-the major drug targets of the twenty-first century? Nat. Rev. Drug. Discov. 1 , 309-315 (2002).

22. Zhang, J.; Yang, P. L. & Gray, N. S. Targeting cancer with small molecule kinase inhibitors. Nat. Rev. Cancer. 9, 28-39 (2009).

23. Lu, L. et al. Hippo signaling is a potent in vivo growth and tumor suppressor pathway in the mammalian liver. Proc. Natl. Acad. Sci. USA 107, 1437-1442 (2010).

24. Zhou, B.-B. S. & Bartek, J. Targeting the checkpoint kinases: chemosensitization versus chemoprotection. Nat. Rev. Cancer 4, 216-225 (2004).

25. Fujimoto, H. ef al. Regulation of the antioncogenic Chk2 kinase by the oncogenic Wip1 phosphatase. Cell Death Differ. 13, 170-1180 (2006).

26. Wilhelm, S. et al. Discovery and development of sorafenib: a multikinase inhibitor for treating cancer. Nat. Rev. Drug Discov. 5, 835-844 (2006).

27. Greuber, E. K.; Smith-Pearson, P.; Wang, J. & Pendergast, A. M. Role of ABL family kinases in cancer: from leukaemia to solid tumours. Nat. Rev. Cancer. 13, 559- 571 (2013).

28. Noren, N. K.; Foos, G.; Hauser, C. A. & Pasquale, E. B. The EphB4 receptor suppresses breast cancer cell tumorigenicity through an Abl-Crk pathway. Nat. Cell Biol. 8, 815-825 (2006).

29. Allington, T. M.; Galliher-Beckley, A. J. & Schiemann, W. P. Activated Abl kinase inhibits oncogenic transforming growth factor-beta signaling and tumorigenesis in mammary tumors. FASEB J. 23, 4231-4243 (2009).

30. Gil-Henn, H. ef al. Arg/Abl2 promotes invasion and attenuates proliferation of breast cancer in vivo. Oncogene 32, 2622-2630 (2013). 31. Hanke s J. H, et a/. Discovery of a novel, potent, and Src family-selective tyrosine kinase inhibitor. Study of Lck- and FynT-de endent T cell activation, J Biol. Chem. 271, 695-701 (1996).

32. Tatfon, L, Mofiey, G. . f Chopra, R. & waja, A. The Src-selective kinase inhibitor PP1 also inhibits Kit and Bcr-Abl tyrosine kinases. J. Biol. Gherri. 278, 4847-4853 (2003).

33. Jester, B. ., Gaj, A., Shornin, C, D., Cox, K. J. & Ghosh, !. Testing the promiscuity of commercial kinase inhibitors against the AGO kinase group using a spiif-iuciferas screen. J Med. Chem. 55, 1626-1537 (2012),

34. Diner, P., Aiao. j. P., So erlund, j,, Sunner tagen, P, & Gr0tlf, M, preparation of 3- subsittuted-1-isopropyi- H-pyrazoio[3,4-d]pyrimM as RET kinase inhibitors. J Med. Chem. 55, 4872-4876 (2012).

35. Apsel, B, et al. Targeted po!ypharmacology: discovery of dual, inhibitors of tyrosine and phosphoinositide. kinases, Nat, Chem. Biol. 4, 691-899 (2008),

38, AntoneHi, A,, et al, CLM29, a multi-target pyrazGiopynmidine derivative, has antineoplastic activity in medullary thyroid cancer in vitro and in vivo. Mo!. Ceii Endocrinol. 393, 56-64 (2014).

37. Sehindier,. T. et al. Crystal structure of Hick in complex with. a Src fa ily-seieetive tyrosine kinase inhibitor, Moi. Ceil. 3, 639-648 (1999).

38. nowies, P. P. et aL Structure and chemical inhibition of the RET tyrosine kinase domain. J Biol. Chem. 281.,.: 33577-87 (2006).

39. Lsu, Y, & Gray, N, S. Rational design of inhibitors that bind to inactive kinase conformations. Nat Chem. Biol. 2, 358-364 (2GQ6).

40. Liu, Y. et al Structural basis for selective inhibition of Src family kinases by PPL. Chem Biol 6, 671-678 ( 999).

41 Dar, A. C, Lopez, M. S. & Shokat, K. M, Small molecule recognition of c-Src via the imatinib-.bin.ding conformation, Qhern. Biol. 15, 1015-1022 (2008).

42. Hann, !Vl M, & Keseru, G, M. Finding the sweet spot: the role of nature and nurture in medicinal chemistry, Hat, Rev. Drug Discov, 1i . 355-365 (2012),

43. Hsieh, A, C. ef a/. The translational landscape of mTOR signalling steers cancer initiation and metastasis. Nature 485, 55-6 (2012).

44. Kerkela R , Grazette L, Yacobi R, Siiescu G, Patten R, Beahm C, Waiters B,

Shevtsov S, Pesant 8, Ciubb FJ, Rosenzweig A, Salomon RN, Van Etten RA, Alroy J, Puraod JB, Force T. (2008), Cardiotoxicity of th cancer therapeutic agent imatinib mesylate. Nature Med Aug;12(8):908-16, 45. Aliington, T. M.; Galliher-Beckley, A. J.; Schiemann, W. P. Activated Abl kinase inhibits oncogenic transforming growth factor-beta signaling and tumorigenesis in mammary tumors. FASEB J. 2009, 23, 4231-4243.

46. Boyce, B.F., Xing, L, Yao, Z., Yamashita, T., Shakespeare, W.C., Wang, Y, Metcalf, C.A., 3rd, Sundaramoorthi, R., Dalgarno, D.C., luliucci, J.D., and Sawyer, T.K. (2006). SRC inhibitors in metastatic bone disease. Clin Cancer Res 12, 629 s-6295s.

47. Brunton, V.G., and Frame, M.C. (2008). Src and focal adhesion kinase as therapeutic targets in cancer. Curr Opin Pharmacol 8, 427-432.

48. Chaturvedi, D., Gao, X., Cohen, M.S., Taunton, J., and Patel, T.B. (2009).

Rapamycin induces transactivation of the EGFR and increases cell survival. Oncogene 28, 1187-1 196.

49. Creedon, H., and Brunton, V.G. (2012). Src kinase inhibitors: promising cancer therapeutics? Crit Rev Oncog 17, 145-159.

50. Girotti R et al. (2015). Paradox-Breaking RAF Inhibitors that Also Target SRC Are Effective in Drug-Resistant BRAF Mutant Melanoma. Cancer Cell 27(1): 85-96.

51. Kerkela R, Grazette L, Yacobi R, lliescu C, Patten R, Beahm C, Walters B, Shevtsov S, Pesant S, Clubb FJ, Rosenzweig A, Salomon RN, Van Etten RA, Alroy J, Durand JB, Force T. (2006). Cardiotoxicity of the cancer therapeutic agent imatinib mesylate. Nature Med 12(8):908-16.

52. Qiu Z, Cang Y, Goff SP. (2010). Abl family tyrosine kinases are essential for basement membrane integrity and cortical lamination in the cerebellum.. J Neurosci. 2010 Oct 27;30(43): 14430-9.

53. De Wispelaere, M,, Lacroix, A.J., and Yang, P.L. (2013). The small molecules AZD0530 and dasatinib inhibit dengue virus RNA replication via Fyn kinase. J Virol 87, 7367-7381.

54. Duxbury, M.S., !to, H., Zinner, M.J., Ashley, S.W., and Whang, E.E. (2004).

Inhibition of SRC tyrosine kinase impairs inherent and acquired gemcitabine resistance in human pancreatic adenocarcinoma cells. Clin Cancer Res 10, 2307-2318.

55. Gi!-Henn, H.; Patsialou, A.; Wang, Y.; Warren, M. S.; Condeelis, J. S.; Ko!eske, A. J. Arg/Abl2 promotes invasion and attenuates proliferation of breast cancer in vivo. Oncogene, 2013, 32, 2622-2630.

56. Hawthorne, V.S., Huang, W.C., Neal, C.L, Tseng, L.M., Hung, M.C, and Yu, D. (2009). ErbB2-mediated Src and signal transducer and activator of transcription 3 activation leads to transcriptional up-regulation of p21Cip1 and chemoresistance in breast cancer cells. Mol Cancer Res 7, 592-600. 57. Kopefz, S,, Lesslie, D P,, Dallas, N.A.., Park, S.L, Johnson, M., Parikh, N.U., Kim, .P., Abbruzzese, J.L, Ellis, LM, Chandra, J., and Gal!ick, G.E, (2009), Synergistic activity of the SRC family kinase inhibiior dasafinib and Qxaliplatin in colon carcinoma cells is mediated by oxidative stress. Cancer Res 6.8, 3842-3849,

58. Mayer, E.L, and rop, LE. (2010). Advances in targeting SRC in the treatment of breast cancer and other solid malignancies. Clin Cancer Res 18, 3528-3532.

59. McCarthy, S.D., Jung, D., Sakac, D., and Branch, D.R. (2014). c-Src and Pyk2 protein tyrosine kinases play protective roles in early HIV-1 infection of CD4÷ T-cell lines, J Acqui immune Defie Syndr 86, 118-126.

80. lyiurriSis, R.J., Fukayama, S., Bosehel!i, F., IVlatteo, J J., Owens, J., Golas, j. ., Patei, D., Lane, G ' ., Liu, Y.8., Garter, L, Jussif, J,, SpaxiSding, V,, Wang, Y.P., Bosenelli, D.H,, iVickew, J.G., Li, X.J,, tOGkhead, S., iiligan, C. t Kharode, Y.P., Dies!, V„ Bai, Y., Foliettie, M,, Bex, FJ., Kornm, B., and Bodine, P.M. (2012). Osteogenic effects of a potent Src-over~Abl~selective kinase inhibitor in the mouse. J Pharmacol Exp The 340, 678-887,

61. Myaui, A., fSJishimura, R, Williams, P. J., Hiraga, T,, Taroura, D. . . Michjgami. T., Mundy, G,R, and Yoneda, T. (2003), G-SRC tyrosine kinase activity is associated vvith tumor colonization in bone and lung in an animal model of huma breast cancer metastasis. Cancer Res 63, 5028-5033,

82, Noren, N, It; FOGS, G.; Rauser, C, A,; Pasquale, E. B (2008). The EphB4 receptor suppresses breast cancer ceil tumorigenicity through an Abl-Crk pathway. Nat. Ceil Biol. 8, 815-825.

63. Park, G.B., Kim, D. s Kim, Y.S., Kim, S„ Lee, H.K., Yang, J.W., and Hur, D.Y. (2014). The Epstein-Barr virus causes- epithelial-flfiesenchymal " transition in human corneal epithelial cells via Syk/src and Akt/Erk signaling pathways, invest Ophthalmol Vis Sci 55, 1770-1779.

64. Sawyers, C.L, : Msl.aughlin, J,, Goga, A., Havfik, M„ and Witte, G. (1994). The nuclear tyrosine kinase c-Abl negatively regulates cell growth, Gel! 77, 121-131 ,

65. Wheeler, D.L. , lida, M,, Kruser, T.J., Nechrebecki, IVLlvIj Dunn, E.F., Armstrong, Ε,Α,, Huang, 8., and Harari, P.M. (2009). Epidermal growth factor receptor cooperates with Src family kinases in acquired resistance to cetuxlrnab. Cancer Biol Ther 8, 696- 703,

66. Zhang, S., Huang, W.C., Li, P., Guo, R, Pon, S.B., Brady, S.W., Xiong, Y., Tseng, LM., Li, S.H., Ding, , Sahin, A A, Esteva, F.J., Hortobagyi, G.N., and Yu, D. (20 ), Combating trastuzumab resistance by targeting SRC, a common node downstream of multiple resistance pathways. Nat Med 7, 461-469.

67. Summy JM, Gallick GE (2003). Src family kinases in tumor progression and metastasis. Cancer Metastasis Rev. 22(4):337-58.

68. Frame MC et ai. (2002). v-Src's hold over actin and celi adhesions. Nat Rev Mo! Cell Biol. 3(4):233-45.

69. Yoo SK et al. (2012) Early redox, Src family kinase, and calcium signaling integrate wound responses and tissue regeneration in zebrafish. J Cell Biol 199, 225-234.

70. Tyryshkin A et al., SRC kinase is a novel therapeutic target in

lymphangioleiomyomatosis. Cancer Res; 74(7) April 1 , 2014.

71. Walcher D et al., C-Peptide induces vascular smooth muscle celi proliferation: involvement of SRC-kinase, phosphatidylinositol 3-kinase, and extracellular signal- regulated kinase 1/2. Circulation Research. 2006; 99: 1 181 -1 187.

72. Cho HM et al., The Src/PLC/PKC/MEK/ERK signaling pathway is involved in aortic smooth muscle ceil proliferation induced by glycated LDL. Molecules and Cells [2005, 19(1):60-66].

73. Yang K1 , Belrose J, Trepanier CH, Lei G, Jackson MF, MacDonald JF. Fyn, a potential target for Alzheimer's disease. J Alzheimers Dis. 2011 , 27, 243-52.

74. Nygaard HB, van Dyck CH, Strittmaiter SM. Fyn kinase inhibition as a novel therapy for Alzheimer's disease. Alzheimers Res Ther. 2014, 6, 8.

75. Kaufman AC1 , Salazar SV, Haas LT, Yang J, Kostylev MA, Jeng AT, Robinson SA, Gunther EC, van Dyck CH, Nygaard HB, Strittmaiter SM. Fyn inhibition rescues established memory and synapse loss in Alzheimer mice. Ann Neurol. 2015, doi:

10.1002/ana.24394 [ahead of print].

76. Nygaard HB, Wagner AF, Bowen GS, Good SP, MacAvoy MG, Strittmatter KA, Kaufman AC, Rosenberg BJ, Sekine-Konno T, Varma P, Chen K, Koleske AJ, Reiman EM, Strittmatter SM, van Dyck CH. A phase lb multiple ascending dose study of the safety, tolerability, and central nervous system availability of AZD0530 (saracatinib) in Alzheimer's disease. Alzheimers Res Ther. 2015, 7, 35.

77. H. Ma, S. Deacon & K. Horiuchi. The challenge of selecting protein kinase assays for lead discovery optimization. Expert Opin. Drug Discov. (2008) 3(6).

78. Patton, E. E.; Dhillon, P.; Amatruda, J. F.; Ramakrishnan, L. Spotlight on zebrafish: translational impact. Dis. Model. Mech. 2014, 7, 731 -733.

79. Gallardo, V. E.; Varshney, G. K.; Lee, M.; Bupp, S.; Xu, L.; Shinn, P.; Crawford, N. P.; Inglese, J.; Burgess, S. M. Phenotype-driven chemical screening in zebrafish for compounds thai inhibit collective ceil rnigration identifies multiple pathways potentially involved in metastatic invasion. Dk, Model. /Wee/?, 2015, 8, 565-576.

80. Xiao, T.: Roeser, T.; Stau ' h, W.; Baler H. A GFP-feased genetic screen reveals mutations that disrupt the architecture of the zebrafish retinotectai projection.

Development 2005, 132, 2955-2967,

81. Gfu, Z ; Gang, Y,; Goff, S. P. c-Ab! tyrosine kinase regulates cardiac growth and development, Proc. Naff. Acad Set USA 2010, 107, 1136-1141.

82. Chis!ock, E. M.; Ring, C; Pendergast, A. M. Ab! kinases are required for vascular funciion, Tse2 expression, and angiopoietin-l -mediated survivai. Pwc, Nail, Acad. Sci, USA 2013, 110, 12432-12417.

83. Animal experiments were performed under Home Office License In compliance with the Animals (Scientific Procedures) Act 1988 and approved by the University of Edinburgh Ethics Committee,