Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
POLYCYCLIC AMIDES AS CYTOTOXIC AGENTS
Document Type and Number:
WIPO Patent Application WO/2020/049286
Kind Code:
A1
Abstract:
The invention relates to a compound of formula (I): or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof; wherein the fused ring moiety is a non-alkylating moiety; and wherein the compounds are useful as medicaments, in particular for use as a drug in an antibody-drug conjugate and in the treatment of a proliferative disease, a bacterial infection, a malarial infection and inflammation.

Inventors:
THURSTON DAVID EDWIN (GB)
JACKSON PAUL JOSEPH MARK (GB)
Application Number:
PCT/GB2019/052445
Publication Date:
March 12, 2020
Filing Date:
September 03, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
FEMTOGENIX LTD (GB)
International Classes:
C07D405/14; A61K31/5513; A61P35/00; C07D471/04; C07D487/04; C07K5/062
Domestic Patent References:
WO2017032983A12017-03-02
WO2017223275A12017-12-28
WO2017201132A22017-11-23
WO2017098257A12017-06-15
WO1998017797A11998-04-30
WO2016040723A12016-03-17
WO1990008187A11990-07-26
WO1990011294A11990-10-04
WO1991001133A11991-02-07
WO2009052249A12009-04-23
WO2004063362A22004-07-29
WO2003042661A22003-05-22
WO2002102235A22002-12-27
WO2003055443A22003-07-10
WO2002099122A12002-12-12
WO2003029421A22003-04-10
WO2003024392A22003-03-27
WO2002098358A22002-12-12
WO2002054940A22002-07-18
WO2002059377A22002-08-01
WO2002030268A22002-04-18
WO2001048204A12001-07-05
WO2004048938A22004-06-10
WO2004032842A22004-04-22
WO2003016475A22003-02-27
WO2002078524A22002-10-10
WO2002099074A22002-12-12
WO2002086443A22002-10-31
WO2003003906A22003-01-16
WO2002064798A12002-08-22
WO2000014228A12000-03-16
WO2003025138A22003-03-27
WO2004065577A22004-08-05
WO2004027049A22004-04-01
WO2004016225A22004-02-26
WO2002089747A22002-11-14
WO2003022995A22003-03-20
WO2004045553A22004-06-03
WO2002092836A22002-11-21
WO2002083866A22002-10-24
WO2003101283A22003-12-11
WO2002101075A22002-12-19
WO2002071928A22002-09-19
WO1994010312A11994-05-11
WO2004022778A12004-03-18
WO2001057188A22001-08-09
WO2001075177A22001-10-11
WO2004000997A22003-12-31
WO2003003984A22003-01-16
WO2002006339A22002-01-24
WO2001088133A22001-11-22
WO2003054152A22003-07-03
WO2003101400A22003-12-11
WO2003105758A22003-12-24
WO2003025148A22003-03-27
WO2004045516A22004-06-03
WO2004040000A22004-05-13
WO2003087768A22003-10-23
WO2002061087A22002-08-08
WO2003016494A22003-02-27
WO2001098351A22001-12-27
WO2001077172A22001-10-18
WO2004001004A22003-12-31
WO2003104275A22003-12-18
WO2004046342A22004-06-03
WO2003083074A22003-10-09
WO2003018621A22003-03-06
WO2001066689A22001-09-13
WO2003087306A22003-10-23
WO2002072596A12002-09-19
WO2001072962A22001-10-04
WO2003104270A22003-12-18
WO2002026822A22002-04-04
WO2002016429A22002-02-28
WO2000040614A22000-07-13
WO2002010382A22002-02-07
WO2001062794A22001-08-30
WO2003083041A22003-10-09
WO2003034984A22003-05-01
WO2002088170A22002-11-07
WO2002016413A22002-02-28
WO2002022808A22002-03-21
WO2004045520A22004-06-03
WO2003062401A22003-07-31
WO1991002536A11991-03-07
WO2004020595A22004-03-11
WO2003048202A22003-06-12
WO1999058658A21999-11-18
WO2000055351A12000-09-21
WO2003077836A22003-09-25
WO2001038490A22001-05-31
WO2003097803A22003-11-27
WO2003089624A22003-10-30
WO2004009622A22004-01-29
WO2003081210A22003-10-02
WO2003089904A22003-10-30
WO2003008537A22003-01-30
WO2003055439A22003-07-10
WO2003025228A12003-03-27
WO2002022636A12002-03-21
WO2002012341A22002-02-14
WO2002013847A22002-02-21
WO2002014503A22002-02-21
WO2001053463A22001-07-26
WO2001041787A12001-06-14
WO2000044899A12000-08-03
WO2000020579A12000-04-13
WO1996030514A11996-10-03
WO2004043361A22004-05-27
WO2004022709A22004-03-18
WO2001000244A22001-01-04
WO2004063709A22004-07-29
WO2004044178A22004-05-27
WO2004031238A22004-04-15
WO2002060317A22002-08-08
WO1999046284A21999-09-16
WO2003029262A22003-04-10
WO2003002717A22003-01-09
WO2002022153A22002-03-21
WO2001046261A12001-06-28
WO2001046232A22001-06-28
WO1998037193A11998-08-27
WO2002002634A22002-01-10
WO2000053216A22000-09-14
WO2004065576A22004-08-05
WO2004020583A22004-03-11
WO2003004529A22003-01-16
WO2003104399A22003-12-18
WO2004000221A22003-12-31
WO2002010187A12002-02-07
WO2001094641A22001-12-13
WO2002002624A22002-01-10
WO2002006317A22002-01-24
WO2002000258A12002-01-03
WO2001040269A22001-06-07
WO2000036107A22000-06-22
WO2004053079A22004-06-24
WO2003004989A22003-01-16
WO2001016318A22001-03-08
WO2002081646A22002-10-17
WO2001040309A22001-06-07
WO2000032752A12000-06-08
WO1998051805A11998-11-19
WO1998051824A11998-11-19
WO1998040403A11998-09-17
WO2003000842A22003-01-03
WO2003023013A22003-03-20
WO2004058309A12004-07-15
WO2004011611A22004-02-05
WO2003045422A12003-06-05
WO2003014294A22003-02-20
WO2003035846A22003-05-01
WO2002094852A22002-11-28
WO2002038766A22002-05-16
WO2002024909A22002-03-28
WO2003072036A22003-09-04
WO2003088808A22003-10-30
WO1999058658A21999-11-18
WO1992007574A11992-05-14
WO2004015426A12004-02-19
WO2002061087A22002-08-08
WO2001072830A22001-10-04
WO2000022129A12000-04-20
WO1999028468A11999-06-10
WO1994028931A11994-12-22
WO1992017497A11992-10-15
WO2004047749A22004-06-10
WO2003072035A22003-09-04
WO2002022660A22002-03-21
WO2003093444A22003-11-13
WO2003029277A22003-04-10
WO2004042346A22004-05-21
WO2003026493A22003-04-03
WO2000075655A12000-12-14
WO1997007198A21997-02-27
WO2003083047A22003-10-09
WO1997044452A11997-11-27
WO2000012130A12000-03-09
WO2004074320A22004-09-02
WO2003009814A22003-02-06
WO2001090304A22001-11-29
WO2004063355A22004-07-29
WO2013017705A12013-02-07
WO2013177055A22013-11-28
WO1993016185A21993-08-19
WO1993001161A11993-01-21
WO1993008829A11993-05-13
WO1996027011A11996-09-06
WO1998050431A21998-11-12
WO2012106587A12012-08-09
WO2009089004A12009-07-16
WO2008077546A12008-07-03
WO2000061739A12000-10-19
WO2001029246A12001-04-26
WO2003085119A12003-10-16
WO2003084570A12003-10-16
WO2005035586A12005-04-21
WO2005035778A12005-04-21
WO2005053742A12005-06-16
WO2002031140A12002-04-18
WO2004056312A22004-07-08
WO2003085107A12003-10-16
WO2003011878A22003-02-13
WO1997030087A11997-08-21
WO1998058964A11998-12-30
WO1999022764A11999-05-14
WO2006029879A22006-03-23
WO2005100402A12005-10-27
WO1999051642A11999-10-14
WO1994029351A21994-12-22
WO2004043963A12004-05-27
Foreign References:
US20180110873A12018-04-26
US4665077A1987-05-12
US4120649A1978-10-17
US5114721A1992-05-19
EP0340109A21989-11-02
US7595292B22009-09-29
US7985783B22011-07-26
US7754681B22010-07-13
EP0624377A21994-11-17
US7521541B22009-04-21
US7723485B22010-05-25
US4816567A1989-03-28
US20030134790A12003-07-17
US20030224454A12003-12-04
EP1394274A22004-03-03
US20030157089A12003-08-21
US20030185830A12003-10-02
US20030064397A12003-04-03
US20030124140A12003-07-03
US0798959A1905-09-05
EP0875569A11998-11-04
US20030129192A12003-07-10
US20040044180A12004-03-04
US20040044179A12004-03-04
US20030096961A12003-05-22
US20030232056A12003-12-18
US20030206918A12003-11-06
EP1347046A12003-09-24
EP0522868A11993-01-13
US20030109676A12003-06-12
US6518404B12003-02-11
US5773223A1998-06-30
US20040005598A12004-01-08
US20030060612A12003-03-27
US20030143557A12003-07-31
US20030219806A12003-11-27
US20030224411A12003-12-04
US5854399A1998-12-29
US5792616A1998-08-11
US20040005538A12004-01-08
US20040101874A12004-05-27
US20020150573A12002-10-17
US5644033A1997-07-01
US6534482B12003-03-18
US20030118592A12003-06-26
US5869445A1999-02-09
EP1439393A22004-07-21
JPH053790B21993-01-18
US20040005320A12004-01-08
US20020042366A12002-04-11
US20030186372A12003-10-02
US20030186373A12003-10-02
US20030119131A12003-06-26
US20030119122A12003-06-26
US20030119126A12003-06-26
US20030119121A12003-06-26
US20030119129A12003-06-26
US20030119130A12003-06-26
US20030119128A12003-06-26
US20030119125A12003-06-26
US20040101899A12004-05-27
US20030165504A12003-09-04
US20030065143A12003-04-03
US20030091580A12003-05-15
US20020034749A12002-03-21
US20040018553A12004-01-29
US20010055751A12001-12-27
US20030194704A12003-10-16
US20030228319A12003-12-11
US20030105292A12003-06-05
US6555339B12003-04-29
US5440021A1995-08-08
US6153408A2000-11-28
US5976551A1999-11-02
US6011146A2000-01-04
US20020193567A12002-12-19
JP2004113151A2004-04-15
EP1295944A22003-03-26
US20040249130A12004-12-09
US20040022727A12004-02-05
US20040197325A12004-10-07
US20030232350A12003-12-18
US20040005563A12004-01-08
US20030124579A12003-07-03
US5571894A1996-11-05
US5587458A1996-12-24
US5869046A1999-02-09
EP0404097A21990-12-27
US6248516B12001-06-19
US5821337A1998-10-13
US7527791B22009-05-05
US6982321B22006-01-03
US7087409B22006-08-08
US6075181A2000-06-13
US6150584A2000-11-21
US5770429A1998-06-23
US7041870B22006-05-09
US20070061900A12007-03-15
US7189826B22007-03-13
US5750373A1998-05-12
US20050079574A12005-04-14
US20050119455A12005-06-02
US20050266000A12005-12-01
US20070117126A12007-05-24
US20070160598A12007-07-12
US20070237764A12007-10-11
US20070292936A12007-12-20
US20090002360A12009-01-01
US5731168A1998-03-24
US20110287009A12011-11-24
US20070178552A12007-08-02
US20110287009A12011-11-24
US4676980A1987-06-30
US20060025576A12006-02-02
US20080069820A12008-03-20
US20030157108A12003-08-21
US20040093621A12004-05-13
US20030115614A12003-06-19
US20020164328A12002-11-07
US20040132140A12004-07-08
US20040110704A12004-06-10
US20040110282A12004-06-10
US20040109865A12004-06-10
US6602684B12003-08-05
US20050123546A12005-06-09
US5500362A1996-03-19
US8697650B22014-04-15
US6737056B12004-05-18
US7332581B22008-02-19
US8969526B22015-03-03
US6194551B12001-02-27
US20050014934A12005-01-20
US7371826B22008-05-13
US5648260A1997-07-15
US5624821A1997-04-29
US5648237A1997-07-15
US5789199A1998-08-04
US5840523A1998-11-24
US5959177A1999-09-28
US6040498A2000-03-21
US6420548B12002-07-16
US7125978B12006-10-24
US6417429B12002-07-09
Other References:
FLATMAN ET AL., J. CHROMATOGR., vol. B 848, 2007, pages 79 - 87
WU ET AL., NATURE BIOTECH., vol. 23, no. 9, 2005, pages 1137 - 1145
DIJKE,P. ET AL., SCIENCE, vol. 264, no. 5155, 1994, pages 1225 - 104
WEIS J.J. ET AL., J. EXP. MED., vol. 167, 1988, pages 1047 - 1066
FRANKLIN ET AL., CANCER CELL, vol. 5, 2004, pages 317 - 328
OFFNER ET AL., SCIENCE, vol. 251, 1991, pages 430 - 3242
CHOTHIALESK, J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
IANEWAY, NATURE, vol. 341, 1989, pages 482
MACKAYMACKAY, TRENDS IMMUNOL, vol. 23, 2002, pages 113 - 5
DURIE ET AL., SCIENCE, vol. 261, 1993, pages 1328 - 30
MOHAN ET AL., J. IMMUNOL, vol. 154, 1995, pages 1470 - 80
FURUSHIMA, K. ET AL., DEV. BIOL., vol. 306, no. 2, 2007, pages 480 - 492
SINHA S.K. ET AL., J. IMMUNOL., vol. 151, 1993, pages 2296 - 5320
CLACKSON ET AL., NATURE, vol. 352, 1991, pages 624 - 628
JAIN, N.SMITH, S. W.GHONE, S.TOMCZUK, B.: "Current ADC Linker Chemistry", PHARMACEUTICAL RESEARCH, vol. 32, no. 11, 2015, pages 3526 - 3540, XP035553874, doi:10.1007/s11095-015-1657-7
CARTER, P., NATURE REVIEWS IMMUNOLOGY, vol. 6, 2006, pages 343 - 357
XIE ET AL., EXPERT. OPIN. BIOL. THER., vol. 6, no. 3, 2006, pages 281 - 291
KOVTUN, CANCER RES., vol. 66, no. 6, 2006, pages 214 - 3121
ERICKSON ET AL., CANCERRES., vol. 66, no. 8, 2006, pages 2328 - 2337
LAMBERT J., CURRENT OPIN. IN PHARMACOL., vol. 5, 2005, pages 543 - 549
HAMANN P., EXPERT OPIN. THER. PATENTS, vol. 15, no. 9, 2005, pages 1087 - 1103
PAYNE, G., CANCER CELL, vol. 3, 2003, pages 207 - 212
TRAIL, CANCER IMMUNOL. IMMUNOTHER., vol. 52, 2003, pages 328 - 337
SYRIGOSEPENETOS, ANTICANCER RESEARCH, vol. 19, 1999, pages 605 - 614
JUNUTULA ET AL., NATURE BIOTECH., vol. 26, no. 8, 2008, pages 925 - 932
DOMAN ET AL., BLOOD, vol. 114, no. 13, 2009, pages 2721 - 2729
MCDONAGH, PROTEIN ENG. DESIGN & SEL., vol. 19, no. 7, 2006, pages 299 - 307
DORONINA ET AL., BIOCONJ. CHEM., vol. 17, 2006, pages 114 - 124
CLIN. CANCERRES., vol. 11, 2005, pages 843 - 852
JEFFREY ET AL., J. MED. CHEM., vol. 48, 2005, pages 1344 - 1358
HAMBLETT ET AL., CLIN. CANCER RES., vol. 10, 2004, pages 7063 - 7070
CANCER RES., vol. 61, no. 15, 2001, pages 5857 - 5860
"Genbank", Database accession no. NP_001774.10
KOHLER ET AL., NATURE, vol. 256, 1975, pages 495
MARKS ET AL., J. MOL. BIOL., vol. 222, 1991, pages 581 - 597
LONBERG, CURR. OPINION, vol. 20, no. 4, 2008, pages 450 - 459
MORRISON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 81, 1984, pages 6851 - 6855
ONCOGENE, vol. 14, no. 11, 1997, pages 1377 - 1382
FEILD, J.A. ET AL., BIOCHEM. BIOPHYS. RES. COMMUN, vol. 266, no. 3, 1999, pages 593 - 602
NATURE, vol. 395, no. 6699, 1998, pages 288 - 291
GAUGITSCH, H.W. ET AL., J. BIOL. CHEM., vol. 267, no. 16, 1992, pages 11267 - 11273
HUBERT, R.S. ET AL., PROC. NATL. ACAD. SCI. U.SA., vol. 96, no. 20, 1999, pages 11531 - 11536
SHIELDS ET AL., J. BIOL. CHEM., vol. 276, no. 29, 2001, pages 27371 - 27375
YAMAGUCHI, N. ET AL., BIOL. CHEM., vol. 269, no. 2, 1994, pages 805 - 808
MAO ET AL., PROC. NATL. ACAD. SCI. U.SA., vol. 93, no. 1, 1996, pages 5910 - 5914
J. BIOL. CHEM., vol. 270, no. 37, 1995, pages 21984 - 21990
PARRISH-NOVAK J. ET AL., J. BIOL. CHEM., vol. 277, no. 22, 2002, pages 47517 - 47523
GENOMICS, vol. 62, no. 2, 1999, pages 281 - 284
NAGASE T. ET AL., DNA RES., vol. 7, no. 2, 2000, pages 143 - 150
ROSS ET AL., CANCER RES., vol. 62, 2002, pages 2546 - 2553
SAKAMOTO A.YANAGISAWA M. ET AL., BIOCHEM. BIOPHYS. RES. COMMUN, vol. 178, 1991, pages 656 - 663
ARAI H. ET AL., JPN. CIRC. J., vol. 56, 1992, pages 1303 - 1307
ELSHOURBAGY N. A. ET AL., J. BIOL. CHEM., vol. 268, 1993, pages 3873 - 3879
HAENDLER B. ET AL., J. CARDIOVASC. PHARMACOL., vol. 20, 1992
TSUTSUMI M. ET AL., GENE, vol. 228, 1999, pages 43 - 49
STRAUSBERG R.L. ET AL., PROC. NATL. ACAD. SCI. U.SA., vol. 99, no. 26, 2002, pages 16899 - 16903
BOURGEOIS C ET AL., J. CLIN. ENDOCRINOL. METAB., vol. 82, 1997, pages 3116 - 3123
OKAMOTO Y. ET AL., BIOL. CHEM., vol. 272, 1997, pages 21589 - 21596
VERHEIJ J.B. ET AL., AM. J. MED. GENET., vol. 108, 2002, pages 223 - 225
HOFSTRA R.M.W. ET AL., EUR. J. HUM. GENET., vol. 5, 1997, pages 180 - 185
PUFFENBERGER E.G. ET AL., CELL, vol. 79, 1994, pages 1257 - 1266
ATTIE T. ET AL., HUM. MOL. GENET., vol. 4, 1995, pages 2407 - 2409
AURICCHIO A. ET AL., HUM. MOL. GENET., vol. 5, 1996, pages 355 - 357
HOFSTRA R.M.W. ET AL., NAT. GENET., vol. 12, 1996, pages 445 - 447
SVENSSON PJ. ET AL., HUM. GENET., vol. 103, 1998, pages 145 - 148
FUCHS S. ET AL., MOL. MED., vol. 7, 2001, pages 115 - 124
PINGAULT V. ET AL., HUM. GENET., vol. 111, 2002, pages 198 - 206
LAB. INVEST., vol. 82, no. 11, 2002, pages 1573 - 1582
XU, X.Z. ET AL., PROC. NATL. ACAD. SCI. U.SA., vol. 98, no. 17, 2001, pages 10692 - 10697
CELL, vol. 109, no. 3, 2002, pages 397 - 407
J. BIOL. CHEM., vol. 278, no. 33, 2003, pages 30813 - 30820
CICCODICOLA ET AL., EMBO J., vol. 8, no. 7, 1989, pages 1987 - 1991
AM. J. HUM. GENET., vol. 49, no. 3, 1991, pages 555 - 565
FUJISAKU ET AL., J. BIOL. CHEM., vol. 264, no. 4, 1989, pages 2118 - 2125
MOORE M. ET AL., PROC. NATL. ACAD. SCI. U.SA., vol. 84, 1987, pages 9194 - 9198
BAREL M. ET AL., MOL. IMMUNOL., vol. 35, 1998, pages 1025 - 1031
WEIS J.J. ET AL., PROC. NATL. ACAD. SCI. U.SA., vol. 83, 1986, pages 5639 - 5643
"GENBANK", Database accession no. 11038674
ERICSSON, T.A. ET AL., PROC. NATL. ACAD. SCI. U.SA., vol. 100, no. 11, 2003, pages 6759 - 6764
BLOOD, vol. 100, no. 8, 2002, pages 2662 - 2669
DOBNER ET AL., EUR. J. IMMUNOL., vol. 22, no. 6, 1992, pages 2795 - 2799
CLARK, H.F. ET AL., GENOME RES., vol. 13, no. 10, 2003, pages 2265 - 2270
IMMUNOGENETICS, vol. 54, no. 2, 2002, pages 87 - 95
XU, M.J. ET AL., BIOCHEM. BIOPHYS. RES. COMMUN, vol. 280, no. 3, 2001, pages 768 - 775
COUSSENS L. ET AL., SCIENCE, vol. 229, no. 4730, 1985, pages 1132 - 1139
YAMAMOTO T. ET AL., NATURE, vol. 319, 1986, pages 230 - 234
SEMBA K. ET AL., PROC. NATL. ACAD. SCI. U.SA., vol. 82, 1985, pages 6497 - 6501
SWIERCZ J.M. ET AL., J. CELL BIOL., vol. 165, 2004, pages 869 - 880
KUHNS J.J. ET AL., J. BIOL. CHEM., vol. 274, 1999, pages 36422 - 36427
MUNGALL A.J. ET AL., NATURE, vol. 425, 2003, pages 805 - 811
EHSANI A ET AL., GENOMICS, vol. 15, 1993, pages 426 - 429
BARNETT T. ET AL., GENOMICS, vol. 3, 1988, pages 59 - 66
TAWARAGI Y. ET AL., BIOCHEM. BIOPHYS. RES. COMMUN, vol. 150, 1988, pages 89 - 96
BLUMBERG H. ET AL., CELL, vol. 104, 2001, pages 9 - 19
DUMOUTIER L. ET AL., J. IMMUNOL., vol. 167, 2001, pages 3545 - 3549
SMELLIE, M.BOSE, D. S.THOMPSON, A. S.JENKINS, T. C.HARTLEY, J. A.THURSTON, D. E., BIOCHEMISTRY, vol. 42, 2003, pages 12617 - 12624
SHEIKH F. ET AL., J. IMMUNOL., vol. 172, 2004, pages 2006 - 2010
GARY S.C. ET AL., GENE, vol. 256, 2000, pages 139 - 147
CHAN,J.WATT, V.M., ONCOGENE, vol. 6, no. 6, 1991, pages 1057 - 1061
ONCOGENE, vol. 10, no. 5, 1995, pages 897 - 905
ANNU. REV. NEUROSCI., vol. 21, 1998, pages 309 - 345
INT. REV. CYTOL., vol. 196, 2000, pages 177 - 244
REITER R.E. ET AL., PROC. NATL. ACAD. SCI. U.SA., vol. 95, 1998, pages 1735 - 1740
GU Z. ET AL., ONCOGENE, vol. 19, 2000, pages 1288 - 1296
NAKAYAMA ET AL., BIOCHEM. BIOPHYS. RES. COMMUN, vol. 277, no. 1, 2000, pages 124 - 127
THOMPSON, J.S. ET AL., SCIENCE, vol. 293, no. 5537, 2001, pages 2108 - 2111
WILSON ET AL., J. EXP. MED., vol. 173, 1991, pages 137 - 146
YU ET AL., J. IMMUNOL., vol. 148, no. 2, 1992, pages 1526 - 1531
MUELLER ET AL., EUR. J. BIOCHEM., vol. 22, 1992, pages 1621 - 1625
HASHIMOTO ET AL., IMMUNOGENETICS, vol. 40, no. 4, 1994, pages 287 - 295
PREUD'HOMME ET AL., CLIN. EXP. IMMUNOL., vol. 90, no. 1, 1992, pages 141 - 146
SAKAGUCHI ET AL., EMBO J., vol. 7, no. 11, 1988, pages 3457 - 3464
BARELLA ET AL., BIOCHEM. J., vol. 309, 1995, pages 773 - 779
TONNELLE ET AL., EMBO J., vol. 4, no. 11, 1985, pages 2839 - 2847
KASAHARA ET AL., IMMUNOGENETICS, vol. 30, no. 1, 1989, pages 411 - 413
MARKS ET AL., J. MOL. BIOL., vol. 222, 1992, pages 581 - 597
STRAUSBERG ET AL., PROC. NATL. ACAD. SCI USA, vol. 99, 2002, pages 16899 - 16903
SERVENIUS ET AL., J. BIOL. CHEM., vol. 262, 1987, pages 8759 - 8766
BECK ET AL., J. MOL. BIOL., vol. 255, 1996, pages 1 - 13
NARUSE ET AL., ISSUE ANTIGENS, vol. 59, 2002, pages 512 - 519
LARHAMMAR ET AL., J. BIOL. CHEM., vol. 260, no. 26, 1985, pages 14111 - 14119
TOUCHMAN ET AL., GENOME RES., vol. 10, 2000, pages 165 - 173
VON HOEGEN ET AL., J. IMMUNOL., vol. 144, no. 12, 1990, pages 4870 - 4877
MIURA ET AL., GENOMICS, vol. 38, no. 3, 1996, pages 299 - 304
MIURA ET AL., BLOOD, vol. 92, 1998, pages 2815 - 2822
DAVIS ET AL., PROC. NATL. ACAD. SCI USA, vol. 98, no. 17, 2001, pages 9772 - 9777
"NCBI", Database accession no. AAG49451
HORIE ET AL., GENOMICS, vol. 67, 2000, pages 146 - 152
LIANG ET AL., CANCER RES., vol. 60, 2000, pages 4907 - 12
GLYNNE- JONES ET AL., INT J CANCER., vol. 94, no. 2, 15 October 2001 (2001-10-15), pages 178 - 84
MCGLINCHEY, R.P. ET AL., PROC. NATL. ACAD. SCI. U.SA., vol. 106, no. 33, 2009, pages 13731 - 13736
KUMMER, M.P. ET AL., J. BIOL. CHEM., vol. 284, no. 4, 2009, pages 2296 - 2306
HARMS, P.W., GENES DEV., vol. 17, no. 21, 2003, pages 2624 - 2629
GERY, S. ET AL., ONCOGENE, vol. 22, no. 18, 2003, pages 2723 - 2727
KIM, M.H. ET AL., MOL. CELL. BIOL., vol. 29, no. 8, 2009, pages 2264 - 2277
TREANOR, J.J. ET AL., NATURE, vol. 382, no. 6586, 1996, pages 80 - 83
DE NOOIJ-VAN DALEN, A G. ET AL., INT. J. CANCER, vol. 103, no. 6, 2003, pages 768 - 774
ZAMMIT, D.J. ET AL., MOL. CELL. BIOL., vol. 22, no. 3, 2002, pages 946 - 952
MALLYA, M. ET AL., GENOMICS, vol. 80, no. 1, 2002, pages 113 - 123
RIBAS, G. ET AL., J. IMMUNOL., vol. 163, no. 1, 1999, pages 278 - 287
SALANTI, G. ET AL., AM. J. EPIDEMIOL., vol. 170, no. 5, 2009, pages 537 - 545
YAMAMOTO, Y. ET AL., HEPATOLOGY, vol. 37, no. 3, 2003, pages 528 - 533
TSUKAMOTO, H. ET AL., CANCER SCI., vol. 100, no. 10, 2009, pages 1895 - 1901
NARITA, N. ET AL., ONCOGENE, vol. 8, no. 34, 2009, pages 3058 - 3068
ISHIKAWA, N. ET AL., CANCER RES., vol. 67, no. 24, 2007, pages 11601 - 11611
MONTPETIT, A.SINNETT, D., HUM. GENET., vol. 105, no. 1-2, 1999, pages 162 - 164
O'DOWD, B.F. ET AL., FEBS LETT., vol. 394, no. 3, 1996, pages 325 - 329
NAVENOT, J.M. ET AL., MOL. PHARMACOL., vol. 75, no. 6, 2009, pages 1300 - 1306
HATA, K. ET AL., ANTICANCER RES., vol. 29, no. 2, 2009, pages 617 - 623
GERHARD, D.S. ET AL., GENOME RES., vol. 14, no. 10B, 2004, pages 2121 - 2127
BISHOP, D.T. ET AL., NAT. GENET., vol. 41, no. 8, 2009, pages 920 - 925
NAN, H. ET AL., INT. J. CANCER, vol. 125, no. 4, 2009, pages 909 - 917
SCHERER, S.E. ET AL., NATURE, vol. 440, no. 7082, 2006, pages 346 - 351
TAKEDA, S. ET AL., FEBS LETT., vol. 520, no. 1-3, 2002, pages 97 - 101
SABBATH ET AL., J. CLIN. INVEST., vol. 75, 1985, pages 756 - 56
ANDREWS ET AL., BLOOD, vol. 68, 1986, pages 1030 - 5
DRICKAMER K, CURR. OPIN. STRUCT. BIOL., vol. 9, no. 5, 1999, pages 585 - 90
VAN RHENEN A ET AL., BLOOD, vol. 110, no. 7, 2007, pages 2659 - 66
CHEN CH ET AL., BLOOD, vol. 107, no. 4, 2006, pages 1459 - 67
MARSHALL AS ET AL., EUR. J. IMMUNOL., vol. 36, no. 8, 2006, pages 2159 - 69
BAKKER AB ET AL., CANCER RES., vol. 64, no. 22, 2005, pages 8443 - 50
MARSHALL AS ET AL., J. BIOL. CHEM., vol. 279, no. 15, 2004, pages 14792 - 802
CHEN ET AL., J. MOL. BIOL., vol. 293, 1999, pages 865 - 881
PRESTA ET AL., CANCER RES., vol. 57, 1997, pages 4593 - 4599
HUDSON ET AL., NAT. MED., vol. 9, 2003, pages 129 - 134
WINTER ET AL., ANN. REV. IMMUNOL, vol. 113, 1994, pages 433 - 455
HOLLINGER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 6444 - 6448
ALMAGROFRANSSON, FRONT. BIOSCI., vol. 13, 2008, pages 1619 - 1633
RIECHMANN ET AL., NATURE, vol. 322, 1988, pages 738 - 329
QUEEN ET AL., PROC. NAT'L ACAD. SCI. USA, vol. 86, 1989, pages 10029 - 10033
DALL'ACQUA ET AL., METHODS, vol. 36, 2005, pages 61 - 68
PADLAN, MOL. IMMUNOL., vol. 28, 1991, pages 489 - 498
KLIMKA ET AL., BR. J. CANCER, vol. 83, 2000, pages 252 - 26o
CARTER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 89, 1992, pages 4285
PRESTA ET AL., J. IMMUNOL, vol. 151, 1993, pages 2623
BACA ET AL., J. BIOL. CHEM., vol. 272, 1997, pages 10678 - 10684
ROSOK ET AL., J. BIOL. CHEM., vol. 271, 1996, pages 22611 - 22618
VAN DIJKVAN DE WINKEL, CURR. OPIN. PHARMACOL., vol. 5, 2001, pages 368 - 74
LONBERG, CURR. OPIN. IMMUNOL., vol. 20, 2008, pages 450 - 459
LONBERG, NAT. BIOTECH., vol. 23, 2005, pages 1117 - 1125
KOZBOR, J. IMMUNOL, vol. 133, 1984, pages 3001
BRUGGEMANN, M. ET AL., J. EXP. MED., vol. 166, 1987, pages 1351 - 1361
BOERNER ET AL., J. IMMUNOL., vol. 147, 1991, pages 60
LI ET AL., PROC. NATL. ACAD. SCI. USA, vol. 103, 2006, pages 3557 - 3562
NI, XIANDAI MIANYIXUE, vol. 26, no. 4, 2006, pages 265 - 268
VOLLMERSBRANDLEIN, HISTOLOGY AND HISTOPATHOLOGY, vol. 20, no. 3, 2005, pages 927 - 937
VOLLMERSBRANDLEIN, METHODS AND FINDINGS IN EXPERIMENTAL AND CLINICAL PHARMACOLOGY, vol. 27, no. 3, 2005, pages 185 - 91
CRAGG, M.S. ET AL., BLOOD, vol. 101, 2003, pages 1045 - 1052
MCCAFFERTY ET AL., NATURE, vol. 305, 1983, pages 537 - 554
OKAZAKI ET AL., J. MOL. BIOL., vol. 336, no. 5, 2004, pages 1239 - 1249
FELLOUSE, PROC. NATL. ACAD. SCI. USA, vol. 101, no. 34, 2004, pages 12467 - 12472
LEE ET AL., J. IMMUNOL. METHODS, vol. 284, no. 1-2, 2004, pages 119 - 132
GRIFFITHS ET AL., EMBO J, vol. 12, 1993, pages 725 - 734
HOOGENBOOMWINTER, J. MOL. BIOL, vol. 227, 1992, pages 381 - 388
TRAUNECKER ET AL., EMBO J., vol. 10, 1991, pages 3655
ZHU ET AL., PROTEIN SCIENCE, vol. 6, 1997, pages 81 - 788
KOSTELNY ET AL., J. IMMUNOL, vol. 148, no. 5, 1992, pages 1547 - 1553
HOLLINGER ET AL., PROC. NATL ACAD. SCI. USA, vol. 90, 1993, pages 6444 - 6448
GRUBER ET AL., J. IMMUNOL, vol. 152, 1994, pages 5368
CHOWDHURY, METHODS MOL. BIOL., vol. 207, 2008, pages 179 - 196
WRIGHT ET AL., TIBTECH, vol. 15, 1997, pages 26 - 32
YAMANE-OHNUKI ET AL., BIOTECH. BIOENG., vol. 87, 2004, pages 614
RIPKA ET AL., ARCH. BIOCHEM. BIOPHYS., vol. 249, 1986, pages 533 - 545
KANDA, Y. ET AL., BIOTECHNOL. BIOENG., vol. 94, no. 4, 2006, pages 68o - 688
RAVETCHKINET, ANNU. REV. IMMUNOL., vol. 9, 1991, pages 457 - 492
HELLSTROM, I. ET AL., PROC. NAT'LACAD. SCI. USA, vol. 83, 1986, pages 7059 - 7o63
HELLSTROM, I ET AL., PROC. NAT'LACAD. SCI. USA, vol. 82, 1985, pages 1499 - 1502
CLYNES ET AL., PROC. NAT'L ACAD. SCI. USA, vol. 95, 1998, pages 652 - 656
GAZZANO-SANTORO ET AL., J. IMMUNOL. METHODS, vol. 202, 1996, pages 163
CRAGG, M.S.M.J. GLENNIE, BLOOD, vol. 103, 2004, pages 2738 - 2743
PETKOVA, S.B. ET AL., INT 7. IMMUNOL., vol. 18, no. 12, 2006, pages 1759 - 1769
DALL'ACQUA ET AL., JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 281, no. 33, 2006, pages 23514 - 23524
SONDERMANN ET AL., NATURE, vol. 406, 20 July 2000 (2000-07-20), pages 267 - 273
IDUSOGIE ET AL., J. IMMUNOL., vol. 164, 2000, pages 4178 - 4184
GUYER ET AL., J. IMMUNOL., vol. 117, 1976, pages 587
KIM ET AL., J. IMMUNOL., vol. 24, 1994, pages 249
KAM ET AL., PROC. NATL. ACAD. SCI. USA, vol. 102, 2005, pages 11600 - 11605
KUMAR, RLOWN, J. W., ONCOLOGY RESEARCH, vol. 248, 2003, pages 221 - 233
GERNGROSS, NAT. BIOTECH., vol. 22, 2004, pages 1409 - 1414
LI ET AL., NAT. BIOTECH., vol. 24, 2006, pages 210 - 215
GRAHAM ET AL., J. GEN VIROL., vol. 36, 1977, pages 59
MATHER, BIOL. REPROD., vol. 23, 1980, pages 243 - 251
MATHER ET AL., ANNALS N.Y. ACAD. SCI., vol. 383, 1982, pages 44 - 68
URLAUB ET AL., PROC. NATL. ACAD. SCI. USA, vol. 77, 1980, pages 4216
ANTONOW, D.JENKINS, T. C.HOWARD, P. W.THURSTON, D. E., BIOORGANIC & MEDICINAL CHEMISTRY, vol. 15, 2007, pages 3041 - 3053
KAMAL, A.REDDY, K. L.DEVAIAH, V.SHANKARAIAH, N.REDDY, D. R., MINI REV MED CHEM, vol. 6, 2006, pages 53 - 69
S. M. BERGE ET AL., J. PHARM. SCI., vol. 66, 1977, pages 1 - 19
K. R. MORRIS: "Polymorphism in Pharmaceutical Solids", 1995
ANTONOW, D.THURSTON, D. E., CHEM REV, vol. 111, 2011, pages 2815 - 2864
CIPOLLA, L.ARAUJO, A. C.AIROLDI, C.BINI, D., ANTICANCER AGENTS MED CHEM, vol. 9, 2009, pages 1 - 31
GERRATANA, B., MED RES REV, vol. 32, 2012, pages 254 - 293
HARTLEY, J. A., EXPERT OPIN INVESTIG DRUGS, vol. 20, 2011, pages 733 - 744
HURLEY, L. H.RECK, T.THURSTON, D. E.LANGLEY, D. R.HOLDEN, K. G.HERTZBERG, R. P.HOOVER, J. R.GALLAGHER, G., JR.FAUCETTE, L. F.MONG,, CHEM RES TOXICOL, vol. 1, 1988, pages 258 - 268
WELLS, G.MARTIN, C. R.HOWARD, P. W.SANDS, Z. A.LAUGHTON, C. A.TIBERGHIEN, A.WOO, C. K.MASTERSON, L. A.STEPHENSON, M. J.HARTLEY, J., J MED CHEM, vol. 49, 2006, pages 5442 - 5461
BRUCOLI, F.HAWKINS, R. M.JAMES, C. H.JACKSON, P. J.WELLS, G.JENKINS, T. C.ELLIS, T.KOTECHA, M.HOCHHAUSER, D.HARTLEY, J. A., J MED CHEM, vol. 56, 2013, pages 6339 - 6351
KOTECHA, M.KLUZA, J.WELLS, G.O'HARE, C. C.FORNI, C.MANTOVANI, R.HOWARD, P. W.MORRIS, P.THURSTON, D. E.HARTLEY, J. A., MOL CANCER THER, vol. 7, 2008, pages 1319 - 1328
PUWADA, M. S.HARTLEY, J. A.JENKINS, T. C.THURSTON, D. E., NUCLEIC ACIDS RES, vol. 21, 1993, pages 3671 - 3675
CLINGEN, P. H.DE SILVA, I. U.MCHUGH, P. J.GHADESSY, F. J.TILBY, M. J.THURSTON, D. E.HARTLEY, J. A., NUCLEIC ACIDS RES, vol. 33, 2005, pages 3283 - 3291
PUWADA, M. S.FORROW, S. A.HARTLEY, J. A.STEPHENSON, P.GIBSON, I.JENKINS, T. C.THURSTON, D. E., BIOCHEMISTRY, vol. 6, 1997, pages 8 - 2484
BARKLEY, M. D.CHEATHAM, S.THURSTON, D. E.HURLEY, L. H., BIOCHEMISTRY, vol. 25, 1986, pages 3021 - 3031
SEIFERT, J.PEZESHKI, S.KAMAL, A.WEISZ, K., ORGANIC & BIOMOLECULAR CHEMISTRY, vol. 10, 2012, pages 6850 - 6860
KOPKA, M. L.GOODSELL, D. S.BAIKALOV, I.GRZESKOWIAK, K.CASCIO, D.DICKERSON, R. E., BIOCHEMISTRY, vol. 33, 1994, pages 13593 - 13610
KIZU, R.DRAVES, P. H.HURLEY, L. H., BIOCHEMISTRY, vol. 32, 1993, pages 8712 - 8722
LEIMGRUBER, W.STEFANOVIC, V.SCHENKER, F.KARR, A.BERGER, J., J AM CHEM SOC, vol. 87, 1965, pages 5791 - 5793
ARIMA, K.KOSAKA, M.TAMURA, G.IMANAKA, H.SAKAI, H., JANTIBIOT (TOKYO, vol. 25, 1972, pages 437 - 444
SATO, S.IWATA, F.YAMADA, S.KAWAHARA, H.KATAYAMA, M., BIOORG MED CHEM LETT, vol. 21, 2011, pages 7099 - 7101
THURSTON D.E.BOSE D.S., CHEM REV, vol. 94, 1994, pages 433 - 465
DAMAYANTHI, Y. ET AL., JOURNAL OF ORGANIC CHEMISTRY, vol. 64, 1999, pages 290 - 292
KUMAR ET AL., HETEROCYCLIC COMMUNICATIONS, vol. 8, 2002, pages 19 - 26
BARALDI, P. G. ET AL., JOURNAL OF MEDICINAL CHEMISTRY, vol. 42, 1999, pages 5131 - 5141
WELLS, G. ET AL., PROC. AM. ASSOC. CANC. RES., vol. 44, 2003, pages 452
REDDY, B. S.SHARMA, S. K.LOWN, J. W., CURRENT MEDICINAL CHEMISTRY, vol. 8, 2001, pages 475 - 508
BLENCOWE, C. A.RUSSELL, A. T.GRECO, F.HAYES, W.THORNTHWAITE, D.W., POLYMER CHEMISTRY, vol. 2, 2011, pages 773 - 790
HAMPSHIRE, A. J.RUSLING, D. A.BROUGHTON-HEAD, V. J.FOX, K. R., METHODS, vol. 42, 2007, pages 128 - 140
DREW, H. R.TRAVERS, A. A., CELL, vol. 37, 1984, pages 491 - 502
WIEGAND, I.HILPERT, K.HANCOCK, R. E., NATURE PROTOCOLS, vol. 3, 2008, pages 163 - 175
Attorney, Agent or Firm:
SCRIPT IP LIMITED et al. (GB)
Download PDF:
Claims:
CLAIMS l. A compound of formula (I):

or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof;

wherein:

q is o or l;

the dotted lines from Z, to Z4 represent single or double bonds;

Z is selected from O, C-R and CH-Rq Z2 is selected from O, C-R2 and CH-R2; Z3 is selected from O, C-R3 and CH-R3; Z4 is selected from O, C-I^ and CH-R^

Ri, R2, R3 and R4 are:

(a) independently selected from H, OH, Ci-i2 alkyl, 0Ci-i2 alkyl, =C(RI4)(RI5), RA and halogen; or

(b) one of Ri and R2; or R2 and R3; or R3 and together with the carbon atoms to which they are attached form a 6-membered aryl ring, or a 5- or 6-membered heteroaryl ring, wherein the non-fused carbons of the aryl or heteroaryl ring are substituted with groups RDi, RD2, RD3 and RD4; and the remaining Ri, R2, R3 groups that do not form a ring are independently selected from H, OH, Ci-ia alkyl, 0Ci-i2 alkyl, RA and halogen; or

(c) one the remaining are independently selected from H, OH, Ci-i2 alkyl, 0Ci-i2 alkyl, RA and halogen; each RI4 and Ri5 are independently selected from H, Ci-i2 alkyl and (CH2)j-Rx;

Rw is selected from Rx, =0, CN, NCO, (CH2)j-0Rx, 0-(CH2)k-0Rx, (CH2)j-C02Rx,

each Rx is independently selected from H, C - 2 alkyl, C5-20 aryl, Ce-26 aralkyl groups, C5-i0 heteroaryl, Ce-ib heteroarylalkyl, C3-20 heterocyclyl; wherein the alkyl, aralkyl, heteroaryl, heteroarylalkyl and heterocyclyl groups are optionally substituted;

RD , RD2, RD3 and RD4 are independently selected from H, OH, C - 2 alkyl, 0C - 2 alkyl, RA and halogen;

Z5 and Z6 together are selected from CR5R6-NR7, CR5R5’-CR6R6’, CR5R6-S, CR5R6-O, CR7=CR5 and NR7-C(=0);

Z7 is C=0 or C=S;

R5, R5’, Re and Re’are independently selected from H, C - 2 alkyl and RA;

R7 is selected from H and C - 2 alkyl;

Re is selected from H, C - 2 alkyl and CH2Ph;

X is O, S, NRl6, CRI6RI7, CRI6RI70, C(=0), C(=0)NRi6, NRI6C(=0), O-C(O), C(0)-0 or is absent;

L is selected from an amino acid, a peptide chain having from 2 to 12 amino acids, a paraformaldehyde chain -(OCH2)I-24-, a polyethylene glycol chain

-(0CH2CH2)m- and -(CH2)m-Li-(CH2)n- wherein

m is o, l, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12;

n is o, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12;

is selected from -(CH2)1-5-, -C(0)-NH-, -NH-, -S(0)0-2-, -CH[(CH2)0-5 RA]-, -Ah- C(0)-NH-(Ar2)0-i-Ar3-, -Ar3-(Ar2)0-i-NH-C(0)-Ar - and -Ar4-;

Ar is an optionally substituted 5-membered heteroarylene;

Ar2 is an optionally substituted 6-membered arylene or heteroarylene;

Ar3 is an optionally substituted 5- to 9-membered heteroarylene ring;

Ar4 is selected from an optionally substituted 3- to 8-membered cycloalkylene, an optionally substituted 3- to 8-membered heterocycloalkene, an optionally substituted 6-membered arylene and an optionally substituted 5- to 9-membered heteroarylene; wherein the optionally substituted Ar , Ar2, Ar3 and Ar4 are optionally substituted with 1, 2 or 3 optional substituents independently selected from OH, C - 2 alkyl, 0C - 2 alkyl and RA;

X2 is O, S, NRl6, CRI6RI7, CRI6RI70, C(=0), C(=0)NRi6, NRI6C(=0), O-C(O), C(0)-0 or is absent;

each R16 and Ri7 are independently selected from H and C - 2 alkyl; r is 1, 2 or 3;

one of each Y and Y2 is independently selected from N-R s, S and O; and the other of each Y and Y2 is CH; each Y3 is independently selected from C-RI9, N and S;

each R s is independently selected from H and C - 2 alkyl;

each R 9 is independently selected from H, OH, C - 2 alkyl and RA; Y4 is N or C-R20;

Y5 is N or C-R’20; and wherein at least one of Y4 and Y5 is C-R20 or C-R’20;

R9 and R o are independently selected from H, C - 2 alkyl and RA;

R20 and R’20 are independently selected from H, C - 2 alkyl and RA;

p is o or l; and

when p is l, then H represents a single bond or then H is a C5 heteroaryl group optionally substituted with l or 2 optional substituent groups independently selected from OH, C - 2 alkyl, 0C - 2 alkyl and RA; and

when p is o, then H is a C9 heteroaryl group optionally substituted with l, 2 or 3 optional substituent groups independently selected from OH, C - 2 alkyl, 0C - 2 alkyl and RA; f is o or 1;

T is:

(i) a C - a alkyl optionally substituted with 1, 2 or 3 substituent groups independently selected from OH, 0C - 2 alkyl and RA;

(ii) a Cg-9 heteroaryl optionally substituted with 1, 2 or 3 substituent groups independently selected from OH, Ci-i2 alkyl, 0Ci-i2 alkyl and RA;

(hi)

Rn, RI2 and Ri3 are independently selected from H, OH, C - 2 alkyl, 0C - 2 alkyl and RA; or

(iv) OH, 0C - 2 alkyl or RA;

each RA is independently selected from (CH2)J-C02R2I, 0-(CH2)k-NR2iR22, C(0)-0- (CH2)k-NR2iR22, C(0)

NH-C(0)-R2I, KI-R33,

0-(Ci-6 alkyl) and NH

each K is independently a bond or a linker moiety having 1-200 non-hydrogen atoms selected from C, N, O, S or halogen, and optionally incorporates alkyl, ether, oxo, carboxyl, carboxamide, carboxamidyl, ester, urethanyl, branched, cyclic, unsaturated, heterocyclyl, aryl or heteroaryl moieties; each R33 is independently an azide, alkyne, bisulfone, carbohydrazide, hydrazine, hydroxylamine, iodoacetamide, isothiocyanate, maleimide, phosphine,

pyrridopyridazine, semihydrazide, succinimidyl ester, sulfodichlorophenol ester, sulfonyl halide, sulfosuccinimidyl ester, 4-sulfotetrafluorophenyl ester,

tetrafluorophenyl ester, thiazole, (CH2)j-C02R34, 0-(CH2)k-NR34R35, C(0)-0-(CH2)k- NH-C(O)- alkyl), NH-C(0)-NR34R35, H or a targeting agent wherein each targeting agent is

independently a protein, a portion of a protein, a polypeptide, a nucleic acid, a hormone, an antibody or an antibody fragment;

each j is independently selected from o, 1, 2, 3, 4, 5 or 6;

each k is independently selected from 1, 2, 3, 4, 5 or 6;

each Rai and R22 is independently selected from KI-R33, H and C - 2 alkyl; and

each R34 and R35 is independently selected from H and C - 2 alkyl.

2. A compound of formula (I) according to claim 1, wherein the compound is a compound of formula (IV):

or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof.

3. The compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof of claim 1 or claim 2, wherein (a) R , R2, R3 and are present and are independently selected from H, OH, C - 2 alkyl, 0C - 2 alkyl, =C(RI4)(RI5) and halogen.

4. The compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof of claim 1 or claim 2, wherein (b) one of R and R2; or R2 and R3; or R3 and together with the carbon atoms to which they are attached form a 6-membered aryl ring such that the non-alkylating moiety: is selected from:

5. The compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof of claim 1 or claim 2, wherein (c) one of R , R2, R3 and R4 is Rw; such that the non-alkylating moiety is selected from:

6. The compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof of any one of the preceding claims, wherein the compound contains a total of o or 1 RA groups. 7. The compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof of any one of the preceding claims, wherein L is -(CH2)m-L -(CH2)n- and L is selected from -(CH2)I-5-,

Ye.is C-H or N;

Y7 is N-R26, O or S; and

R23, R24 and R25 are independently selected from H, OH, C - 2 alkyl, 0C - 2 alkyl and RA; and R26 is H or C 1-12 alkyl.

8. The compound of formula (I) of any of the preceding claims, wherein the compound is selected from:

or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof; wherein

Ys is selected from N-R28, S and O;

Y9 is selected from C-R29 and N;

one of Y o and Yu is independently selected from N-R2s, S and O; and the other of Y 0 and Yn is C-H;

Y 2 is selected from C-R29, N and S;

R27 is selected from H, OH, C - 2 alkyl, 0C - 2 alkyl and RA;

R28 is selected from H and C - 2 alkyl; and

each R29 is selected from H, OH, C - 2 alkyl, 0C - 2 alkyl and RA.

9. The compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof of any of claims l to 8, linked, either directly or indirectly, to a targeting agent to provide a targeting conjugate. to. The compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof of any of claims l to 9, further comprising one or more linker groups.

11. A pharmaceutical composition comprising a compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof of any one of claims 1 to 10, and a pharmaceutically acceptable carrier, diluent, or excipient.

12. A compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof of any one of claims 1 to 10, or a pharmaceutical composition according to claim 11, for use as a medicament.

13. A compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof of any one of claims 1 to 10, for use as a drug in an antibody-drug conjugate.

14. A compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof of any one of claims 1 to 10, or a pharmaceutical composition according to claim 11, for use in the treatment of a proliferative disease, a bacterial infection, a malarial infection and inflammation.

15. A compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, or pharmaceutical composition for use in the treatment of a proliferative disease according to claim 14, wherein the proliferative disease is selected from bladder cancer, bone cancer, bowel cancer, brain cancer, breast cancer, cervical cancer, colon cancer, head and neck cancer, leukemia, liver cancer, lung cancer, lymphoma, melanoma, oesophageal cancer, oral cancer, ovarian cancer, pancreatic cancer, prostate cancer, rectal cancer, renal cancer, retinoblastoma, sarcoma, skin cancer, stomach cancer, testicular cancer, thyroid cancer and uterine cancer.

Description:
POLYCYCLIC AMIDES AS CYTOTOXIC AGENTS

FIELD OF THE INVENTION

The invention relates to novel cytotoxic agents comprising multiple fused rings similar to guanine-alkylating moieties, such as pyrrolobenzodiazepines (PBDs), but comprising groups that are non-alkylating. In particular, the invention relates to novel non- alkylating compounds linked via the A-ring to a side chain comprising multiple aromatic groups, and to pharmaceutically acceptable salts thereof, which are useful as medicaments, in particular as anti-proliferative agents.

BACKGROUND

The pyrrolobenzodiazepines (PBDs) are a group of compounds some of which have been shown to be sequence-selective DNA minor-groove binding agents. The PBDs were originally discovered in Streptomyces species [1-5]. They are tricyclic in nature, and are comprised of fused 6-7-5-membered rings that comprise an anthranilate (A ring), a diazepine (B ring) and a pyrrolidine (C ring) [3]. They are characterized by an electrophilic Nio=Cn imine group (as shown below) or the hydrated equivalent, a carbinolamine [NH-CH(OH)], or a carbinolamine alkyl ether ([NH-CH(OR, where R = alkyl)] which can form a covalent bond to a C2-amino group of guanine in DNA to form a DNA adduct [6].

Carbinolamine Imine Carbinolamine alkyl ether

The natural products interact in the minor groove of the DNA helix with excellent fit (i.e., good“isohelicity”) due to a right-handed longitudinal twist induced by a chiral

Cua-position which has the (^-configuration [6]. The DNA adduct has been reported to inhibit a number of biological processes including the binding of transcription factors [7-9] and the function of enzymes such as endonucleases [10, 11] and RNA polymerase (12). PBD monomers (e.g., anthramycin) have been shown by footprinting [6], NMR [13, 14], molecular modeling [15] and X-ray crystallography [16] to span three base pairs and to have a thermodynamic preference for the sequence 5’-Pu-G-Pu- 3’ (where Pu = purine, and G is the reacting guanine) [17] and a kinetic preference for Py-5-Py (where Py = Pyrimidine). PBDs are thought to interact with DNA by first locating at a low-energy binding sequence (i.e., a s’-Pu-G-Pu-3’ triplet) through Van der Waals, hydrogen bonding and electrostatic interactions [7]. Then, once in place, a nucleophilic attack by the exocyclic C2-amino group of the central guanine occurs to form the covalent adduct [7]. Once bound, the PBD remains anchored in the DNA minor groove, avoiding DNA repair by causing negligible distortion of the DNA helix [16]. The ability of PBDs to form an adduct in the minor groove and crosslink DNA enables them to interfere with DNA processing and, hence, their potential for use as antiproliferative agents. Hence, the ability of these compounds to undergo alkylation to form a covalent adduct was considered vital to their effectiveness as antiproliferative agents.

A number of monomeric PBD structures have been isolated from Streptomyces species, including anthramycin [18] the first PBD, tomamycin [19], and more recently usabamycin [20] from a marine sediment Streptomyces species in a marine sediment. This has led to the development of a large range of synthetic analogues which have been reviewed [1, 21]. More recently, a number of monomeric PBD structures that are linked through their C8 position to pyrroles and imidazoles have been reported WO

2007/039752, WO 2013/164593 [22-27].

In addition to pyrrolobenzodiazepines (PBDs), comprising three fused 6-7-5- membered rings), other guanine alkylating moieties, such as C2-substituted PBDs (including C2-endo, Ci/C2-endo, and C2/C3-endo PBDs), pyrridinobenzodiazepines (PDDs), comprising three fused 6-7-6-membered rings), indolinobenzodiazapenes (IBDs, comprising four fused 6-7-5-6 membered rings), and tetrahydroisoquinoline- benzodiazapines (QBDs, comprising four fused 6-7-6-6 membered rings) are known.

As with PBDs the ability of these other classes of compounds to undergo alkylation to form a covalent adduct is the key to their effectiveness as antiproliferative agents.

Non-alkylating PBDs (i.e., dilactams) have been extensively reported in the literature and exhibit minimal stabilisation of the DNA. In a similar manner to alkylating PBDs, dilactams are isohelical with the DNA minor groove due to their chiral Cua-position, and therefore possess weak DNA-binding properties through non-covalent hydrogen bonding and other interactions such as van der Waals. This is reflected in the literature where a series of dilactams [28] provided a thermal stabilization (i.e., AT m ) of up to approximately 3°C with calf thymus DNA, comparing unfavourably with a simple PBD monomer such as anthramycin which provides a AT m of 13.1°C under the same conditions. This suggests dilactams do not stabilise DNA. Furthermore, other libraries of PBD dilactams have shown similar AT m ranges (e.g., up to approximately 2.4°C for C2-aryl substituted dilactams) [29], suggesting a‘ceiling’ of 3°C for dilactam molecules. Studies on PBD dilactam-distamycin conjugates (containing a PBD dilactam molecule linked to three pyrroles via a trimethylene spacer) illustrated a mean GI 50 of >iomM across the NCI cell line panel compared to 0.04mM for the equivalent PBD imine- containing molecule, suggesting the alkylation event is critical for potent cytotoxicity.

The present inventors have surprisingly found that a non-alkylating multiple fused ring moieties, which are not cytotoxic themselves, can form an effective cytotoxic agent when attached to a suitable side chain. Such suitable side chains comprise multiple aromatic groups and are also not cytotoxic themselves. Hence, the fact that the overall compounds produce effective cytotoxic agents is unexpected, in particular, as they do not undergo alkylation. The cytotoxicity occurs as a result of the DNA-binding ability and sequence-selectivity of the compounds. Although potent cytotoxicity is still maintained, the fact that the alkylating ability of the compounds is removed should result in compounds with higher tolerability in mice and humans, and therefore a far wider therapeutic index (TI) than DNA-alkylating agents. These compounds have the advantage of improved tolerability as compared to the known guanine alkylating compounds. SUMMARY

In a first aspect, the present invention provides a compound of formula (I):

or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof;

wherein:

q is o or 1;

the dotted lines from Z to Z 4 represent single or double bonds;

Z is selected from O, C-R and CH-Rq Z 2 is selected from O, C-R 2 and CH-R 2 ; Z 3 is selected from O, C-R 3 and CH-R 3 ; Z 4 is selected from O, C-IU, and CH-IU,;

R , R 2 , R 3 and R4 are:

(a) independently selected from H, OH, C - i2 alkyl, 0C - 2 alkyl, =C(R I4 )(R I5 ), RA and halogen; or

(b) one of R and R 2 ; or R 2 and R 3 ; or R 3 and IU, together with the carbon atoms to which they are attached form a 6-membered aryl ring, or a 5- or 6-membered heteroaryl ring, wherein the non-fused carbons of the aryl or heteroaryl ring are substituted with groups RD , RD 2 , RD 3 and RD 4 ; and the remaining R ls R 2 , R 3 and R4 groups that do not form a ring are independently selected from H, OH, C - 2 alkyl, 0C - i2 alkyl, R A and halogen; or

(c) one of R , R 2 , R 3 and R4 is R w ; and the remaining of R , R 2 , R 3 and R4 are independently selected from H, OH, C - i2 alkyl, 0C - i2 alkyl, R A and halogen; each R I4 and R 15 are independently selected from H, C - i2 alkyl and (CH 2 ) j -R x ;

R w is selected from R x , =0, CN, NCO, (CH 2 ) j -0R x , 0-(CH 2 ) k -0R x , (CH 2 ) j -C0 2 R x ,

each R x is independently selected from H, 0- 12 alkyl, C 5-20 aryl, C 6 -26 aralkyl groups, C 5-i0 heteroaryl, 0 6-i6 heteroarylalkyl, C 3-20 heterocyclyl; wherein the alkyl, aralkyl, heteroaryl, heteroarylalkyl and heterocyclyl groups are optionally substituted;

RD , RD 2 , RD 3 and RD 4 are independently selected from H, OH, C - i2 alkyl, 0C - i2 alkyl, R A and halogen;

Z 5 and Z 6 together are selected from CR 5 R6-NR 7 , CR 5 R 5 ’-CR6R6’, CR5R6-S, CR5R6-O,

(=0); independently selected from H, C - i2 alkyl and R A ;

R 7 is selected from H and 0- 12 alkyl;

Rg is selected from H, C - i2 alkyl and CH 2 Ph;

X is O, S, NR 6, CR l6 R 17 , CR I6 R 17 0, C(=0), C(=0)NR i6 , NR I6 C(=0), O-C(O), C(0)-0 or is absent;

L is selected from an amino acid, a peptide chain having from 2 to 12 amino acids, a paraformaldehyde chain -(0CH 2 ) _ 24 -, a polyethylene glycol chain

-(0CH 2 CH 2 ) m - and -(CH 2 ) m -L -(CH 2 ) n - wherein

m is o, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12;

n is o, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12;

is selected from - C(0)-NH-(Ar 2 ) 0-1 -A At i is an optionally substituted 5-membered heteroarylene;

Ar 2 is an optionally substituted 6-membered arylene or heteroarylene;

Ar 3 is an optionally substituted 5- to 9-membered heteroarylene ring;

Ar 4 is selected from an optionally substituted 3- to 8-membered cycloalkylene, an optionally substituted 3- to 8-membered heterocycloalkene, an optionally substituted 6-membered arylene and an optionally substituted 5- to 9-membered heteroarylene; wherein the optionally substituted Ar , Ar 2 , Ar 3 and Ar 4 are optionally substituted with 1, 2 or 3 optional substituents independently selected from OH, C - i2 alkyl, 0C - i2 alkyl and R A ;

X 2 is O, S, NR 16 , CR l6 R 17 , CR I6 R 17 0, C(=0), C(=0)NR i6 , NR I6 C(=0), O-C(O), C(0)-0 or is absent;

each R 16 and R 17 are independently selected from H and C - i2 alkyl;

r is 1, 2 or 3;

one of each Y and Y 2 is independently selected from N-R s, S and O; and the other of each Y and Y 2 is CH;

each Y 3 is independently selected from C-R I9 , N and S;

each R g is independently selected from H and C,- 12 alkyl;

each R 19 is independently selected from H, OH, 0- 12 alkyl and R A ;

Y 4 is N or C-R 20 ;

Y 5 is N or C-R’ 20 ; and wherein at least one of Y 4 and Y 5 is C-R 20 or C-R’ 20 ;

R g and R 10 are independently selected from H, 0- 12 alkyl and R A ;

R 20 and R’ 20 are independently selected from H, 0- 12 alkyl and R A ;

p is o or 1; and

when p is 1, then H represents a single bond or H is a C 5 heteroaryl group optionally substituted with 1 or 2 optional substituent groups independently selected from OH, C - 2 alkyl, 0C - 2 alkyl and R A ; and

when p is o, then H is a C 9 heteroaryl group optionally substituted with 1, 2 or 3 optional substituent groups independently selected from OH, C - 2 alkyl, 0C - 2 alkyl and R A ;

f is o or 1;

T is:

(i) a C - 2 alkyl optionally substituted with 1, 2 or 3 substituent groups independently selected from OH, 0C - 2 alkyl and R A ;

(ii) a C 5-9 heteroaryl optionally substituted with 1, 2 or 3 substituent groups independently selected from OH, C - 2 alkyl, 0C - 2 alkyl and R A ;

(iii)

Rn, R I2 and R, 3 are independently selected from H, OH, Ci-i 2 alkyl, 0Ci-i 2 alkyl and R A ; or

(iv) OH, OC - 12 alkyl or R A ;

each R A is independently selected from (CH 2 ) j -C0 2 R 2i , 0-(CH 2 ) k -NR 2i R 22 , C(0)-0-

(CH 2 ) k -NR 21 R 22 , C(0)

NH-C(0)-R 21 , Ki-Rs j ,

CKC ! -e alkyl) and NH

each K is independently a bond or a linker moiety having 1-200 non-hydrogen atoms selected from C, N, O, S or halogen, and optionally incorporates alkyl, ether, oxo, carboxyl, carboxamide, carboxamidyl, ester, urethanyl, branched, cyclic, unsaturated, heterocyclyl, aryl or heteroaryl moieties;

each R 33 is independently an azide, alkyne, bisulfone, carbohydrazide, hydrazine, hydroxylamine, iodoacetamide, isothiocyanate, maleimide, phosphine,

pyrridopyridazine, semihydrazide, succinimidyl ester, sulfodichlorophenol ester, sulfonyl halide, sulfosuccinimidyl ester, 4-sulfotetrafluorophenyl ester,

tetrafluorophenyl ester, thiazole, (CH 2 ) j -C0 2 R 34 , 0-(CH 2 ) k -NR 34 R 35 , C(0)-0-(CH 2 ) k -

NR 34 R 35 , C(0)-NR 34 R 35 , (CH 2 )J-NR 34 R 35 , NR 35 NH 2 , C(0)-NH-(CH 2 )J-NR 34 R 35 , NH-C(O)

R 35 , C(0)-NH-(CH 2 ) k -C(=NH)NR 34 R 35 , (CH 2 ) j -S0 2 -NR 34 R 35 , C(=NH)-0-(C 1-6 alkyl), NH-C(0)-NR 34 R 35 , H or a targeting agent wherein each targeting agent is

independently a protein, a portion of a protein, a polypeptide, a nucleic acid, a hormone, an antibody or an antibody fragment;

each j is independently selected from o, l, 2, 3, 4, 5 or 6;

each k is independently selected from 1, 2, 3, 4, 5 or 6;

each R 2 , and R 22 is independently selected from K -R 33 , H and Ci-i 2 alkyl; and each R 34 and R 35 is independently selected from H and Ci-i 2 alkyl.

In a further aspect, the present invention provides a compound of formula (I) with is a compound of formula (II):

or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof; wherein:

q is o or l;

the dotted lines from Z to Z 4 represent single or double bonds;

Z is selected from O, C-R and CH-Rq Z 2 is selected from O, C-R 2 and CH-R 2 ; Z 3 is selected from O, C-R 3 and CH-R 3 ; Z 4 is selected from O, C-R z , and CH-R z ,;

R , R 2 , R 3 and I¾ are:

(a) independently selected from H, OH, C - i2 alkyl, 0C - i2 alkyl, =C(R I4 )(R I5 ), R A and halogen; or

(b) one of R and R 2 ; or R 2 and R 3 ; or R 3 and I¾ together with the carbon atoms to which they are attached form a 6-membered aryl ring, or a 5- or 6-membered heteroaryl ring, wherein the non-fused carbons of the aryl or heteroaryl ring are substituted with groups RD , RD 2 , RD 3 and RD 4 ; and the remaining R ls R 2 , R 3 and I¾ groups that do not form a ring are independently selected from H, OH, C - 2 alkyl, 0C - i2 alkyl, R A and halogen; or

(c) one of R , R 2 , R 3 and I¾ is R w ; and the remaining of R , R 2 , R 3 and I¾ are independently selected from H, OH, C - i2 alkyl, 0C - i2 alkyl, R A and halogen; each R I4 and R 15 are independently selected from H, C - i2 alkyl and (CH 2 ) j -R x ;

R w is selected from R x , =0, CN, NCO, (CH 2 ) j -0R x , 0-(CH 2 ) k -0R x , (CH 2 ) j -C0 2 R x ,

each R x is independently selected from H, 0- 12 alkyl, C 5-20 aryl, C 6-26 aralkyl groups, C 5-i0 heteroaryl, 0 6-i6 heteroarylalkyl, C 3-20 heterocyclyl; wherein the alkyl, aralkyl, heteroaryl, heteroarylalkyl and heterocyclyl groups are optionally substituted;

RD , RD 2 , RD 3 and RD 4 are independently selected from H, OH, C - i2 alkyl, 0C - i2 alkyl, R A and halogen;

Z 5 and Z 6 are selected from CR 5 R6-NR 7 , CR 5 R 5 ’-CR 6 R 6 ’, CR5R6-S, CR5R6-O, CR 7 =CR 5 and NR 7 -C(=0);

Z 7 is C=0 or C=S;

R 5 , R 5 ’, Re, and R 6 ’are independently selected from H, C - i2 alkyl and R A ;

R 7 is selected from H and 0- 12 alkyl; Re is selected from H, C - 2 alkyl and CH 2 Ph;

X is O, S, Nf , CR I6 R 17 , CR I6 R 17 0, C(=0), C(=0)NR i6 , NR I6 C(=0), O-C(O), C(0)-0 or is absent;

L is selected from an amino acid, a peptide chain having from 2 to 12 amino acids, a paraformaldehyde chain -(0CH 2 ) _ 24 -, a polyethylene glycol chain

-(0CH 2 CH 2 )m- and -(CH 2 ) m -L -(CH 2 ) n - wherein

m is o, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12;

n is o, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12;

is selected from -(CH 2 ) I-5 -, -C(0)-NH-, -NH-, -S(0) 0-2 -, -CH[(CH 2 ) 0-5 R A ]-, -AT I - C(0)-NH-(Ar 2 ) 0-1 -Ar 3 -, -Ar 3 -(Ar 2 ) 0-i -NH-C(0)-Ar 1 - and -Ar 4 -;

AT I is an optionally substituted 5-membered heteroarylene;

Ar 2 is an optionally substituted 6-membered arylene or heteroarylene;

Ar 3 is an optionally substituted 5- to 9-membered heteroarylene ring;

Ar 4 is selected from an optionally substituted 3- to 8-membered cycloalkylene, an optionally substituted 3- to 8-membered heterocycloalkene, an optionally substituted 6-membered arylene and an optionally substituted 5- to 9-membered heteroarylene; wherein the optionally substituted Ar , Ar 2 , Ar 3 and Ar 4 are optionally substituted with 1, 2 or 3 optional substituents independently selected from OH, C - i2 alkyl, 0C - i2 alkyl and R A ;

X 2 is O, S, NR 16 , CR l6 R 17 , CR I6 R 17 0, C(=0), C(=0)NR i6 , NR I6 C(=0), O-C(O), C(0)-0 or is absent;

each R 16 and R 17 are independently selected from H and C - i2 alkyl;

r is 1, 2 or 3;

one of each Y and Y 2 is independently selected from N-R s, S and O; and the other of each Y and Y 2 is CH;

each Y 3 is independently selected from C-R 1 9, N and S;

each R g is independently selected from H and C,- 12 alkyl;

each R 19 is independently selected from H, OH, 0- 12 alkyl and R A ;

Y 4 is N or C-R 20 ;

Y 5 is N or C-R’ 20 ; and wherein at least one of Y 4 and Y 5 is C-R 20 or C-R’ 20 ;

R g and R 10 are independently selected from H, 0- 12 alkyl and R A ;

R 20 and R’ 20 are independently selected from H, 0- 12 alkyl and R A ;

p is o or 1; and

when p is 1, then H is a C 5 heteroaryl group optionally substituted with 1 or 2 optional substituent groups independently selected from OH, C - i2 alkyl, 0C - i2 alkyl and R A ; and when p is o, then H is a C g heteroaryl group optionally substituted with l, 2 or 3 optional substituent groups independently selected from OH, C - 2 alkyl, OC 1-12 alkyl and R A ;

T is:

(i) a C - 2 alkyl optionally substituted with 1, 2 or 3 substituent groups independently selected from OH, OC 1-12 alkyl and R A ;

(ii) a C 5-9 heteroaryl optionally substituted with 1, 2 or 3 substituent groups independently selected from OH, C - 2 alkyl, OC 1-12 alkyl and R A ; or

(hi)

R 11 , R 12 and R 13 are independently selected from H, OH, C - 2 alkyl, OC 1-12 alkyl and R A ;

each R A is independently selected from (CH 2 )j-C0 2 R 2i , 0-(CH 2 ) k -NR 2i R 22 , C(0)-0- (CH 2 ) k -NR 2i R 22 , C(0)-NR 2I R 22 , (CH 2 )j-NR 2i R 22 , NR 2I NH 2 , C(0)-NH-(CH 2 )j-NR 2i R 22 , C(0)-NH-(CH 2 ) k -C(=NH)NR 2i R 22 , (CH 2 )j-S0 2 -NR 2i R , C(=NH)-0-(C - 6 alkyl) and NH-

C(0)-NR 2I R 22 ; and

each j is independently selected from o, 1, 2, 3, 4, 5 or 6;

each k is independently selected from 1, 2, 3, 4, 5 or 6;

each R 2I and R 22 is independently selected from H and C - 2 alkyl.

In a further aspect, there is provided a compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof as described herein, linked, either directly or indirectly, to a targeting agent to provide a targeting conjugate.

In a further aspect, there is provided a compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof as described herein, linked to a linking group. In a further aspect, the present invention provides a pharmaceutical composition comprising a compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof as described herein, and a

pharmaceutically acceptable carrier, diluent, or excipient. The pharmaceutical composition of the present invention may further comprise one or more (e.g. two, three or four) further active agents. In a further aspect, there is provided a compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, or a pharmaceutical composition as described herein, for use as a medicament.

In a further aspect, there is provided a compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, or a pharmaceutical composition as described herein, for use in a method of therapy.

In a further aspect, there is provided a compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof as described herein, for use as a drug in an antibody-drug conjugate.

In certain aspects, the compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, may be used as a payload on a tumour-targeting agent (e.g., antibody, antibody fragment, hormone, etc.).

In a further aspect, the compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, may be linked, either directly or indirectly, to a targeting agent (e.g., antibody, antibody fragment, hormone, etc.) to provide a targeted conjugate. In a further aspect, the compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, may contain a linker group, wherein the targeting agent is attached to the compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, through the linker group. The target conjugates of the present disclosure may contain one or multiple compounds of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof. A variety of target conjugates are known in the art and may be used with a compound of formula (I) and salts or solvates thereof. For example, in a particular aspect the target conjugate is an antibody-drug conjugate, wherein one or more compounds of formula (I) are linked, directly or indirectly, to the antibody. Therefore, the compound of formula (I) and salts or solvates thereof, may be used as a payload on a targeted conjugate.

In a further aspect, there is provided a compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, or a pharmaceutical composition as described herein, for use in the treatment of a proliferative disease, a bacterial infection, a malarial infection and inflammation.

In a further aspect, the present invention provides a method of treatment of a patient suffering from a proliferative disease, comprising administering to said patient a therapeutically effective amount of a compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, or a pharmaceutical composition comprising a compound of formula (I).

In a further aspect, the compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, may be administered alone or in combination with other treatments, either simultaneously or sequentially depending upon the condition to be treated.

Further particular and preferred aspects are set out in the accompanying independent and dependent claims. Features of the dependent claims maybe combined with features of the independent claims as appropriate, and in combinations other than those explicitly set out in the claims.

DEFINITIONS

The following abbreviations are used throughout the specification: Ac acetyl; Alloc allyloxycarbonyl; BAIB/PIDA bis(acetoxy)iodobenzene/ (diacetoxyiodo)benzene/ phenyliodine(III) diacetate; Boc tert-butoxycarbonyl; DHP dihydropyran; DMAP 4- dimethylaminopyridine; DMF dimethylformamide; EDCI i-Ethyl-3-(3-dimethylamino- propyl)carbodiimide; Et ethyl; HATU (i-[Bis(dimethylamino)methylene]-iH-i,2,3- triazolo[4,5-b]-pyridinium 3-oxid hexafluorophosphate); Me methyl; p-CeH 4 para- substituted phenylene; Ph phenyl; p-TSA /PTSA p-Toluenesulfonic acid; TBAF tetrabutylammonium fluoride; TEMPO (2,2,6,6-tetramethyl-piperidin-i-yl)oxyl; TFA trifluoro-acetic acid; THF tetrahydrofuran and TIPS-C1 triisopropylsilyl chloride.

“Substituted”, when used in connection with a chemical substituent or moiety (e.g., an alkyl group), means that one or more hydrogen atoms of the substituent or moiety have been replaced with one or more non-hydrogen atoms or groups, provided that valence requirements are met and that a chemically stable compound results from the substitution. “Optionally substituted” refers to a parent group which may be unsubstituted or which maybe substituted with one or more substituents. Suitably, unless otherwise specified, when optional substituents are present the optional substituted parent group comprises from one to three optional substituents. Where a group may be“optionally substituted with l, 2 or 3 groups”, this means that the group may be substituted with o, l, 2 or 3 of the optional substituents. Suitably, the group is substituted with 1, 2 or 3 of the optional substituents. Where a group is“optionally substituted with one or two optional substituents”, this means that the group maybe substituted with o, 1 or 2 of the optional substituents. Suitably, the group maybe optionally substituted with o or 1 optional substituents. In some aspects, suitably the group is not optionally substituted. In other aspects, suitably the group is substituted with 1 of the optional substituents.

Optional substituents may be selected from C - 2 alkyl, C 2-7 alkenyl, C 2-7 alkynyl, C - i2 alkoxy, C 5-20 aryl, C 3-i0 cycloalkyl, C 3-i0 cycloalkenyl, C 3-i0 cycloalkynyl, C 3-20

heterocyclyl, C 3-20 heteroaryl, acetal, acyl, acylamido, acyloxy, amidino, amido, amino, aminocarbonyloxy, azido, carboxy, cyano, ether, formyl, guanidino, halo, hemiacetal, hemiketal, hydroxamic acid, hydroxyl, imidic acid, imino, ketal, nitro, nitroso, oxo, oxycarbonyl, oxycarboyloxy, sulfamino, sulfamyl, sulfate, sulfhydryl, sulfmamino, sulfinate, sulfino, sulfinyl, sulfinyloxy, sulfo, sulfonamido, sulfonamino, sulfonate, sulfonyl, sulfonyloxy, uredio groups. In some aspects, the optional substituents are 1, 2 or 3 optional substituents independently selected from OH, C - i2 alkyl, 00- 12 alkyl, R A and halogen. More suitably, the optional substituents are selected from OH, C - i2 alkyl and 0C - i2 alkyl; more suitably, the optional substituents are selected from 0- 12 alkyl and 0C - i2 alkyl.

“Independently selected” is used in the context of statement that, for example,“each Ru, and R I7 are independently selected from H and CV 12 alkyl,...” and means that each instance of the functional group, e.g. R 16 , is selected from the listed options

independently of any other instance of R 16 or R I7 in the compound. Hence, for example, H may be selected for the first instance of R 16 in the compound; methyl may be selected for the next instance of R 16 in the compound; and ethyl may be selected for the first instance of R iy in the compound.

C - 2 alkyl: refers to straight chain and branched saturated hydrocarbon groups, generally having from 1 to 12 carbon atoms; suitably a C -u alkyl; suitably a C - 0 alkyl; suitably a C -9 alkyl; suitably a C - 8 alkyl; more suitably a C -7 alkyl; more suitably a C -6 alkyl; more suitably a C -5 alkyl; more suitably a C -4 alkyl; more suitably a C -3 alkyl. Examples of alkyl groups include methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl, i- butyl, t-butyl, pent-i-yl, pent-2-yl, pent-3-yl, 3-methylbut-i-yl, 3-methylbut-2-yl, 2- methylbut-2-yl, 2,2,2-trimethyleth-i-yl, n-hexyl, n-heptyl, and the like.

“Alkylene” refers to a divalent radical derived from an alkane which may be a straight chain or branched, as exemplified by -CH 2 CH 2 CH 2 CH 2 -.

The term“amino acid” refers to both the twenty“canonical” or“natural” amino acids, as well“non-canonical” amino acids, also referred to as“unnatural” amino acids, such as modified or synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function similarly to naturally occurring amino acids. Naturally occurring amino acids are those encoded by the genetic code, i.e. they are amino acids selected from alanine, argenine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine and valine. Modified amino acids include, e.g., hydroxyproline, g-carboxyglutamate, and O-phosphoserine. Amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, e.g., an a carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs may have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid. Amino acid mimetics refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that functions similarly to a naturally occurring amino acid.

“C 6-26 aralkyl” refers to an arylalkyl group having 6 to 26 carbon atoms and comprising an alkyl group substituted with an aiyl group. Suitably the alkyl group is a C -6 alkyl group and the aryl group is phenyl. Examples of C 6-26 aralkyl include benzyl and phenethyl. In some cases the C 6-26 aralkyl group maybe optionally substituted and an example of an optionally substituted C 6-26 aralkyl group is 4-methoxylbenzyl.

“C5-20 Aryl”: refers to fully unsaturated monocyclic, bicyclic and polycyclic aromatic hydrocarbons having at least one aromatic ring and having a specified number of carbon atoms that comprise their ring members (e.g., C 5-20 aryl refers to an aryl group having from 5 to 20 carbon atoms as ring members). The aiyl group maybe attached to a parent group or to a substrate at any ring atom and may include one or more non- hydrogen substituents unless such attachment or substitution would violate valence requirements. Suitably, a C 6-i4 aryl is selected from a C 6-i2 aryl, more suitably, a C 6-i0 aryl. Examples of aryl groups include phenyl.

“Arylene” refers to a divalent radical derived from an aryl group, e.g. -O ό H 4 - which is the arylene derived from phenyl.

“C 3-8 cycloalkyl” or“3- to 8-membered cycloalkyl” means a closed ring of carbon atoms having 3 to 8 carbon atoms, preferably 3 to 7 carbon atoms, more preferably 3 to 6 carbon atoms and encompasses, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.

“C 3-8 cycloalkylene” or“3- to 8-membered cycloalkylene” refers to a divalent radical derived from a cycloalkyl group, e.g. -C 6 H 12 -.

Halogen or halo: refers to a group selected from F, Cl, Br, and I. Suitably, the halogen or halo is F or Cl.

“C 5-10 heteroaryl” or“5- to 10-membered heteroaryl”: refers to unsaturated monocyclic or bicyclic aromatic groups comprising from 5 to 10 ring atoms, whether carbon or heteroatoms, of which from 1 to 5 are ring heteroatoms. Suitably, any monocyclic heteroaryl ring has from 5 to 6 ring atoms and from 1 to 3 ring heteroatoms. Suitably each ring heteroatom is independently selected from nitrogen, oxygen, and sulfur. The bicyclic rings include fused ring systems and, in particular, include bicyclic groups in which a monocyclic heterocycle comprising 5 ring atoms is fused to a benzene ring.

The heteroaryl group may be attached to a parent group or to a substrate at any ring atom and may include one or more non-hydrogen substituents unless such attachment or substitution would violate valence requirements or result in a chemically unstable compound.

Examples of monocyclic heteroaryl groups include, but are not limited to, those derived from:

Nu pyrrole, pyridine;

(X: furan;

Su thiophene;

N Ou oxazole, isoxazole, isoxazine; N 2 0 I : oxadiazole (e.g. i-oxa-2,3-diazolyl, i-oxa-2,4-diazolyl, i-oxa-2,5-diazolyl, l-oxa- 3,4-diazolyl);

N 3 O I : oxatriazole;

N 1 S 1 : thiazole, isothiazole;

N 2 : imidazole, pyrazole, pyridazine, pyrimidine, pyrazine;

N 3 : triazole, triazine; and,

N 4 : tetrazole.

Examples of heteroaryl which comprise fused rings, include, but are not limited to, those derived from:

(X: benzofuran, isobenzofuran;

N : indole, isoindole, indolizine, isoindoline;

S : benzothiofuran;

N 1 O 1 : benzoxazole, benzisoxazole;

N 1 S 1 : benzothiazole;

N 2 : benzimidazole, indazole;

0 2 : benzodioxole;

N 2 0 I : benzofurazan;

N 2 S I : benzothiadiazole;

N 3 : benzotriazole; and

N 4 : purine (e.g., adenine, guanine), pteridine;

“heteroarylene” refers to a divalent radical derived from a heteroaryl group (such as those described above) as exemplified by pyridinyl -[C 5 H 3 N]-. Heteroarylenes maybe monocyclic, bicyclic, or tricyclic ring systems. Representative heteroarylenes, are not limited to, but may be selected from triazolylene, tetrazolylene, oxadiazolylene, pyridylene, furylene, benzofuranylene, thiophenylene, benzothiophenylene, quinolinylene, pyrrolylene, indolylene, oxazolylene, benzoxazolylene, imidazolylene, benzimidazolylene, thiazolylene, benzothiazolylene, isoxazolylene, pyrazolylene, isothiazolylene, pyridazinylene, pyrimidinylene, pyrazinylene, triazinylene, cinnolinylene, phthalazinylene, quinazolinylene, pyrimidylene, azepinylene, oxepinylene, and quinoxalinylene. Heteroarylenes are optionally substituted.

“C 6-i6 heteroarylalkyl” refers to an alkyl group substituted with a heteroaryl group. Suitably the alkyl is a C -6 alkyl group and the heteroaryl group is C 5-i0 heteroaryl as defined above. Examples of C 6-i6 heteroarylalkyl groups include pyrrol-2-ylmethyl, pyrrol-3-ylmethyl, pyrrol-4-ylmethyl, pyrrol-3-ylethyl, pyrrol-4-ylethyl, imidazol-2- ylmethyl, imidazol-4-ylmethyl, imidazol-4-ylethyl, thiophen-3-ylmethyl, furan-3- ylmethyl, pyridin-2-ylmethyl, pyridin-2-ylethyl, thiazol-2-ylmethyl, thiazol-4-ylmethyl, thiazol-2-ylethyl, pyrimidin-2-ylpropyl, and the like.

“C3- 2 0 heterocyclyl”: refers to saturated or partially unsaturated monocyclic, bicyclic or polycyclic groups having ring atoms composed of 3 to 20 ring atoms, whether carbon atoms or heteroatoms, of which from 1 to 10 are ring heteroatoms. Suitably, each ring has from 3 to 7 ring atoms and from 1 to 4 ring heteroatoms (e.g., suitably C 3-5 heterocyclyl refers to a heterocyclyl group having 3 to 5 ring atoms and 1 to 4 heteroatoms as ring members). The ring heteroatoms are independently selected from nitrogen, oxygen, and sulphur.

As with bicyclic cycloalkyl groups, bicyclic heterocyclyl groups may include isolated rings, spiro rings, fused rings, and bridged rings. The heterocyclyl group maybe attached to a parent group or to a substrate at any ring atom and may include one or more non-hydrogen substituents unless such attachment or substitution would violate valence requirements or result in a chemically unstable compound.

Examples of monocyclic heterocyclyl groups include, but are not limited to, those derived from:

N : aziridine, azetidine, pyrrolidine, pyrroline, 2H-pyrrole or 3H-pyrrole, piperidine, dihydropyridine, tetrahydropyridine, azepine;

(X: oxirane, oxetane, tetrahydrofuran, dihydrofuran, tetrahydropyran, dihydropyran, pyran, oxepin;

S : thiirane, thietane, tetrahydrothiophene, tetrahydrothiopyran, thiepane;

0 2 : dioxoiane, dioxane, and dioxepane;

0 3 : trioxane;

N 2 : imidazoiidine, pyrazolidine, imidazoline, pyrazoline, piperazine:

N 1 O 1 : tetrahydrooxazole, dihydrooxazole, tetrahydroisoxazole, dihydroisoxazole, morpholine, tetrahydrooxazine, dihydrooxazine, oxazine;

N 1 S 1 : thiazoline, thiazolidine, thiomorpholine;

N 2 0 I : oxadiazine;

O 1 S 1 : oxathiole and oxathiane (thioxane); and

N 1 O 1 S 1 : oxathiazine.

Examples of substituted monocyclic heterocyclyl groups include those derived from saccharides, in cyclic form, for example, furanoses, such as arabinofuranose, lyxofuranose, ribofuranose, and xylofuranse, and pyranoses, such as aliopyranose, altropyranose, glucopyranose, mannopyranose, gulopyranose, idopyranose, galactopyranose, and talopyranose.

“3- to 8-membered heterocycloalkyl,” refers to a closed ring of comprising carbon atoms and heteroatoms. The heterocyclosalkyl may comprise one, or three

heteroatoms. Suitably the heteroatoms are selected from the group consisting of O, N and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized. Heterocycloalkyl groups typically comprise from 3 to 8 ring member atoms, preferably from 3 to 7 ring member atoms, more preferably from 3 to 6 ring member atoms, and most preferably from 3 to 5 ring member atoms. Heteroalkyl groups may be optionally substituted.

“heterocycloalkylene” refers to a divalent group derived from heteroalkyl (as discussed above). For heterocycloalkylene groups, heteroatoms can also occupy either or both of the positions where the heterocycloalkylene group is attached to the rest of the compound. Heteroalkylene groups maybe optionally substituted.

“Nucleic acid”, refers to a linear polymer of nucleosides (including deoxyribo- nucleosides, ribonucleosides, or analogs thereof) joined by inter-nucleosidic linkages. Nucleic acid may encompass the term“polynucleotide” as well as“oligonucleotide”.

The linear polymer may be represented by a sequence of letters, such as“ATGCCTG,” where it will be understood that the nucleotides are in 5' to 3' order from left to right and that“A” denotes deoxyadenosine,“C” denotes deoxycytidine,“G” denotes deoxyguanosine, and“T” denotes deoxythymidine, unless otherwise noted. Another natural nucleotide is“U”, denoting uridine. The letters A, C, G, T and U can be used to refer to the bases themselves, to nucleosides, or to nucleotides comprising the bases, as is standard in the art. In naturally occurring nucleic acids, the inter-nucleoside linkage is typically a phosphodiester bond, and the subunits are referred to as“nucleotides.” Nucleic acids may also include other inter-nucleoside linkages, such as phosphoro- thioate linkages, and the like. Such analogs of nucleotides that do not include a phosphate group are considered to fall within the scope of the term“nucleotid”" as used herein, and nucleic acids comprising one or more inter-nucleoside linkages that are not phosphodiester linkages are still referred to as "polynucleotides”,“oligonucleotides”, etc.

Nitrogen protecting groups Nitrogen protecting groups are well known in the art and are groups that block or protect the nitrogen groups from further reaction. Nitrogen protecting groups are exemplified by carbamates, such as methyl or ethyl carbamate, 9-fluorenylmethyloxy- carbonyl (Fmoc), substituted ethyl carbamates, carbamates cleaved by 1,6-beta- elimination, ureas, amides, peptides, alkyl and aryl derivatives. Carbamate protecting groups have the general formula:

In this specification a zig-zag line (or wavy line n / ' ) indicates the point of

attachment of the shown group (e.g. the protecting group above) to the rest of the compound of formula (I). Suitable nitrogen protecting groups may be selected from acetyl, trifluoroacetyl, t-butyloxy-carbonyl (BOC), benzyloxycarbonyl (Cbz) and 9- fluorenylmethyloxy-carbonyl (Fmoc).

A large number of possible carbamate nitrogen protecting groups are listed on pages 706 to 771 of Wuts, P.G.M. and Greene, T.W., Protective Groups in Organic Synthesis, 4 th Edition, Wiley-lnterscience, 2007, and in P. Kocienski, Protective Groups, 3rd Edition (2005) which are incorporated herein by reference.

Particularly preferred protecting groups include Alloc (allyloxycarbonyl), Troc (2,2,2- Trichloroethyl carbonate), Teoc [2-(Trimethylsilyl)ethoxycarbony], BOC (tert- butyloxycarbonyl), Doc (2,4-dimethylpent-3-yloxycarbonyl), Hoc (cyclohexyloxy- carbonyl), TcBOC (2,2,2-trichloro-tert-butyloxycarbonyl), Fmoc (9- fluorenylmethyloxycarbonyl), l-Adoc (i-Adamantyloxycarbonyl) and 2-Adoc (2- adamantyloxycarbonyl).

Hydroxyl protecting groups

Hydroxyl protecting groups are well known in the art, a large number of suitable groups are described on pages 16 to 366 of Wuts, P.G.M. and Greene, T.W., Protective Groups in Organic Synthesis, 4 th Edition, Wiley-lnterscience, 2007, and in P. Kocienski, Protective Groups, 3rd Edition (2005) which are incorporated herein by reference.

Classes of particular interest include silyl ethers, methyl ethers, alkyl ethers, benzyl ethers, esters, benzoates, carbonates, and sulfonates. Particularly preferred protecting groups include THP ( tetrahydropyranyl ether). An“acceptor human framework” for the purposes herein is a framework comprising the amino acid sequence of a light chain variable domain (VL) framework or a heavy chain variable domain (VH) framework derived from a human immunoglobulin framework or a human consensus framework, as defined below. An acceptor human framework "derived from" a human immunoglobulin framework or a human consensus framework may comprise the same amino acid sequence thereof, or it may contain amino acid sequence changes. In some embodiments, the number of amino acid changes are 10 or less, 9 or less, 8 or less, 7 or less, 6 or less, 5 or less, 4 or less, 3 or less, or 2 or less. In some embodiments, the VL acceptor human framework is identical in sequence to the VL human immunoglobulin framework sequence or human consensus framework sequence.

“Affinity” refers to the strength of the sum total of noncovalent interactions between a single binding site of a molecule (e.g., an antibody) and its binding partner (e.g., an antigen). Unless indicated otherwise, as used herein, "binding affinity" refers to intrinsic binding affinity which reflects a 1 : 1 interaction between members of a binding pair (e.g., antibody and antigen). The affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (Kd). Affinity can be measured by common methods known in the art, including those described herein. Specific illustrative and exemplary embodiments for measuring binding affinity are described in the following.

An“affinity matured” antibody refers to an antibody with one or more alterations in one or more hypervariable regions (HVRs), compared to a parent antibody which does not possess such alterations, such alterations resulting in an improvement in the affinity of the antibody for antigen.

The term“antibody” is used herein in the broadest sense and encompasses various antibody structures, including but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they exhibit the desired antigen-binding activity.

An“antibody fragment” refers to a molecule other than an intact antibody that comprises a portion of an intact antibody and that binds the antigen to which the intact antibody binds. Examples of antibody fragments include but are not limited to Fv, Fab, Fab', Fab'-SH, F(ab')2; diabodies; linear antibodies; single-chain antibody molecules (e.g. scFv); and multispecific antibodies formed from antibody fragments. The term“chimeric” antibody refers to an antibody in which a portion of the heavy and/or light chain is derived from a particular source or species, while the remainder of the heavy and/or light chain is derived from a different source or species.

The“class” of an antibody refers to the type of constant domain or constant region possessed by its heavy chain. There are five major classes of antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgGi, IgG2, IgG3, IgG4, IgAi, and IgA2. The heavy chain constant domains that correspond to the different classes of immunoglobulins are called a, d, e, g, and m, respectively.

The term“cytotoxic agent” as used herein refers to a substance that inhibits or prevents a cellular function and/ or causes cell death or destruction. Cytotoxic agents include, but are not limited to, radioactive isotopes (e.g., At 211 , 1 131 , 1 125 , Y 90 , Re 186 , Rel88 Sm 153 , Bi 212 , P 32 , Pb 212 and radioactive isotopes of Lu); chemotherapeutic agents or drugs (e.g., methotrexate, adriamicin, vinca alkaloids (vincristine, vinblastine, etoposide), doxorubicin, melphalan, mitomycin C, chlorambucil, daunorubicin or other

intercalating agents); growth inhibitory agents; enzymes and fragments thereof such as nucleolytic enzymes; antibiotics; toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, including fragments and/or variants thereof; and the various antitumor or anticancer agents disclosed below.

By“co-administering” is meant intravenously administering two (or more) drugs during the same administration, rather than sequential infusions of the two or more drugs. Generally, this will involve combining the two (or more) drugs into the same IV bag prior to co-administration thereof.

A drug that is administered“concurrently” with one or more other drugs is

administered during the same treatment cycle, on the same day of treatment as the one or more other drugs, and, optionally, at the same time as the one or more other drugs. For instance, for cancer therapies given every 3 weeks, the concurrently administered drugs are each administered on day-i of a 3-week cycle.

A“chemotherapeutic agent” refers to a chemical compound useful in the treatment of cancer. Examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide (CYTOXAN®); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide and trimethylomelamine; acetogenins (especially bullatacin and bullatacinone); delta-9- tetrahydrocannabinol (dronabinol, MARINOL®); beta-lapachone; lapachol;

colchicines; betulinic acid; a camptothecin (including the synthetic analogue topotecan (HYCAMTIN®), CPT-11 (irinotecan, CAMPTOSAR®), acetylcamptothecin, scopolectin, and 9-aminocamptothecin); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); podophyllotoxin;

podophyllinic acid; teniposide; cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CB1-TM1); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, chlorophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosoureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and

ranimnustine; antibiotics such as the enediyne antibiotics (e.g., calicheamicin, especially calicheamicin gammail and calicheamicin omegali (see, e.g., Nicolaou et ah, Angew. Chem Inti. Ed. Engl., 33 : 183-186 (1994)); CDP323, an oral alpha-4 integrin inhibitor; dynemicin, including dynemicin A; an esperamicin; as well as

neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including ADRIAMYCIN®, morpholino-doxorubicin, cyanomorpholino- doxorubicin, 2- pyrrolino-doxorubicin, doxorubicin HC1 liposome injection (DOXIL®), liposomal doxorubicin TLC D-99 (MYOCET®), peglylated liposomal doxorubicin (CAELYX®), and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, porfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate, gemcitabine (GEMZAR®), tegafur (UFTORAL®), capecitabine (XELODA®), an epothilone, and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6- mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone;

aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfornithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; 2- ethylhydrazide; procarbazine; PSK® polysaccharide complex (JHS Natural Products, Eugene, OR); razoxane; rhizoxin; sizofiran; spirogermanium; tenuazonic acid;

triaziquone; 2,2’,2’-trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethan; vindesine (ELDISINE®,

FILDESIN®); dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside (“Ara-C"); thiotepa; taxoid, e.g., paclitaxel (TAXOL®), albumin-engineered nanoparticle formulation of paclitaxel (ABRAXANETM), and docetaxel (TAXOTERE®); chloranbucil; 6-thioguanine; mercaptopurine;

methotrexate; platinum agents such as cisplatin, oxaliplatin (e.g., ELOXATIN®), and carboplatin; vincas, which prevent tubulin polymerization from forming microtubules, including vinblastine (VELBAN®), vincristine (ONCOVIN®), vindesine (ELDISINE®, FILDESIN®), and vinorelbine (NAVELBINE®); etoposide (VP- 16); ifosfamide;

mitoxantrone; leucovorin; novantrone; edatrexate; daunomycin; aminopterin;

ibandronate; topoisomerase inhibitor RFS 2000; difluoromethyl ornithine (DMFO); retinoids such as retinoic acid, including bexarotene (TARGRETIN®);

bisphosphonates such as clodronate (for example, BONEFOS® or OSTAC®), etidronate (DIDROCAL®), NE-58095, zoledronic acid/zoledronate (ZOMETA®), alendronate (FOSAMAX®), pamidronate (AREDIA®), tiludronate (SKELID®), or risedronate (ACTONEL®); troxacitabine (a 1,3- dioxolane nucleoside cytosine analog); antisense oligonucleotides, particularly those that inhibit expression of genes in signaling pathways implicated in aberrant cell proliferation, such as, for example, PKC- alpha, Raf, H-Ras, and epidermal growth factor receptor (EGF- R); vaccines such as THERATOPE® vaccine and gene therapy vaccines, for example, ALLOVECTIN® vaccine, LEUVECTIN® vaccine, and VAXID® vaccine; topoisomerase 1 inhibitor (e.g., LURTOTECAN®); rmRH (e.g., ABARELIX®); BAY439006 (sorafenib; Bayer); SU- 11248 (sunitinib, SUTENT®, Pfizer); perifosine, COX-2 inhibitor (e.g., celecoxib or etoricoxib), proteosome inhibitor (e.g., PS341); bortezomib (VELCADE®); CCI-779; tipifarnib (R11577); orafenib, ABT510; Bcl-2 inhibitor such as oblimersen sodium (GENASENSE®); pixantrone; EGFR inhibitors; tyrosine kinase inhibitors; serine- threonine kinase inhibitors such as rapamycin (sirolimus, RAPAMUNE®); farnesyltransferase inhibitors such as lonafarnib (SCH 6636, SARASARTM); and pharmaceutically acceptable salts, acids or derivatives of any of the above; as well as combinations of two or more of the above such as CHOP, an abbreviation for a combined therapy of cyclophosphamide, doxorubicin, vincristine, and prednisolone; and FOLFOX, an abbreviation for a treatment regimen with oxaliplatin

(ELOXATINTM) combined with 5-FU and leucovorin.

Chemotherapeutic agents as defined herein include“anti-hormonal agents” or “endocrine therapeutics” which act to regulate, reduce, block, or inhibit the effects of hormones that can promote the growth of cancer. They may be hormones themselves, including, but not limited to: anti-estrogens with mixed agonist/antagonist profile, including, tamoxifen (NOLVADEX®), 4-hydroxytamoxifen, toremifene

(FARESTON®), idoxifene, droloxifene, raloxifene (EVISTA®), trioxifene, keoxifene, and selective estrogen receptor modulators (SERMs) such as SERM3; pure anti- estrogens without agonist properties, such as fulvestrant (FASLODEX®), and EM800 (such agents may block estrogen receptor (ER) dimerization, inhibit DNA binding, increase ER turnover, and/or suppress ER levels); aromatase inhibitors, including steroidal aromatase inhibitors such as formestane and exemestane (AROMASIN®), and nonsteroidal aromatase inhibitors such as anastrazole (ARFMIDEX®), letrozole (FEMARA®) and aminoglutethimide, and other aromatase inhibitors include vorozole (RIVISOR®), megestrol acetate (MEGASE®), fadrozole, and 4(5)-imidazoles;

lutenizing hormone-releaseing hormone agonists, including leuprolide (LUPRON® and ELIGARD®), goserelin, buserelin, and tripterelin; sex steroids, including progestines such as megestrol acetate and medroxyprogesterone acetate, estrogens such as diethylstilbestrol and premarin, and androgens/retinoids such as

fluoxymesterone, all transretionic acid and fenretinide; onapristone; anti- progesterones; estrogen receptor down- regulators (ERDs); anti-androgens such as flutamide, nilutamide and bicalutamide; and pharmaceutically acceptable salts, acids or derivatives of any of the above; as well as combinations of two or more of the above.

“Drug”,“drug substance”,“active pharmaceutical ingredient”, and the like, refer to a compound (e.g., compounds of Formula (I) and compounds specifically named above) that may be used for treating a subject in need of treatment.

“Effector functions” refer to those biological activities attributable to the Fc region of an antibody, which vary with the antibody isotype. Examples of antibody effector functions include: Clq binding and complement dependent cytotoxicity (CDC); Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC);

phagocytosis; down regulation of cell surface receptors (e.g. B cell receptor); and B cell activation.

The term“epitope” refers to the particular site on an antigen molecule to which an antibody binds.

The“epitope 4D5” or“4D5 epitope” or“4D5” is the region in the extracellular domain of HER2 to which the antibody 4D5 (ATCC CRL 10463) and trastuzumab bind. This epitope is close to the transmembrane domain of HER2, and within domain IV of HER2. To screen for antibodies which bind to the 4D5 epitope, a routine cross-blocking assay such as that described in Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory, Ed Harlow and David Lane (1988), can be performed. Alternatively, epitope mapping can be performed to assess whether the antibody binds to the 4D5 epitope of HER2 (e.g. any one or more residues in the region from about residue 550 to about residue 610, inclusive, of HER2 (SEQ ID NO: 39).

The“epitope 2C4” or“2C4 epitope” is the region in the extracellular domain of HER2 to which the antibody 2C4 binds. In order to screen for antibodies which bind to the 2C4 epitope, a routine cross-blocking assay such as that described in Antibodies, A

Laboratory Manual, Cold Spring Harbor Laboratory, Ed Harlow and David Lane (1988), can be performed. Alternatively, epitope mapping can be performed to assess whether the antibody binds to the 2C4 epitope of HER2. Epitope 2C4 comprises residues from domain II in the extracellular domain of HER2. The 2C4 antibody and pertuzumab bind to the extracellular domain of HER2 at the junction of domains I, II and III (Franklin et al. Cancer Cell 5:317-328 (2004)). Anti-HER2 murine antibody 7C2 binds to an epitope in domain I of HER2. See, e.g., PCT Publication No. WO 98/17797. This epitope is distinct from the epitope bound by trastuzumab, which binds to domain IV of HER2, and the epitope bound by pertuzumab, which binds to domain II of HER2. By binding domain IV, trastuzumab disrupts ligand- independent HER2-HER3 complexes, thereby inhibiting downstream signaling (e.g. PI3K/AKT). In contrast, pertuzumab binding to domain II prevents ligand-driven HER2 interaction with other HER family members (e.g. HER3, HERl or HER4), thus also preventing downstream signal transduction. Binding of MAb 7C2 to domain I does not result in interference of trastuzumab or pertuzumab binding to domains IV and II, respectively, thereby offering the potential of combining a MAb 7C2 ADC with trastuzumab, trastuzumab emtansine (T-DM-i), and/or pertuzumab. Murine antibody 7C2, 7C2.B9, is described in PCT Publication No. WO 98/17797. An anti-HER2 7C2 humanized antibody is disclosed in WO2016/040723 Al.

“Excipient” refers to any substance that may influence the bioavailability of a drug, but is otherwise pharmacologically inactive.

The term“Fc region” herein is used to define a C-terminal region of an immunoglobulin heavy chain that contains at least a portion of the constant region. The term includes native sequence Fc regions and variant Fc regions. In one embodiment, a human IgG heavy chain Fc region extends from Cys226, or from Pro230, to the carboxyl-terminus of the heavy chain. However, the C-terminal lysine (Lys447) of the Fc region may or may not be present. Unless otherwise specified herein, numbering of amino acid residues in the Fc region or constant region is according to the EU numbering system, also called the EU index, as described in Rabat et al., Sequences of Proteins of

Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD, 1991.

“Framework” or“FR” refers to variable domain residues other than hypervariable region (HVR) residues. The FR of a variable domain generally consists of four FR domains: FRi, FR2, FR3, and FR4. Accordingly, the HVR and FR sequences generally appear in the following sequence in VH (or VL): FRI-HI(LI)-FR2-H2(L2)-FR3-H3(L3)- FR4.

The terms“full length antibody,”“intact antibody,” and“whole antibody” are used herein interchangeably to refer to an antibody having a structure substantially similar to a native antibody structure or having heavy chains that contain an Fc region as defined herein.

The terms“host cell,”“host cell line,” and“host cell culture” are used interchangeably and refer to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells. Host cells include“transformants” and“transformed cells,” which include the primary transformed cell and progeny derived therefrom without regard to the number of passages. Progeny may not be completely identical in nucleic acid content to a parent cell, but may contain mutations. Mutant progeny that have the same function or biological activity as screened or selected for in the originally transformed cell are included herein. A“human antibody” is one which possesses an amino acid sequence which corresponds to that of an antibody produced by a human or a human cell or derived from a non- human source that utilizes human antibody repertoires or other human antibody- encoding sequences. This definition of a human antibody specifically excludes a humanized antibody comprising non-human antigen-binding residues.

A“human consensus framework” is a framework which represents the most commonly occurring amino acid residues in a selection of human immunoglobulin VL or VH framework sequences. Generally, the selection of human immunoglobulin VL or VH sequences is from a subgroup of variable domain sequences. Generally, the subgroup of sequences is a subgroup as in Rabat et ah, Sequences of Proteins of Immunological Interest, Fifth Edition, NIH Publication 91-3242, Bethesda MD (1991), vols. 1-3. In one embodiment, for the VL, the subgroup is subgroup kappa I as in Rabat et ah, supra. In one embodiment, for the VH, the subgroup is subgroup III as in Rabat et ah, supra.

A“humanized” antibody refers to a chimeric antibody comprising amino acid residues from non-human HVRs and amino acid residues from human FRs. In certain embodiments, a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the HVRs (e.g.,

CDRs) correspond to those of a non-human antibody, and all or substantially all of the FRs correspond to those of a human antibody. A humanized antibody optionally may comprise at least a portion of an antibody constant region derived from a human antibody. A“humanized form” of an antibody, e.g., a non-human antibody, refers to an antibody that has undergone humanization.

The term“hypervariable region” or“HVR,” as used herein, refers to each of the regions of an antibody variable domain which are hypervariable in sequence and/ or form structurally defined loops (“hypervariable loops”). Generally, native four-chain antibodies comprise six HVRs; three in the VH (Hi, H2, H3), and three in the VL (Li, L2, L3). HVRs generally comprise amino acid residues from the hypervariable loops and/or from the“complementarity determining regions” (CDRs), the latter being of highest sequence variability and/ or involved in antigen recognition. Exemplaiy hypervariable loops occur at amino acid residues 26-32 (Li), 50-52 (L2), 91-96 (L3), 26-32 (Hi), 53-55 (H2), and 96-101 (H3). (Chothia and Lesk, J. Mol. Biol. 196:901-917 (1987).) Exemplaiy CDRs (CDR-Li, CDR-L2, CDR-L3, CDR-Hi, CDR-H2, and CDR- H3) occur at amino acid residues 24-34 of Li, 50-56 of L2, 89-97 of L3, 31-35B of Hi, 50-65 of H2, and 95-102 of H3. (Rabat et ah, Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD (1991).) With the exception of CDRi in VH, CDRs generally comprise the amino acid residues that form the hypervariable loops. CDRs also comprise“specificity

determining residues,” or“SDRs,” which are residues that contact antigen. SDRs are contained within regions of the CDRs called abbreviated-CDRs, or a- CDRs. Exemplary a-CDRs (a-CDR-Li, a-CDR-L2, a-CDR-L3, a-CDR-Hi, a-CDR-H2, and a-CDR-H3) occur at amino acid residues 31-34 of LI, 50-55 of L2, 89-96 of L3, 31-35B of HI, 50-58 of H2, and 95-102 of H3. (See Almagro and Fransson, Front. Biosci. 13 : 1619- 1633 (2008).) Unless otherwise indicated, HVR residues and other residues in the variable domain (e.g., FR residues) are numbered herein according to Rabat et ah, supra.

An“immunoconjugate” is an antibody conjugated to one or more heterologous molecule(s), including but not limited to a cytotoxic agent.

The term“immunosuppressive agent” as used herein for adjunct therapy refers to substances that act to suppress or mask the immune system of the mammal being treated herein. This would include substances that suppress cytokine production, down-regulate or suppress self-antigen expression, or mask the MHC antigens.

Examples of such agents include 2-amino-6-aryl-5-substituted pyrimidines (see U.S. Pat. No. 4,665,077); non-steroidal anti-inflammatory drugs (NSAIDs); ganciclovir, tacrolimus, glucocorticoids such as cortisol or aldosterone, anti-inflammatory agents such as a cyclooxygenase inhibitor, a 5- lipoxygenase inhibitor, or a leukotriene receptor antagonist; purine antagonists such as azathioprine or mycophenolate mofetil (MMF); alkylating agents such as cyclophosphamide; bromocryptine; danazol;

dapsone; glutaraldehyde (which masks the MHC antigens, as described in U.S. Pat. No. 4,120,649); anti-idiotypic antibodies for MHC antigens and MHC fragments;

cyclosporin A; steroids such as corticosteroids or glucocorticosteroids or glucocorticoid analogs, e.g., prednisone, methylprednisolone, including SOLU-MEDROL®

methylprednisolone sodium succinate, and dexamethasone; dihydrofolate reductase inhibitors such as methotrexate (oral or subcutaneous); anti-malarial agents such as chloroquine and hydroxychloroquine; sulfasalazine; leflunomide; cytokine or cytokine receptor antibodies including anti-interferon-alpha, -beta, or -gamma antibodies, anti- tumor necrosis factor(TNF)-alpha antibodies (infliximab (REMICADE®) or adalimumab), anti-TNF-alpha immunoadhesin (etanercept), anti-TNF-beta antibodies, anti-interleukin-2 (IL-2) antibodies and anti-IL-2 receptor antibodies, and anti- interleukin-6 (IL-6) receptor antibodies and antagonists (such as ACTEMRA™

(tocilizumab)); anti-LFA-i antibodies, including anti-CDua and anti-CDi8 antibodies; anti-L3T4 antibodies; heterologous anti-lymphocyte globulin; pan-T antibodies, preferably anti-CD3 or anti-CD4/CD4a antibodies; soluble peptide containing a LFA-3 binding domain (WO 90/08187); streptokinase; transforming growth factor-beta (TGF-beta); streptodornase; RNA or DNA from the host; FK506; RS-61443;

chlorambucil; deoxyspergualin; rapamycin; T-cell receptor (Cohen et al, U.S. Pat. No. 5,114,721); T-cell receptor fragments (Offner et al, Science, 251 : 430-432 (1991); WO 90/11294; Ianeway, Nature, 341 : 482 (1989); and WO 91/01133); BAFF antagonists such as BAFF antibodies and BR3 antibodies and ZTNF4 antagonists (for review, see Mackay and Mackay, Trends Immunol, 23 : 113-5 (2002) and see also definition below); biologic agents that interfere with T cell helper signals, such as anti- CD40 receptor or anti-CD40 ligand (CD 154), including blocking antibodies to CD40-CD40 ligand (e.g., Durie et al, Science, 261 : 1328-30 (1993); Mohan et al, J. Immunol, 154: 1470- 80 (1995)) and CTLA4-Ig (Finck et al, Science, 265: 1225-7 (1994)); and T-cell receptor antibodies (EP 340,109) such as T10B9. Some preferred immunosuppressive agents herein include cyclophosphamide, chlorambucil, azathioprine, leflunomide, MMF, or methotrexate.

An“isolated antibody” is one which has been separated from a component of its natural environment. In some embodiments, an antibody is purified to greater than 95% or 99% purity as determined by, for example, electrophoretic (e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatographic (e.g., ion exchange or reverse phase HPLC). For review of methods for assessment of antibody purity, see, e.g., Flatman et al., J. Chromatogr. B 848:79-87 (2007).

An“isolated nucleic acid" refers to a nucleic acid molecule that has been separated from a component of its natural environment. An isolated nucleic acid includes a nucleic acid molecule contained in cells that ordinarily contain the nucleic acid molecule, but the nucleic acid molecule is present extrachromosomally or at a chromosomal location that is different from its natural chromosomal location.

“Isolated nucleic acid encoding an antibody” refers to one or more nucleic acid molecules encoding antibody heavy and light chains (or fragments thereof), including such nucleic acid molecule(s) in a single vector or separate vectors, and such nucleic acid molecule(s) present at one or more locations in a host cell.

The term“HER2,” as used herein, refers to any native, mature HER2 which results from processing of a HER2 precursor protein in a cell. The term includes HER2 from any vertebrate source, including mammals such as primates (e.g. humans and cynomolgus monkeys) and rodents (e.g., mice and rats), unless otherwise indicated. The term also includes naturally occurring variants of HER2, e.g., splice variants or allelic variants. The amino acid sequence of an exemplary human HER2 precursor protein, with signal sequence (with signal sequence, amino acids 1-22) is shown in SEQ ID NO: 64. The amino acid sequence of an exemplary mature human HER2 is amino acids 23-1255 of SEQ ID NO: 64.

The term“HER2 -positive cell” refers to a cell that expresses HER2 on its surface. The term“monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical and/ or bind the same epitope, except for possible variant antibodies, e.g., containing naturally occurring mutations or arising during production of a monoclonal antibody preparation, such variants generally being present in minor amounts. In contrast to polyclonal antibody preparations, which typically include different antibodies directed against different determinants

(epitopes), each monoclonal antibody of a monoclonal antibody preparation is directed against a single determinant on an antigen. Thus, the modifier“monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the present invention maybe made by a variety of techniques, including but not limited to the hybridoma method, recombinant DNA methods, phage- display methods, and methods utilizing transgenic animals containing all or part of the human immunoglobulin loci, such methods and other exemplary methods for making monoclonal antibodies being described herein.

A“naked antibody” refers to an antibody that is not conjugated to a heterologous moiety (e.g., a cytotoxic moiety) or radiolabel. The naked antibody may be present in a pharmaceutical formulation.

“Native antibodies” refer to naturally occurring immunoglobulin molecules with varying structures. For example, native IgG antibodies are heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light chains and two identical heavy chains that are disulfide-bonded. From N- to C-terminus, each heavy chain has a variable region (VH), also called a variable heavy domain or a heavy chain variable domain, followed by three constant domains (CHI, CH2, and CH3). Similarly, from N- to C-terminus, each light chain has a variable region (VL), also called a variable light domain or a light chain variable domain, followed by a constant light (CL) domain. The light chain of an antibody may be assigned to one of two types, called kappa (K) and lambda (l), based on the amino acid sequence of its constant domain.

“Percent (%) amino acid sequence identity” with respect to a reference polypeptide sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the reference polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative

substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. For purposes herein, however, % amino acid sequence identity values are generated using the sequence comparison computer program ALIGN-2. The ALIGN-2 sequence comparison computer program was authored by Genentech, Inc., and the source code has been filed with user documentation in the U.S. Copyright Office, Washington D.C., 20559, where it is registered under U.S. Copyright Registration No. TXU510087. The ALIGN-2 program is publicly available from Genentech, Inc., South San Francisco, California, or maybe compiled from the source code. The ALIGN-2 program should be compiled for use on a UNIX operating system, including digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary.

In situations where ALIGN-2 is employed for amino acid sequence comparisons, the % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given amino acid sequence A that has or comprises a certain % amino acid sequence identity to, with, or against a given amino acid sequence B) is calculated as follows:

too times the fraction X/Y

where X is the number of amino acid residues scored as identical matches by the sequence alignment program ALIGN-2 in that program's alignment of A and B, and where Y is the total number of amino acid residues in B. It will be appreciated that where the length of amino acid sequence A is not equal to the length of amino acid sequence B, the % amino acid sequence identity of A to B will not equal the % amino acid sequence identity of B to A. Unless specifically stated otherwise, all % amino acid sequence identity values used herein are obtained as described in the immediately preceding paragraph using the ALIGN-2 computer program

The term“PD-i axis binding antagonist” refers to a molecule that inhibits the interaction of a PD-i axis binding partner with either one or more of its binding partner, so as to remove T-cell dysfunction resulting from signaling on the PD-i signaling axis - with a result being to restore or enhance T-cell function (e.g., proliferation, cytokine production, target cell killing). As used herein, a PD-i axis binding antagonist includes a PD-i binding antagonist, a PD-Li binding antagonist and a PD-L2 binding antagonist.

The term "PD-i binding antagonist" refers to a molecule that decreases, blocks, inhibits, abrogates or interferes with signal transduction resulting from the interaction of PD- 1 with one or more of its binding partners, such as PD-Li, PD-L2. In some embodiments, the PD-i binding antagonist is a molecule that inhibits the binding of PD-i to one or more of its binding partners. In a specific aspect, the PD-i binding antagonist inhibits the binding of PD-i to PD-Li and/or PD-L2. For example, PD-i binding antagonists include anti-PD-i antibodies, antigen binding fragments thereof, immunoadhesins, fusion proteins, oligopeptides and other molecules that decrease, block, inhibit, abrogate or interfere with signal transduction resulting from the interaction of PD-i with PD-Li and/or PD-L2. In one embodiment, a PD-i binding antagonist reduces the negative co-stimulatory signal mediated by or through cell surface proteins expressed on T lymphocytes mediated signaling through PD-i so as render a dysfunctional T-cell less dysfunctional (e.g., enhancing effector responses to antigen recognition). In some embodiments, the PD-i binding antagonist is an anti-PD- 1 antibody. In a specific aspect, a PD-i binding antagonist is MDX-1106 (nivolumab) described herein. In another specific aspect, a PD-i binding antagonist is MK- 3475 (lambrolizumab) described herein. In another specific aspect, a PD-i binding antagonist is CT-01 1 (pidilizumab) described herein. In another specific aspect, a PD-i binding antagonist is AMP-224 described herein.

The term“PD-Li binding antagonist” refers to a molecule that decreases, blocks, inhibits, abrogates or interferes with signal transduction resulting from the interaction of PD- Li with either one or more of its binding partners, such as PD-i, B7-1. In some embodiments, a PD-Li binding antagonist is a molecule that inhibits the binding of PD- Li to its binding partners. In a specific aspect, the PD-Li binding antagonist inhibits binding of PD-Li to PD-i and/or B7-1. In some embodiments, the PD-Li binding antagonists include anti-PD-Li antibodies, antigen binding fragments thereof, immunoadhesins, fusion proteins, oligopeptides and other molecules that decrease, block, inhibit, abrogate or interfere with signal transduction resulting from the interaction of PD-Li with one or more of its binding partners, such as PD-i, B7-1. In one embodiment, a PD-Li binding antagonist reduces the negative co-stimulatory signal mediated by or through cell surface proteins expressed on T lymphocytes mediated signalling through PD-Li so as to render a dysfunctional T-cell less dysfunctional (e.g., enhancing effector responses to antigen recognition). In some embodiments, a PD-Li binding antagonist is an anti-PD-Li antibody. In a specific aspect, an anti-PD-Li antibody is YW243.55. S70 described herein. In another specific aspect, an anti- PD-Li antibody is MDX-1105 described herein. In still another specific aspect, an anti-PD- Li antibody is MPDL3280A described herein. In still another specific aspect, an anti-PD-Li antibody is MEDI4736 described herein.

The term“PD-L2 binding antagonist” refers to a molecule that decreases, blocks, inhibits, abrogates or interferes with signal transduction resulting from the interaction of PD- L2 with either one or more of its binding partners, such as PD-i. In some embodiments, a PD-L2 binding antagonist is a molecule that inhibits the binding of PD-L2 to one or more of its binding partners. In a specific aspect, the PD-L2 binding antagonist inhibits binding of PD-L2 to PD-i. In some embodiments, the PD-L2 antagonists include anti-PD-L2 antibodies, antigen binding fragments thereof, immunoadhesins, fusion proteins, oligopeptides and other molecules that decrease, block, inhibit, abrogate or interfere with signal transduction resulting from the interaction of PD-L2 with either one or more of its binding partners, such as PD-i. In one embodiment, a PD-L2 binding antagonist reduces the negative co-stimulatoiy signal mediated by or through cell surface proteins expressed on T lymphocytes mediated signaling through PD-L2 so as render a dysfunctional T-cell less

dysfunctional (e.g., enhancing effector responses to antigen recognition). In some embodiments, a PD-L2 binding antagonist is an immunoadhesin.

A“fixed” or“flat” dose of a therapeutic agent herein refers to a dose that is

administered to a human patient without regard for the weight (WT) or body surface area (BSA) of the patient. The fixed or flat dose is therefore not provided as a mg/kg dose or a mg/ m 2 dose, but rather as an absolute amount of the therapeutic agent. A“loading” dose herein generally comprises an initial dose of a therapeutic agent administered to a patient, and is followed by one or more maintenance dose(s) thereof. Generally, a single loading dose is administered, but multiple loading doses are contemplated herein. Usually, the amount of loading dose(s) administered exceeds the amount of the maintenance dose(s) administered and/or the loading dose(s) are administered more frequently than the maintenance dose(s), so as to achieve the desired steady-state concentration of the therapeutic agent earlier than can be achieved with the maintenance dose(s).

A“maintenance” dose herein refers to one or more doses of a therapeutic agent administered to the patient over a treatment period. Usually, the maintenance doses are administered at spaced treatment intervals, such as approximately every week, approximately every 2 weeks, approximately every 3 weeks, or approximately every 4 weeks, preferably every 3 weeks.

“Infusion” or“infusing” refers to the introduction of a drug-containing solution into the body through a vein for therapeutic purposes. Generally, this is achieved via an intravenous (IV) bag.

An“intravenous bag” or“IV bag” is a bag that can hold a solution which can be administered via the vein of a patient. In one embodiment, the solution is a saline solution (e.g. about 0.9% or about 0.45% NaCl). Optionally, the IV bag is formed from polyolefin or polyvinal chloride.

The term“variable region” or“variable domain” refers to the domain of an antibody heavy or light chain that is involved in binding the antibody to antigen. The variable domains of the heavy chain and light chain (VH and VL, respectively) of a native antibody generally have similar structures, with each domain comprising four conserved framework regions (FRs) and three hypervariable regions (HVRs). (See, e.g., Kindt et al. Kuby Immunology, 6 th ed., W.H. Freeman and Co., page 91 (2007).) A single VH or VL domain may be sufficient to confer antigen-binding specificity.

Furthermore, antibodies that bind a particular antigen may be isolated using a VH or VL domain from an antibody that binds the antigen to screen a library of

complementary VL or VH domains, respectively. See, e.g., Portolano et al, J. Immunol. 150:880-887 (1993); Clarkson et al, Nature 352:624-628 (1991). The term“vector,” as used herein, refers to a nucleic acid molecule capable of propagating another nucleic acid to which it is linked. The term includes the vector as a self- replicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced. Certain vectors are capable of directing the expression of nucleic acids to which they are operatively linked. Such vectors are referred to herein as“expression vectors.”

A“free cysteine amino acid” refers to a cysteine amino acid residue which has been engineered into a parent antibody, has a thiol functional group (-SH), and is not paired as an intramolecular or intermolecular disulfide bridge.

The term“or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof’ means that pharmaceutically acceptable salt, solvate, tautomeric, stereoisomeric forms of the shown structure are also included. Mixtures thereof means that mixture of these forms may be present, for example, the compounds of the invention may include both a tautomeric form and a pharmaceutically acceptable salt.

“Pharmaceutically acceptable” substances refers to those substances which are within the scope of sound medical judgment suitable for use in contact with the tissues of subjects without undue toxicity, irritation, allergic response, and the like,

commensurate with a reasonable benefit-to-risk ratio, and effective for their intended use.

“Pharmaceutical composition” refers to the combination of one or more drug substances and one or more excipients.

As used herein,“solvate” refers to a complex of variable stoichiometry formed by a solute (e.g. formulas (l)-(i) (A), (B), (C), (D), or any other compound herein or a salt thereof) and a solvent. Pharmaceutically acceptable solvates may be formed for crystalline compounds wherein solvent molecules are incorporated into the crystalline lattice during crystallization. The incorporated solvent molecules can be water molecules or non-aqueous molecules, such as but not limited to, ethanol, isopropanol, dimethyl sulfoxide, acetic acid, ethanolamine, and ethyl acetate molecules.

The term“subject” as used herein refers to a human or non-human mammal.

Examples of non-human mammals include livestock animals such as sheep, horses, cows, pigs, goats, rabbits and deer; and companion animals such as cats, dogs, rodents, and horses.

“Therapeutically effective amount” of a drug refers to the quantity of the drug or composition that is effective in treating a subject and thus producing the desired therapeutic, ameliorative, inhibitory or preventative effect. The therapeutically effective amount may depend on the weight and age of the subject and the route of administration, among other things.

“Treating” refers to reversing, alleviating, inhibiting the progress of, or preventing a disorder, disease or condition to which such term applies, or to reversing, alleviating, inhibiting the progress of, or preventing one or more symptoms of such disorder, disease or condition.

“Treatment” refers to the act of“treating”, as defined immediately above.

As used herein the term“comprising” means“including at least in part of’ and is meant to be inclusive or open ended. When interpreting each statement in this specification that includes the term“comprising”, features, elements and/or steps other than that or those prefaced by the term may also be present. Related terms such as“comprise” and “comprises” are to be interpreted in the same manner.

The term“consisting essentially of’ limits the scope of a claim to the specified materials or steps“and those that do not materially affect the basic and novel characteristic(s)” of the claimed invention. When the phrase“consisting essentially of’ appears in a clause of the body of a claim, rather than immediately following the preamble, it limits only the element set forth in that clause.

The term“consisting of’ excludes any element, step, or ingredient not specified in the claim;“consisting of’ defined as“closing the claim to the inclusion of materials other than those recited except for impurities ordinarily associated therewith. When the phrase“consists of’ appears in a clause of the body of a claim, rather than immediately following the preamble, it limits only the element set forth in that clause; other elements are not excluded from the claim as a whole. It should be understood that while various embodiments in the specification are presented using“comprising” language, under various circumstances, a related embodiment is also described using “consisting essentially of’ or“consisting of’ language. Suitable Compounds

Suitably, the compound of formula (I) is a compound of formula (III):

or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof.

Suitably, the compound of formula (I) is a compound of formula (IV):

or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof.

Suitably, the compound of formula (I) is a compound of formula (V):

or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof.

Suitably for compounds of formulas (III), (IV) or (V) f is l. Suitably, the compound of formula (I) is selected from:

;

or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof; wherein the dotted lines from Z to Z 4 represent single or double bonds;

Yg is selected from N-R 28 , S and O;

Y 9 is selected from C-R 2g and N;

one of Y o and Yu is independently selected from N-R 2 s, S and O; and the other of Y i0 andYu is C-R 2g ;

Y 3 is selected from C-R 2g , N and S;

R 27 is selected from H, OH, C - i2 alkyl, 0C - i2 alkyl and R A ;

R 28 is selected from H and C - i2 alkyl; and

each R 2g is selected from H, OH, C - i2 alkyl, 0C - i2 alkyl and R A .

Suitably, the compound of formula (I) is selected from formula (VI), (VII) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof.

Suitably, the compound of formula (I) is selected from:

or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof. Suitably the substituents are as described for formula (VI)-(VIII).

Suitably, the compound of formula (I) is selected from:

or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof. Suitably the substituents are as described for formula (VI)-(VIII).

More suitably, the compound of formula (I) is selected from:

or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof. Suitably the substituents are as described for formula (VI)-(VIII).

In one embodiment, suitably, the compound of formula (I) is:

or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof. Suitably the substituents are as described for formula (VI)-(VIII).

In another embodiment, suitably, the compound of formula (I) is:

(xviii) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof. Suitably the substituents are as described for formula (VI)-(VIII).

In another embodiment, suitably, the compound of formula (I):

or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof. Suitably the substituents are as described for formula (VI)-(VIII).

In another embodiment, suitably, the compound of formula (I) is:

or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof. Suitably the substituents are as described for formula (VI)-(VIII).

In another embodiment, suitably, the compound of formula (I) is:

or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof. Suitably the substituents are as described for formula (VI)-(VIII). a

In some aspects, q is o. In other aspects, q is 1. Dotted lines

In one aspect, suitably the dotted lines from Z t to Z 2 and from Z 3 to Z 4 are double bonds and the remaining dotted line is a single bond. In another aspect, suitably, one of the dotted lines from Z to Z 2 , Z 2 to Z 3 , or Z 3 to Z 4 is a double bond and the remaining dotted lines are single bonds. In another aspect, suitably, all of the dotted lines from Z , Z 2 , Z 3 and Z 4 are single bonds.

Z T Z ?, Z 2 and Z

Suitably, zero, one, two or three of Z , Z 2 , Z 3 and Z 4 are O. In one aspect, suitably, one, two or three of Z , Z 2 , Z 3 and Z 4 are O.

In one aspect, suitably only one of Z , Z 2 , Z 3 and Z 4 is O. Hence, in this aspect, the compound of formula (I) is selected from:

or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof. Suitably, f is l.

More suitably, Z t is selected from C-R and CH-R ; Z 2 is selected from C-R 2 and CH-R 2 ; Z 3 is selected from C-R 3 and CH-R 3 ; and Z 4 is selected C-R z , and CH-R, which may be represented by showing the compound of formula (I) as:

or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof. Suitably, f is 1. More suitably, Z t , Z 2 , Z 3 and Z 4 are selected from CH and CH 2 .

Z ^ and Z

Z 5 and Z 6 together are selected from CR 5 R 6 -NR 7 , CR 5 R 5 ’-CR 6 R 6 ’, CR 5 R 6 -S, CR 5 R 6 -0, CR 7 =CR 5 and NR 7 -C(=0) which can be positioned in either direction, such that the compound of formula (I) is selected from:

,

4i or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof. Suitably, f is 1.

More suitably, Z 5 and Z 6 are CR 5 R 6 -NR 7 and the compound has the structure of formula (XVIII) or (XIX). Most suitably, Z 5 and Z 6 are CR 5 R 6 -NR 7 and the compound has the structure of formula (XVIII).

Suitably, Z y is C=S. More suitably, Z y is C=0.

More suitably, in one aspect Z 5 and Z 6 are CR 5 R 6 -NR 7 ; Z 7 is C=0 and the structure of formula (I) is:

or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof. Suitably, f is l.

More suitably, in one aspect Z 5 and Z 6 are CR 5 R 6 -NR 7 ; Z 7 is C=0, and T) is (iii) and the structure of formula (I) is:

or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof. Suitably, f is 1.

R . R ? . R 2 and R^

R , R 2 , R 3 and R 4 are not always present in the compound of formula (I) , i.e. if any of Z , Z 2 , Z 3 or Z 4 are O. Suitably, at least one of R , R 2 , R 3 and R 4 are present in the compound of formula (I). Suitably, at least two of R , R 2 , R 3 and R 4 are present. More suitably, at least three of R , R 2 , R 3 and R 4 are present. Most suitably, all of R , R 2 , R 3 and R z , are present. In some aspects, suitably, at least one of R , R 2 , R 3 and R4 is H; suitably, at least two of R , R 2 , R 3 and R z , are H; suitably, at least three of R , R 2 , R 3 and I¾ are H; suitably, R = R 2 = R 3 = R 1 = H . In one aspect, suitably, R , R 2 , R 3 and I¾ where present are (a) independently selected from H, OH, C - i2 alkyl, 0C - i2 alkyl, =C(R I4 )(R I5 ), RA and halogen.

In another aspect, suitably, R , R 2 , R 3 and R4 where present are (a) independently selected from H, OH, C - 2 alkyl, 0C - i2 alkyl, =C(R I4 )(R I5 ), RA and halogen; with the proviso that a maximum of one (i.e. o or 1) of R , R 2 , R 3 and R4 is a RA group and/ or with the proviso that a maximum of one (i.e. o or 1) of R , R 2 , R 3 and R4 is a

=C(R I4 )(R I5 ) group.

Suitably, in some aspects, a single =C(R I4 )(R I5 ) group is present such that the non- alkylating moiety is selected from:

wherein the dotted lines between Z 2 to Z 4 represent single or double bonds;

wherein the dotted lines between Z 3 to Z 4 represent a single or double bond;

wherein the dotted lines between Z to Z 2 represent a single or double bond; and

wherein the dotted lines between Z to Z 3 represent single or double bonds. As discussed above, suitably, all of R , R 2 , R 3 and R z , are present, hence, suitably in this aspect (a), the non-alkylating moiety:

comprises the three ABC fused-ring structure as shown but does not comprise a fused D-ring. In one aspect, suitably, (a) R , R 2 , R 3 and R 4 are selected such that one of R , R 2 , R 3 and R 4 is =C(R I4 )(R I5 ); and the remaining R ls R 2 , R 3 and R 4 are independently selected from H, OH, C - 2 alkyl, 0C - i2 alkyl and halogen. Suitably, R 2 is =C(R I4 )(R I5 ) and R , R 3 and R 4 are independently selected from H, OH, C - i2 alkyl, 0C - i2 alkyl and halogen. More suitably, in this aspect, the non-alkylating moiety is:

In another aspect, suitably (a) Ri, R 2 , R 3 and R4 where present are (a) independently selected from H, OH, Ci- i2 alkyl, 0Ci- i2 alkyl, R A and halogen. In another aspect, one of Ri, R 2 , R 3 and I¾ is R A . More suitably, one ofRi, R 2 , R 3 and I¾ is R A and other three are H. In another aspect, more suitably, (a) Ri, R 2 , R 3 and R z , are independently selected from H, OH, Ci- i2 alkyl, 0Ci- i2 alkyl and halogen. More suitably, (a) Ri, R 2 , R 3 and R z , are independently selected from H, Ci- i2 alkyl and 00- 12 alkyl. More suitably, (a) Ri = R 2 = R 3 = R z , = H. Suitably, (b) one of R and R 2 ; or R 2 and R 3 ; or R 3 and I¾ together with the carbon atoms to which they are attached form a 6-membered aryl ring, or a 5- or 6-membered heteroaryl ring, wherein the non-fused carbons of the aryl or heteroaryl ring are substituted with groups RD , RD 2 , RD 3 and RD 4 ; and the remaining R ls R 2 , R 3 and I¾ groups that do not form a ring are independently selected from H, OH, C - i2 alkyl, 0C - i2 alkyl, R A and halogen. Hence, in this option the non-alkylating moiety comprises a fused D-ring.

Suitably, for option (b) one of R and R 2 ; or R 2 and R 3 ; or R 3 and R 4 together with the carbon atoms to which they are attached form a 5- or 6-membered heteroaryl ring, such that the non-alkylating moiety:

wherein t is o or l; and when t is o then one of Zg, Z 9 and Z 9 are selected from NR 2i , S, O and the remaining of Zg, Z 9 and Z 9 are independently selected from N, CH, C-OH, C-(C - alkyl), C-0(C - 2 alkyl) and C-R A ; and when t is l then one of Zg, Z 9 , Z 9 and Z 10 are N and the remaining of Zg, Z 9 , Z 9 and Z 10 are independently selected from N, CH, C-OH, C-(C - alkyl), C-0(C - 2 alkyl) and C-R A .

More suitably, for option (b) one of R and R 2 ; or R 2 and R 3 ; or R 3 and R 4 together with the carbon atoms to which they are attached form a 6-membered aryl ring such that the non-alkylating moiety:

is selected from:

wherein the dotted line between Z and Z 2 is a single or double bond;

wherein the dotted line between Z 3 and Z 4 is a single or double bond.

More suitably, for option (b) the non-alkylating moiety is selected from:

Suitably, for option (c) the non-alkylating moiety is selected from:

Suitably, for option (c) the non-alkylating moiety is (C2).

Suitably, for option (c) the non-alkylating moiety is selected from:

Suitably, for option (c) the non-alkylating moiety is (C6).

More suitably, for option (c) one out of R , R 2 R 3 and R 4 is a C - 2 alkyl, a phenyl ring or a C 5-9 heteroaryl group and these groups are optionally substituted with 1, 2 or 3 optional groups selected from OH, C - i2 alkyl, 0C - i2 alkyl, R A and halogen; and where present the remaining of R , R 2 , R 3 and R 4 are independently selected from H, OH, C - i2 alkyl, 0C - i2 alkyl, R A and halogen. More suitably, for option (c) R is H; R 2 is ci Ci- 12 alkyl, a phenyl ring or a C 5-9 heteroaryl group optionally substituted with 1, 2 or 3 optional groups selected from OH, C - 2 alkyl, 0Ci- i2 alkyl, R A and halogen; R 3 is H.

More suitably for option (c) q is o and the non-alkylating moiety is selected from:

In some aspects, for options (b) or (c) the remaining R ls R 2 , R 3 and R 4 , that do not form a ring or that are not R w , are independently selected from H, OH, Ci- i2 alkyl, 0Ci- i2 alkyl and halogen. More suitably, for options (b) or (c) the remaining R ls R 2 , R 3 and R 4 are independently selected from H, Ci- i2 alkyl and 0Ci- i2 alkyl. More suitably, for options (b) or (c) the remaining R ls R 2 , R 3 and R 4 are H.

In some aspects for options (b) or (c) one the remaining R ls R 2 , R 3 and R 4 , that do not form a ring or that are not R w , is R A and the other remaining groups are independently selected from H, Ci- i2 alkyl and 0Ci- i2 alkyl; more suitably, the other remaining groups are H.

RD , RD ?, RD 2 and RD

In one aspect, suitably one of RD , RD 2 , RD 3 and RD 4 is R A .. Suitably, one RD , RD 2 , RD 3 and RD 4 is R A and other three are independently selected from H, OH, Ci- i2 alkyl, 0Ci- i2 alkyl and halogen. Suitably, one RD , RD 2 , RD 3 and RD 4 is R A and other three are independently selected from H, C - 0 alkyl and OCi- 1 0 alkyl. Suitably, one RD , RD 2 , RD 3 and RD 4 is R A and other three are independently selected from H, methyl, ethyl, n- propyl, i-propyl, O-methyl, O-ethyl, O-(n-propyl) and O-(i-propyl). Suitably, one RD , RD 2 , RD 3 and RD 4 is R A and other three are H.

More suitably, RDi, RD 2 , RD 3 and RD 4 are independently selected from H, OH, Ci-i 0 alkyl, OCi- 1 0 alkyl and halogen. More suitably, Ru, R 12 and R 13 are independently selected from H, OH, Ci-8 alkyl, OC 1 -8 alkyl and halogen. More suitably, Ru, R 12 and R 13 are independently selected from H, OH, Ci -6 alkyl, OCi -6 alkyl and F, Cl, Br and I. More suitably, RD , RD 2 , RD 3 and RD 4 are independently selected from H, OH, methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl, i-butyl, t-butyl, O-methyl, O-ethyl, 0-(n- propyl), O-(i-propyl), O-(n-butyl), O-(s-butyl), O-(i-butyl), O-(t-butyl) and F, Cl, Br and I. More suitably, RD , RD 2 , RD 3 and RD 4 are independently selected from H, methyl, ethyl, n-propyl, i-propyl, O-methyl, O-ethyl, O-(n-propyl) and O-(i-propyl). More suitably, RD , RD 2 , RD 3 and RD 4 are H.

R„ R ft and R v

Suitably, R 5 , R & and R 7 are independently selected from H and C - 0 alkyl; suitably, R 5 ,

R 6 and R 7 are independently selected from H and C - 8 alkyl; more suitably, R 5 , R & and R 7 are independently selected from H and C -6 alkyl.

More suitably, R 5 , R & and R 7 are independently selected from H, methyl, ethyl, n- propyl, i-propyl, n-butyl, s-butyl, i-butyl and t-butyl.

Ea

Suitably Rs is selected from H, C - 0 alkyl and CH 2 Ph. Suitably Rs is selected from H, C - 8 alkyl and CH 2 Ph. Suitably Rs is selected from H, C -6 alkyl and CH 2 Ph.

Suitably Rs is selected from H, methyl, ethyl and CH 2 Ph.

More suitably Rs is selected from methyl and ethyl.

More suitably Rs is methyl.

Ro and Rm

Suitably, R g and R 10 are independently selected from H and C - 0 alkyl,; suitably, R g and R o are independently selected from H and C -s alkyl; more suitably, R g and R 10 are independently selected from H and C -6 alkyl.

More suitably, R g and R 10 are independently selected from H, methyl, ethyl, n-propyl, i- propyl, n-butyl, s-butyl, i-butyl and t-butyl.

In one aspect, R g is R A and R i0 is selected from H and C - i2 alkyl; suitably, R g is R A and R o is H. In a more suitably alternative aspect, R g and R 10 are H.

R , R and R 2

Suitably, Ru, R 12 and R 13 are independently selected from H, OH, C - 0 alkyl, OC - 0 alkyl and R A . Suitably, Ru, R 12 and R 13 are independently selected from H, OH, C -s alkyl, OC -8 alkyl and R A . Suitably, Ru, R 12 and R 13 are independently selected from H, OH, C - 6 alkyl, OC -6 alkyl and R A .

More suitably, Ru, R 12 and R 13 are independently selected from H, OH, methyl, ethyl, n- propyl, i-propyl, n-butyl, s-butyl, i-butyl, t-butyl, O-methyl, O-ethyl, O-(n-propyl), 0-(i- propyl), O-(n-butyl), O-(s-butyl), O-(i-butyl), O-(t-butyl) and RA.

In one aspect, suitably, Ru, R and R TS are independently selected from H, OH, C - 2 alkyl and 0C - 2 alkyl. More suitably, Ru, R 12 and R 13 are independently selected from H, C - 2 alkyl and OC 1-12 alkyl.

In another aspect, more suitably, one of Ru, R 12 and R 13 is RA and the remaining of Ru, R 12 and R I3 are independently selected from H, OH, C 1-12 alkyl and OC 1-12 alkyl. More suitably, one of Ru, R 12 and R TS is RA and the remaining of Ru, R 12 and R TS are independently selected from H, C - 2 alkyl and OC 1-12 alkyl. More suitably, one of Ru,

R 12 and R I3 is RA and the remaining of Ru, R 12 and R 13 are H.

R, and RI

Suitably, each R 34 and R 15 are independently selected from H, C HO alkyl and (CH 2 ) j -R x ; suitably, each R 34 and R 15 are independently selected from H, C - 8 alkyl and (CH 2 ) j -R x ; more suitably, each R 34 and R 15 are independently selected from H, C -6 alkyl and (CH 2 ) j - R x .

More suitably, each R 34 and R 15 are independently selected from H, methyl, ethyl, n- propyl, i-propyl, n-butyl, s-butyl, i-butyl and t-butyl.

R fi and Ru

Suitably, R 16 and R 17 are independently selected from H and C - 0 alkyl,; suitably, R 16 and R I7 are independently selected from H and C - 8 alkyl; more suitably, R 16 and R 17 are independently selected from H and C -6 alkyl.

More suitably, R 16 and R iy are independently selected from H, methyl, ethyl, n-propyl, i- propyl, n-butyl, s-butyl, i-butyl and t-butyl.

R g Suitably, each Rg is independently selected from H and C- 0 alkyl; suitably, each Rg is independently selected from H and C- 8 alkyl; more suitably, each Rg is independently selected from H and C -6 alkyl.

More suitably, each Rg is independently selected from H, methyl, ethyl, n-propyl, i- propyl, n-butyl, s-butyl, i-butyl and t-butyl. More suitably, each Rg is independently selected from H, methyl and ethyl.

In one aspect, an Rg is H. In another aspect, an Rg is a C- 2 alkyl.

Rg

Suitably, each R g is independently selected from H, OH and C- 0 alkyl; suitably, each Rg is independently selected from H, OH and C- 8 alkyl; more suitably, each Rg is independently selected from H, OH and C -6 alkyl.

More suitably, each Rg is independently selected from H, OH, methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl, i-butyl and t-butyl. More suitably, each R g is independently selected from H, methyl and ethyl.

In one aspect, Rg is R A . In a more suitable aspect, Rg is a C- 2 alkyl. In a most suitable aspect, Rg is H.

Rn and R n

Suitably, R 20 and R’ 20 are independently selected from H and C- 0 alkyl; suitably, R 20 and R’ 20 are independently selected from H and C- 8 alkyl; more suitably, R 20 and R’ 20 are independently selected from H and C -6 alkyl.

More suitably, R 20 and R’ 20 are independently selected from H, methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl, i-butyl and t-butyl.

In one aspect, R 20 is R A . In a more suitably alternative aspect, R 20 is H.

In one aspect, R’ 20 is R A . In a more suitably alternative aspect, R’ 20 is H.

R and R ??

Suitably, each R 2I and R 22 are independently selected from K I -R 33 , H and C- 0 alkyl; suitably, each R 2I and R 22 are independently selected from K I -R 33 , H and C- 8 alkyl; more suitably, each R 2I and R 22 are independently selected from K I -R 33 , H and C -6 alkyl. In one aspect, suitably one of R 2i and R 22 is K I -R 33 , and each of the remaining R 2I and R 22 are independently selected from H and C - i2 alkyl.

Suitably, each R 2I and R 22 are independently selected from H and C - 0 alkyl; suitably, each R 21 and R 22 are independently selected from H and C - 8 alkyl; more suitably, each R 21 and R 22 are independently selected from H and C -6 alkyl.

More suitably, each R 2I and R 22 are independently selected from H, methyl, ethyl, n- propyl, i-propyl, n-butyl, s-butyl, i-butyl and t-butyl.

R 22 . R | and R

Suitably, R 23 , R 24 and R 25 are independently selected from H, OH, C - 0 alkyl, OC - 0 alkyl and R A . Suitably, R 23 , R 24 and R 25 are independently selected from H, OH, C - 8 alkyl, OC 1-8 alkyl and R A . More suitably, R 23 , R 24 and R 25 are independently selected from H, OH, C - 6 alkyl, OC -6 alkyl and R A .

More suitably, R 23 , R 24 and R 25 are independently selected from H, OH, methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl, i-butyl, t-butyl, O-methyl, O-ethyl, O-(n-propyl), O- (i-propyl), O-(n-butyl), O-(s-butyl), O-(i-butyl), O-(t-butyl) and R A .

R26

Suitably, R 2& is selected from H and C - 0 alkyl; suitably, R 2& is selected from H and C - 8 alkyl; more suitably, R 2& is selected from H and C - 6 alkyl.

More suitably, R 2& is selected from H, methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl, i-butyl and t-butyl.

Suitably, R 27 is selected from H, OH, C - 0 alkyl, OC - 0 alkyl and R A . Suitably, R 27 is selected from H, OH, C - 8 alkyl, OC 1-8 alkyl and R A . More suitably, R 27 is selected from H, OH, C - 6 alkyl, OC -6 alkyl and R A . In another aspect, R 27 is R A . In an alternative aspect, R 27 is selected from H, OH, C -6 alkyl, OC -6 alkyl

More suitably, R 27 is selected from H, OH, methyl, ethyl, n-propyl, i-propyl, n-butyl, s- butyl, i-butyl, t-butyl, O-methyl, O-ethyl, O-(n-propyl), O-(i-propyl), O-(n-butyl), 0-(s- butyl), O-(i-butyl), O-(t-butyl) and R A . In one aspect, R 27 is selected from OH, C - 2 alkyl, 0C - i2 alkyl and RA. Suitably, R 27 is RA. More suitably, R 27 is selected from OH, C - i2 alkyl and 0C - i2 alkyl.

In another aspect, more suitably, R 27 is selected from H, OH, C - i2 alkyl and 0CV 12 alkyl. More suitably, R 27 is selected from H, C - i2 alkyl and 0C - i2 alkyl. More suitably, R 27 is

H.

R2S

Suitably, R 2 8 is selected from H and C - 0 alkyl; suitably, R 2 8 is selected from H and C -8 alkyl; more suitably, R 2 8 is selected from H and C -6 alkyl.

More suitably, R 2 8 is selected from H, methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl, i-butyl and t-butyl. More suitably, R 2 8 is H, methyl or ethyl. 29

Suitably, R 2g is selected from H, OH, Ci-i 0 alkyl, OCi- 10 alkyl and R A . Suitably, R 2g is selected from H, OH, Ci- 8 alkyl, OC 1-8 alkyl and R A . More suitably, R 2g is selected from H, OH, C -6 alkyl, OC 1-6 alkyl and R A .

More suitably, R 2g is selected from H, OH, methyl, ethyl, n-propyl, i-propyl, n-butyl, s- butyl, i-butyl, t-butyl, O-methyl, O-ethyl, O-(n-propyl), O-(i-propyl), O-(n-butyl), 0-(s- butyl), O-(i-butyl), O-(t-butyl) and RA.

In one aspect, R 2g is selected from OH, 0- 12 alkyl, 0C - i2 alkyl and RA. Suitably, R 2g is RA. More suitably, R 2g is selected from OH, C - i2 alkyl and 0C - i2 alkyl.

In another aspect, more suitably, R 2g is selected from H, OH, C - i2 alkyl and 0CV 12 alkyl. More suitably, R 2g is selected from H, CV 12 alkyl and 0C - i2 alkyl. More suitably, R 2g is

H.

R30

Suitably, R 30 is selected from H, OH, C - 0 alkyl, OC - 10 alkyl and R A . Suitably, R 30 is selected from H, OH, C -8 alkyl, OC 1 -8 alkyl and R A . More suitably, R 30 is selected from H, OH, C -6 alkyl, OC 1 -6 alkyl and R A . More suitably, R 30 is selected from H, OH, methyl, ethyl, n-propyl, i-propyl, n-butyl, s- butyl, i-butyl, t-butyl, O-methyl, O-ethyl, O-(n-propyl), O-(i-propyl), O-(n-butyl), 0-(s- butyl), O-(i-butyl), O-(t-butyl) and RA.

In one aspect, R 30 is selected from OH, C - 2 alkyl, 0C - 2 alkyl and RA. Suitably, R 30 is RA. More suitably, R 30 is selected from OH, C - 2 alkyl and 0C - 2 alkyl.

In another aspect, more suitably, R 30 is selected from H, OH, C - i2 alkyl and 0CV 12 alkyl. More suitably, R 30 is selected from H, C - i2 alkyl and 0C - i2 alkyl. More suitably, R 30 is

H.

Rai

Suitably, R is selected from H, OH, C - 0 alkyl, OC - 1 0 alkyl and RA. Suitably, R is selected from H, OH, C -8 alkyl, OC 1 -8 alkyl and RA. More suitably, R is selected from H, OH, C -6 alkyl, OC -6 alkyl and RA.

More suitably, R 3I is selected from H, OH, methyl, ethyl, n-propyl, i-propyl, n-butyl, s- butyl, i-butyl, t-butyl, O-methyl, O-ethyl, O-(n-propyl), O-(i-propyl), O-(n-butyl), 0-(s- butyl), O-(i-butyl), O-(t-butyl) and RA.

In one aspect, R 31 is selected from OH, 0- 12 alkyl, 0C - i2 alkyl and RA. Suitably, R 31 is RA. More suitably, R 31 is selected from OH, C - i2 alkyl and 0C - i2 alkyl.

In another aspect, more suitably, R 31 is selected from H, OH, C - i2 alkyl and 0CV 12 alkyl. More suitably, R 31 is selected from H, 0,- 12 alkyl and 0C - i2 alkyl. More suitably, R 31 is

H.

Suitably each R 32 is independently selected from H, C - i2 alkyl and phenyl. Suitably, each R 32 is independently selected from H, methyl, ethyl and phenyl. More suitably, each R 32 is independently selected from H, methyl and ethyl.

R33

Suitably each R 33 is a targeting agent or is a reactive moiety capable of reacting with a targeting agent. Where R 33 is a reactive moiety it can react with functional groups such as aldehdes, amines, disulfides, ketones thiols in the targeting agent, or in Staudinger reactions, Pictet-Spengler reactions and/ or Click-type chemistry with the targeting agent. For some reactive moieties suitable coupling reagents are used to react the reactive moiety with a targeting agent, for example, where R 33 is a carboxylic acid

[(CH 2 ) j -C0 2 R 34 ] carbodiimide coupling reagents may be used.

Suitably, each R 33 is independently an azide, alkynes, bisulfone, carbohydrazide, hydroxylamine, iodoacetamide, isothiocyanate, maleimide, phosphine, semihydrazide, succinimidyl ester and sulfonyl halide, C0 2 H, C0 2 CH 3 , C0 2 CH 2 CH 3 , 0-(CH 2 ) k -NH 2 , C(=0)-0-(CH 2 ) k -NH 2 , (CH 2 ) j -NH 2 , NH-CH 3 , S(0) 2 -CH 3 , S(0) 2 -NHCH 3 , S(0) 2 -N(CH 3 ) 2 , C(=NH)-0-CH 3 , C(=NH)-0-CH 2 CH 3 , NH-C(0)-NH 2 , NH-C(0)-NH 2 , H or is a targeting agent.

More suitably, each R 33 is independently an maleimide, C0 2 H, C0 2 CH 3 , C0 2 CH 2 CH 3 , 0-(CH 2 ) k -NH 2 , (CH 2 ) j -NH 2 , NH-CH 3 or is a targeting agent.

In one aspect, suitably, each R 33 is independently an azide, alkynes, bisulfone, carbohydrazide, hydroxylamine, iodoacetamide, isothiocyanate, maleimide, phosphine, semihydrazide, succinimidyl ester and sulfonyl halide, C0 2 H, C0 2 CH 3 , C0 2 CH 2 CH 3 , O- (CH 2 ) k -NH 2 , C(=0)-0-(CH 2 ) k -NH 2 , (CH 2 ) j -NH 2 , NH-CH 3 , S(0) 2 -CH 3 , S(0) 2 -NHCH 3 , S(0) 2 -N(CH 3 ) 2 , C(=NH)-0-CH 3 , C(=NH)-0-CH 2 CH 3 , NH-C(0)-NH 2 or NH-C(0)-NH 2 . More suitably, in some aspects, each R 33 is maleimide:

A number of other chemistries are known for attachment of compounds to antibodies. US 7,595,292 (Brocchini et al.) refers to linkers that form thioesters with the sulfurs in a disulfide bond of an antibody. US 7,985,783 (Carico et al.) refers to the introduction of aldehyde residues into antibodies, which are used to couple compounds to the antibody.

In another aspect, each R 33 is independently a targeting agent wherein each targeting agent is independently a protein, a portion of a protein, a peptide, a nucleic acid, a hormone, an antibody or an antibody fragment. The targeting agent may bind to a tumor-associated antigen, a cancer-stem-cell associated antigen or a viral antigen. Suitably, each targeting agent is independently a protein, a portion of a protein, a polypeptide, a nucleic acid, an antibody or an antibody fragment. More suitably, each targeting agent is independently an antibody or an antibody fragment. More suitably, each targeting agent is an antibody.

In various embodiments, the targeting agent may bind to a target selected from an acute myeloid leukemia (AML M4) cell, an acute promyelocytic leukemia cell, an acute lymphoblastic leukemia cell, an acute lymphocytic leukemia cell, a chronic lymphocytic leukemia cell, a chronic myeloid leukemia cell, a chronic T-cell lymphocytic leukemia, a myelodysplasia syndromic cell, a multiple myeloma cell, a prostate carcinoma cell, a renal cell adenocarcinoma cell, a pancreatic adenocarcinoma cell, a lung carcinoma cell or a gastric adenocarcinoma cell, a gastric adenocarcinoma cell, a breast cancer cell, a colon cancer cell, a melanoma cell, a thyroid cancer cell, an ovarian cancer cell, a bladder cancer cell, a liver cancer cell, a head and neck cancer cell, an esophageal cancer cell, a hodgkin lymphoma cell, a non- hodgkin lymphoma cell, a mesothelioma cell, a neuroblastoma cell, a neuroendocrine tumor cell, a neurofibromatosis type 1 (NFi) cell, a neurofibromatosis type 2 (NF2) or an osteosarcoma cell.

In another aspect, each R 33 is H.

R 34 and R 35

Suitably each R 34 and R 35 is independently selected from H and C -6 alkyl. Suitably, each R 34 and R 35 is independently selected from H, methyl and ethyl.

Ki

Linker K is a bond or is a moiety having 1-200 nonhydrogen atoms selected from C, N, O, S, or halogen, and optionally incorporates alkyl, ether, oxo, carboxyl, carboxamide, carboxamidyl, ester, urethanyl, branched, cyclic, unsaturated, amino acid, heterocyclyl, aryl or heteroaryl moieties. Linker K may be unbranched or branched, flexible or rigid, short or long and may incorporate any combination of moieties as deemed useful. In some embodiments, at least a portion of the linker K may have a polyalkylene oxide polymeric region, which may enhance solubility of the compound of formula (I). In some embodiments, the linker K may have a repeating unit of ethylene glycol, and may have 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49 or 50 ethylene glycol units. In other embodiments, the linker K may include an alkylene chain. Suitably, the alkylene chain comprises -CH 2 - groups in a chain that is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, li or 12 carbons in length. In some embodiments a proportion of the linker K comprises an ethylene glycol repeating unit or an alkylene chain and another proportion of linker K comprises one or more amino acid moieties. In some embodiments, at least a portion of Linker K may include one or more amino acid moieties which may provide enhanced solubility for the compound of formula (I) or may provide amino acid sequences to enhance target binding, enhance compatibility with a targeting agent, or enhance target binding recognition. In other embodiments, the linker K may include one or more amino acid moieties that provide a suitable substrate motif for a protease. When a set of amino acid moieties are incorporated into the linker K that provide a substrate motif specific for a selected protease, the cytotoxic drug compound of formula (I) maybe released from a target bound conjugate to provide localized cytotoxic effects. Such substrate motifs are known in the art and may be incorporated into the linker as desired to provide selective release from the target bound conjugate. This selectivity can be based on known presence of a desired protease within the localized delivery region of the conjugate drug. Other polymeric types of moieties maybe incorporated in the linker K , such as polyacids,

polysaccharides, or polyamines. Other moieties such as substituted aromatic or heteroaromatic moieties may be used to enhance rigidity or provide synthetically accessible sites on substituents therein for linking to reactive moieties or to the compound of formula (I).

The linker K can also include a variety of other connecting groups that connect the ethylene glycol portion to the amino acid sequence, or connect the ethylene glycol or amino acid sequence to R * , or the compound of formula (I). For example, the amino acid sequence can be connected to the compound of formula (I) via a 4- amino benzyl carboxylate group.

More suitably, the linker K is:

(i) -K 2 -X AA -, (ii) -X AA -C(0)-K 2 -, (iii) -X AA -NH-K 2 -, (iv) -NH-X AA -C(0)-K 2 -, (V) -NH-K 2 - C(0)-X AA -, (vi) -C(0)-X AA -NH-K 2 -, (vii) -C(0)-K 2 -NH-X AA -, (viii) -0-CH 2 -p-C 6 H 4 -NH- X AA -C(0)-K 2 -, (ix) -C(0)-0-CH 2 -P-C 6 H 4 -NH-X AA -C(0)-K 2 -, (X) -0-CH 2 -p-C 6 H 4 -NH-K 2 - C(0)-XAA-, (xi) -C(0)-0-CH 2 -P-C 6 H 4 -NH-K 2 -C(0)-XAA-, (xii) -0-CH 2 -P-C 6 H 4 -NH-XAA- C(0)-K 2 -NH-, (xiii) -C(0)-0-CH 2 -P-C 6 H 4 -NH-X AA -C(0)-K 2 -NH-, (xiv) -0-CH 2 -p-C 6 H 4 - NH-K 2 -C(0)-X AA -NH-, (XV) -C(0)-0-CH 2 -P-C 6 H 4 -NH-K 2 -C(0)-X AA -NH-, (xvi) -X AA -, (xvii) -C(0)-X AA -, (xviii) -NH-X AA - or (xix) -C(0)-X AA -NH-; wherein X AA is an amino acid sequence; and K 2 is -[CH 2 CH 2 0] 0-5o - or -[CH 2 ] 0 Suitably, K 2 is -[CH 2 CH 2 0] o5o - comprising o, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49 or 50 ethylene glycol units. More suitably, K 2 is -[CH 2 ] 0 12 - comprising o, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 carbons.

The linker K may be attached to R 33 and the rest of the compound of formula (I) in either direction. Suitably, XAA is closest to R 33 and K 2 is closest to the rest of the molecule. More suitably, K 2 is closest to R 33 and XAA is closest to the rest of the molecule.

More suitably, the linker K is (i), (ii), (iii), (iv), (vi), (viii), (ix), (x), (xi) or (xvii).

In some embodiments, the linker K can include 8 ethylene glycol units. Several commercially available ethylene glycol groups (polyethylene glycol, PEG) are suitable in the linker K , such as H 2 N-dPEG® 8 -C(0)OH, having a discrete (“d”) polyethylene glycol having 8 ethylene glycol repeating units. Other discrete PEG units are commercially available and known to one of skill in the art, such as by Advanced ChemTech. Suitably, the linker K comprises the formula: -HN-PEG 8 -C(0)-Val-Ala- wherein PEGs has 8 ethylene glycol units. Suitably, the linker K comprises the formula:

Suitably, for the above embodiment, the HN group is directly linked to R 33 . The amino acid portion of the linker K can include any suitable number of amino acid moieties, as described above. For example, the amino acid sequence XAA can include from l to too amino acid moieties, or from l to to amino acid moieties, or from l to 5 amino acid moieties. Suitably, the linker K comprises an amino acid sequence XAA that has 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more amino acid moieties. More suitably, the linker K comprises an amino acid sequence XAA that consists of 2 amino acid moieties.

More suitably, the linker K comprises an amino acid sequence XAA that includes the amino acid sequence Val-Ala. More suitably, the amino acid sequence XAA is:

X

Suitably ¾ is selected from O, S, NH, CH 2 , CH 2 0, C(=0), C(=0)NR l6 , NR l6 C(=0), O- C(O) and C(0)-0;

Suitably, X is selected from O, C(=0), C(=0)NR I6 and NR I6 C(=0).

More suitably X is selected from O, C(=0)NH and NHC(=0).

More suitably X is O.

L

Suitably, L is selected from a peptide chain having from 2 to 5 amino acids, from 2 to 4 amino acids, from 2 to 3 amino acids; a paraformaldehyde chain -(0CH 2 ) I-I2 -, - (0CH 2 ) I-U -, -(OOH 2 ) 1-10 -, -(OCH.k-g-, -(OOH 2 ) 1-8 -, -(OCH 2 ) 1-7 -, -(OOH 2 ) 1-6 -, -(0CH 2 ) !- 5 -, -(OCH 2 ) I-4 -, -(OCH 2 ) I-3 -; a polyethylene glycol chain -(OCH 2 CH 2 ) I-5 -, -(0CH 2 CH 2 ) _ 4 -, -(0CH 2 CH 2 ) I-3 -; and -(CH. H-CCH n -.

More suitably, L is -(CH 2 ) m -L -(CH 2 ) n -, wherein L is selected from -(CH 2 ) I-5 -, -C(O)- NH-, -NH-, -S(0) 0-2 -, -CH[(CH 2 ) 0-5 R A ]-, -Ar ! -CCOi-NH-CArOo- -Arg-, -Ar 3 -(Ar 2 )o- -NH- C(0)-Ar - and -Ar 4 -. More suitably, L is -(CH 2 ) m -L -(CH 2 ) n -, wherein L is selected from -(CH 2 ) I-5 - and -Ar 4 -.

More suitably, L is -(CH 2 ) m -L -(CH 2 ) n -, wherein L is selected from -(CH 2 ) I-5 -,

Y 6 .is C-H or N;

Y 7 is N-R 26 , O or S; and

R 2 3, R 2 4 and R 25 are independently selected from H, OH, CV 12 alkyl, 0C - i2 alkyl and R A ; and

R 26 is H or C 1-12 alkyl. In one aspect, suitably Y 6 is C-H. In another aspect, suitably Y 6 is N. Y z

Suitably, Y 7 is NH, N-CH 3 , N-CH 2 CH3, O or S; and

ATI

Suitably, Ar s selected from pyrrolylene, N-methylpyrrolylene, furanylene, thiophenylene, imidazolylene, N-methylimidazolylene, oxazolylene or thiazolylene, wherein these groups may be optionally substituted with l, 2 or 3 optional substituents independently selected from OH, C - 2 alkyl, 0C - i2 alkyl and RA.

Ar 4 is selected from an optionally substituted 3- to 8-membered cycloalkylene, an optionally substituted 3- to 8-membered heterocycloalkene, an optionally substituted 6-membered arylene and an optionally substituted 5- to 9-membered heteroarylene; wherein the optionally substituted Ar , Ar 2 , Ar 3 and Ar 4 are optionally substituted with 1, 2 or 3 optional substituents independently selected from OH, C - i2 alkyl, 0C - i2 alkyl and RA.

Suitably, Ar 4 is:

wherein one of Y i3 and Y i4 is independently selected from N-H, N-(C I-6 alkyl), S and O; and the other of Y i3 and Y i4 is CH; and Y i5 is independently selected from C-H, C-(C - 6 alkyl), C-R A , N, S and COH.

Suitably, An is selected from pyrrolylene, N-methylpyrrolylene, imidazolylene or N- methylimidazolylene, wherein these groups may be optionally substituted with 1 or 2 optional substituents independently selected OH, C - i2 alkyl, 0C - i2 alkyl and RA.

An,

Suitably, Ar 2 is an optionally substituted phenylene or pyridylene.

More suitably, Ar 2 is:

6o

wherein Y 17 is N or CH; Y s is N or CH; and wherein at least one of Y i7 and Y 8 is CH; and R 3O is H, OH, C - 2 alkyl, 0C - 2 alkyl and RA.

More suitably, Ar 2 is an optionally substituted phenylene.

Ar 3

Suitably, Ar 3 is selected from pyrrolylene, N-methylpyrrolylene, furanylene, thiophenylene, imidazolylene, N-methylimidazolylene, oxazolylene, thiazolylene, pyridylene, indolylene, N-methylindolylene, benzofuranylene, benzothiophenylene, benzimidazolylene, N-methylbenzoimidazolylene, benzooxazolylene or

benzothiazolylene wherein these groups may be optionally substituted with l, 2 or 3 independently selected optional substituents selected from OH, C - i2 alkyl, 0C - i2 alkyl and RA.

Suitably, Ar 3 is selected from pyrrolylene, N-methylpyrrolylene, thiophenylene, imidazolylene, N-methylimidazolylene, oxazolylene, thiazolylene, indolylene, N- methylindolylene, benzofuranylene, benzothiophenylene, benzimidazolylene, N- methylbenzoimidazolylene, wherein these groups may be optionally substituted with 1, 2 or 3 independently selected optional substituents selected from OH, C - i2 alkyl, 00- 12 alkyl and RA.

Suitably, Ar 3 is selected from pyrrolyl, N-methylpyrrolyl, thiophenyl, N-methyl- imidazolyl, oxazolyl, thiazolyl, benzothiophenyl, N-methylbenzoimidazolyl and benzothiazolyl wherein these groups may be optionally substituted with 1, 2 or 3 independently selected optional substituents selected from OH, C - i2 alkyl, 0C - i2 alkyl and RA.

Suitably, Ar 3 is

wherein Y 9 is selected from NH, N-CH 3 , S and O;

Y 20 is selected from CH and N;

Y 2i is selected from NH, N-CH 3 , S and O;

6i Y is selected from CH and N; and

R 3I is selected from H, OH, C - 2 alkyl, OC 1-12 alkyl and R A .

Ar 4

Ar 4 is selected from an optionally substituted 3- to 8-membered cycloalkylene, an optionally substituted 3- to 8-membered heterocycloalkene, an optionally substituted 6-membered arylene and an optionally substituted 5- to 9-membered heteroarylene; wherein these groups are optionally substituted with 1, 2 or 3 optional substituents independently selected from OH, C - 2 alkyl, OC 1-12 alkyl and R A .

More suitably, Ar 4 is selected from an optionally substituted 6-membered arylene and an optionally substituted 5- to 9-membered heteroarylene.

More suitably, Ar 4 is selected from an optionally substituted phenylene and an optionally substituted pyridinylene.

X 2

Suitably X 2 is selected from O, S, NH, CH ¾ CH 2 0, C(=0), C(=0)NR l6 , NR l6 C(=0), O- C(O) and C(0)-0 or is absent.

Suitably X 2 is selected from O, C(=0), C(=0)NR I6 and NR I6 C(=0) or is absent.

More suitably X 2 is selected from O, C(=0)NH and NHC(=0). Suitably X 2 is the same as X .

More suitably X 2 is O. r

Suitably, r is 1 or 2. More suitably, r is 1. i-membered ring containing Y , Y andY ^

Each 5-membered ring containing Y l5 Y 2 and Y 3 :

is a heteroaryl ring. There can be more than one 5-membered ring containing Y l5 Y 2 and Y 3 when r is more than 1. Where there is more than one 5-membered ring containing Y l5 Y 2 and Y 3 , the substituents for each ring are independently selected such that each 5-membered ring may be the same or different. Suitably, the 5-membered ring group containing Y l5 Y 2 and Y 3 is selected from:

wherein each ring is the same or different. For example, the 5-membered ring group containing Y l5 Y 2 and Y 3 may be:

wherein each R g is independently selected from H and C - i2 alkyl. Each heteroaryl ring containing Y l5 Y 2 and Y 3 is selected from the following groups:

Suitably, one of each Y and Y 2 is independently selected from NH, N-(C I-6 alkyl), S and O; and the other of each Y and Y 2 is CH. More suitably, one of each Y and Y 2 is independently selected from NH, N-CH 3 , N-CH 2 CH 3 , S and O; and the other of each Y and Y 2 is CH. Hence, Y , Y 2 and Y 3 for each ring are independently selected from the Y , Y 2 and Y 3 of any other ring.

More suitably, one of each Y and Y 2 is independently selected from NH, N-CH 3 , N- CH 2 CH 3 ; and the other of each Y and Y 2 is CH. Suitably, each Y 3 is independently selected from C-H, C-(C - 6 alkyl), C-OH, N and S. More suitably, each Y 3 is independently selected from C-H, C-CH 3 , C-CH 2 CH 3 , C-OH, N and S. In one aspect, a Y 3 is C-OH, so a 5-membered ring containing Y l5 Y 2 and Y 3 is selected from:

In a further aspect, more suitably, each Y 3 is independently selected from C-H, C-CH 3 ,

C-CH 2 CH 3 , N and S.

More suitably, each 5-membered ring containing Y l5 Y 2 and Y 3 is selected from:

6-membered ring containing Y and Y*

Suitably, the 6-membered ring containing Y 4 and Y 5 is:

Suitably, the 6-membered ring containing Y 4 and Y 5 is selected from:

More suitably, the 6-membered ring containing Y 4 and Y 5 is:

In one aspect, the 6-membered ring containing Y 4 and Y 5 is selected from:

More suitably, the 6-membered ring containing Y 4 and Y 5 is selected from:

Most suitably, the 6-membered ring containing Y 4 and Y 5 is selected from:

Most suitably, the 6-membered ring containing Y 4 and Y 5 is phenylene.

Suitably, Y 4 is selected from N, CH, C-(C - 6 alkyl). More suitably, Y 4 is selected from N, CH, C-CH 3 , C-CH 2 CH 3 . More suitably, Y 4 is N or CH. More suitably, Y 4 is N;

alternatively, and most suitably, Y 4 is CH.

Suitably, Y 5 is selected from N, CH, C-(C - 6 alkyl). More suitably, Y 5 is selected from N, CH, C-CH 3 , C-CH 2 CH 3 . More suitably, Y 5 is N or CH. More suitably, Y 5 is N;

alternatively, and most suitably, Y 5 is CH.

Hi

In one aspect, suitably H is a C 9 heteroaryl group optionally substituted with 1, 2 or 3 optional substituent groups independently selected from OH, C - 2 alkyl, OC 1-12 alkyl and R A . Suitably, H is a C 9 heteroaryl group selected from a 9-membered ring containing Yg and Y 9 .

In another aspect, suitably H is a C 5 heteroaryl group optionally substituted with 1 or 2 optional substituent groups independently selected from OH, C 1-12 alkyl, OC 1-12 alkyl and R A . Suitably, H is a C 5 heteroaryl group selected from a 5-membered ring containing Y 10 , Y u and Y i2 .

In one aspect, H is a C 5 heteroaryl or C 9 heteroaryl group and is substituted with 1, 2 or 3 of the optional groups. Suitably, the H group is substituted with 1 or 2 of the optional groups; more suitably, the H group is substituted with 1 of the optional groups. In one aspect, one of the optional groups is R A and the remaining optional groups are independently selected from OH, C - 2 alkyl and OC 1-12 alkyl. More suitably, the optional groups for H are independently selected from OH, C - 2 alkyl and OC 1-12 alkyl. More suitably, the optional groups for H are independently selected from C 1-12 alkyl and OC 1-12 alkyl. More suitably, in an alternative aspect H is an unsubstituted C 5 heteroaryl group or an unsubstituted C 9 heteroaryl group.

Q-membered ring containing Yg and Y

Suitably, the 9-membered ring containing Yg and Y 9 is selected from:

Suitably, the 9-membered ring containing Yg and Y 9 is selected from:

In some aspects, more suitably, the 9-membered ring containing Yg and Y 9 is:

In other aspects, more suitably, the 9-membered ring containing Yg and Y 9 is:

Most suitably, R 27 is H and the 9-membered ring containing Yg and Y 9 is

Suitably, the 9-membered ring containing Yg and Y 9 is (Y-2); Most suitably, the 9- membered ring containing Yg and Y 9 is (Y-i).

Yg is selected from N-R 2g , S and O. Suitably, Yg is selected from N-H, N-(C I-6 alkyl), S and O. More suitably, Yg is N-H, N-CH 3 , N-CH 2 CH 3 , S or O. Y 9 is selected from C-R 29 and N. Suitably, Ys is selected from C-H, C-(Cu 6 alkyl), N and R A . More suitably, Y 9 is C-H, C-CH 3 , C-CH 2 CH 3 or N. i-membered ring containing Ym, Yu andY

The 5-membered ring containing Y 10 , Y u and Y i2 :

is a heteroaryl ring. The heteroaryl ring containing Y 10 , Yu andY i2 is selected from one of the following groups:

Suitably, one ofY 0 and Yu is independently selected from NH, N-(C I-6 alkyl), S and O; and the other of Y i0 and Yu is CH. More suitably, one of Y i0 and Yu is independently selected from NH, N-CH 3 , N-CH 2 CH 3 , S and O; and the other of Y i0 and Y is CH.

More suitably, one of Y i0 and Yu is independently selected from NH, N-CH 3 , N- CH 2 CH 3 ; and the other of Y i0 and Yu is CH. Suitably, Y i2 is selected from C-R 29 , N and

S.

In one aspect, Y i2 is C-OH, so the 5-membered ring containing Y 10 , Yu and Y i2 is selected from:

In a further aspect, more suitably, Y i2 is selected from C-H, C-CH 3 , C-CH 2 CH 3 , N and S. More suitably, the 5-membered ring containing Y 10 , Yu and Y 12 is selected from:

Ti

Suitably, T is (i), (ii) or (iii). Suitably, T is (i), (ii) or (iv). Suitably, T is (i), (iii) or (iv).More suitably, T is (ii), (iii) or (iv).

Suitably, T is (i) or (ii). Suitably, T is (i) or (iii). Suitably, T is (i) or (iv). More suitably, T is (ii) or (iii). More suitably, T is (ii) or (iv). More suitably, T is (iii) or (iv). Suitably, T is (i). Suitably, T is (ii). Suitably, Suitably, where T is (i) or (ii) it is substituted with 1, 2 or 3 of the optional groups.

Suitably, where T is (i) or (ii) it is substituted with 1 or 2 of the optional groups; more suitably, where T is (i) or (ii) it is substituted with 1 of the optional groups.

In one aspect T is unsubstituted and contains no optional groups. Hence, T is (i) an unsubstituted C - i2 alkyl, or (ii) an unsubstituted C 5 heteroaryl group, (iii)

where Ru, R 12 and R 13 are H; or (iv) OH, 0C - i2 alkyl or R A .

Suitably, in another aspect, the optional groups for T are independently selected from OH, C - 2 alkyl and 0C - i2 alkyl. Suitably, the optional groups for T are independently selected from C - i2 alkyl and 0C - i2 alkyl.

In a more suitable aspect, one of the optional groups is R A and the remaining optional groups are independently selected from OH, C - i2 alkyl and 0C - i2 alkyl. More suitably, one of the optional groups is R A and the remaining optional groups are independently selected from C - i2 alkyl and 0C - i2 alkyl. More suitably, T is substituted with one R A group. Where T is (ii) an optionally substituted C 5-g heteroaryl, suitably it is selected from triazolyl, tetrazolyl, oxadiazolyl, pyridyl, furyl, benzofuranyl, thiophenyl,

benzothiophenyl, pyrrolyl, indolyl, oxazolyl, benzoxazolyl, imidazolyl, benzimidazolyl, thiazolylene, benzothiazolyl, isoxazolyl, pyrazolyl, isothiazolyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl and pyrimidyl, all of which may be optionally substituted. More suitably, where T is (ii) an optionally substituted C 5-g heteroaryl it is an optionally substituted C 5 heteroaryl or an optionally substituted C g heteroaryl. More suitably, T is (ii) an optionally substituted C 5-g heteroaryl selected from furyl, benzofuranyl, thiophenyl, benzothiophenyl, pyridyl, pyrrolyl, indolyl, oxazolyl, benzoxazolyl, imidazolyl, benzimidazolyl, thiazolylene, benzothiazolyl, isoxazolyl, pyrazolyl, all of which may be optionally substituted.

More suitably, T is (i) a C - 2 alkyl optionally substituted with l, 2 or 3 substituent groups selected from OH, OC 1-12 alkyl and R A ; or

(iii)

More suitably, T is (iii)

More suitably, T is (iii) and is selected from:

More suitably, T is (iv) OH, OC 1 - 12 alkyl or RA. More suitably, T is (iv) OH, OC 1 -6 alkyl or RA. More suitably, T is (iv) OH, OCH 3 , OCH 2 CH 3 or RA. More suitably, T is (iv) RA.

EA

Suitably, each R A is independently selected from (CH 2 ) j -C0 2 R 2i , 0-(CH 2 ) k -NR 2i R 22 , C(=0)-0-(CH 2 ) k -NR 2i R 22 , C(=0)-NR 21 R 22 , (CH 2 ) j -NR 21 R 22 , NR 21 NH 2 , C(=0)-NH- (CH 2 ) j -NR 2i R 22 , C(=0)-NH-(CH 2 ) k -C(=NH)NR 21 R 22 , (CH 2 ) j -S(0) 2 -NR 21 R 22 , C(=NH)-0- (C 1 -6 alkyl) and NH-C(0)-NR 21 R 22 .

Suitably, each R A is independently selected from (CH 2 ) j -C0 2 H, (CH 2 ) j -C0 2 CH 3 , (CH 2 ) j - C0 2 CH 2 CH 3 , 0-(CH 2 ) k -NH 2 , 0-(CH 2 ) k -NH-CH 3 , C(=0)-0-(CH 2 ) k -NH 2 , C(=0)-0-

More suitably, each R A is independently selected from C0 2 H, C0 2 CH 3 , C0 2 CH 2 CH 3 , O- (CH 2 ) k -NH 2 , C(=0)-0-(CH 2 ) k -NH 2 , (CH 2 ) j -NH 2 , NH-CH 3 , S(0) 2 -CH 3 , S(0) 2 -NHCH 3 , S(0) 2 -N(CH 3 ) 2 , C(=NH)-0-CH 3 , C(=NH)-0-CH 2 CH 3 , NH-C(0)-NH 2 and NH-C(O)- NH 2 .

More suitably, each R A is independently selected from C0 2 H, C0 2 CH 3 , C0 2 CH 2 CH 3 , O- (CH 2 ) k -NH 2 , (CH 2 ) j -NH 2 and NH-CH 3 .

More suitably, each R A group is selected from 0-(CH 2 ) k -NH 2 and (CH 2 ) j -NH 2 .

In some aspects, suitably, each R A is independently selected from (CH 2 ) j -C0 2 R 2i , O- (CH 2 ) k -NR 2i R 22 , C(0)-0-(CH 2 ) k -NR 21 R 22 , C(0)-NR 21 R 22 , (CH 2 ) j -NR 21 R 22 , NH-C(0)-R 21 , K I -R 33 and (CH 2 ) j -S0 2 -NR 2i R 22 .

In some aspects, suitably, each R A is independently selected from C0 2 H, C0 2 CH 3 , C0 2 CH 2 CH 3 , C0 2 K 1 -R 33 , 0-(CH 2 ) k -NH-K -R 33 , 0-(CH 2 ) k -NH 2 , C(0)-0-(CH 2 ) k -NH-K 1 - R 33 , C(0)-0-(CH 2 ) k -NH 2 , C(0)-NH-K I -R 33 , C(0)-NH 2 , NH-K I -R 33 , NH 2 , NH-C(0)-CH 3 , NH-C(0)-K I -R33, K ! -R 33 , S0 2 -NH-K 1 -R 33 and S0 2 -NH 2 . In some aspects, suitably, one R A is selected from C0 2 K -R 33 , 0-(CH 2 ) k -NH-K -R 33 , C(0)-0-(CH 2 ) k -NH-K -R 33 , CCOi-NH-K -R^NH-K -Raa, NH-CCOi-K -R^, K I -R 33 and S0 2 -NH-K 1 -R 33 . Suitably, the compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof contains a total of o, l, 2 or 3 R A groups. Suitably, the compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof contains a total of o or 1 R A groups. In some aspects, the compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof contains zero R A groups (i.e. R A groups are absent). More suitably, the compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof contains a total of 1 R A group (i.e. a single R A group is present). R w

In one aspect, R w is Rx, =0, CN, NCO, (CH 2 ) j -0Rx, 0-(CH 2 ) k -0Rx, (CH 2 ) j -C0 2 Rx, (CH 2 ) j -NR 21 R x , 0-(CH 2 ) k -NR 21 Rx, C(0)-NR 21 R x , C(0)-0-(CH 2 ) k -NR 21 R x ,

C(0)-NH-(CH 2 ) j -NR 21 R x , C(0)-NH-C 6 H 4 -(CH 2 ) j -Rx, C(0)-NH-(CH 2 ) k -C(=NH)NR 21 R x , C(0)-NH-(CH 2 ) j -R x , NH-C(0)-(CH 2 ) j -Rx, 0-(CH 2 ) k -NH-C(0)-R x ,

0-(CH 2 ) k -C(0)-NH-Rx, (CH 2 ) j -S0 2 R x , 0-S0 2 R x , (CH 2 ) j -S0 2 -NR 21 R x , (CH 2 ) j -C(0)R x ,

(CH 2 ) j -C(0)NR 21 R x , NR 21 NH 2 , C(=NH)-0-R X or NH-C(0)-NR 21 R x .

Suitably, R w is selected from R x , =0, CN, NCO, (CH 2 ) j -0Rx, (CH 2 ) j -C0 2 Rx,

(CH 2 ) j -NR 21 R x , C(0)-NR 21 R x , C(0)-0-(CH 2 ) k -NR 21 R x , C(0)-NH-(CH 2 ) j -NR 21 R x , C(0)-NH-C 6 H 4 -(CH 2 ) j -Rx, C(0)-NH-(CH 2 ) k -C(=NH)NR 21 R x , C(0)-NH-(CH 2 ) j -R x ,

NH-C(0)-(CH 2 ) j -R x , (CH 2 ) j -S0 2 R x , 0-S0 2 R x , (CH 2 ) j -S0 2 -NR 21 R x , (CH 2 ) j -C(0)R x , (CH 2 ) j -C(0)NR 21 R x , NR 21 NH 2 , C(=NH)-0-R X and NH-C(0)-NR 21 R x .

In one aspect, more suitably, R w is selected from R x , (CH 2 ) j -0Rx, (CH 2 ) j -C0 2 Rx, C(0)-NH-C 6 H 4 -(CH 2 ) j -R x , C(0)-NH-(CH 2 ) j -R x , NH-C(0)-(CH 2 ) j -R x and (CH 2 ) j -C(0)R x .

In another aspect, more suitably, R w is selected (CH 2 ) j -NR 2i R x , C(0)-NR 2i R x ,

C(0)-0-(CH 2 ) k -NR 21 Rx, C(0)-NH-(CH 2 ) j -NR 21 R x , (CH 2 ) j -S0 2 -NR 21 R x , (CH 2 ) j - C(0)NR 21 R x , NR 21 NH 2 and NH-C(0)-NR 21 R x . Rx Suitably, each R x is independently selected from H, C -6 alkyl, C 6 12 aryl, C 7-I8 aralkyl, C 5- 10 heteroaryl, C 6-i6 heteroarylalkyl, C 3-i2 heterocyclyl; wherein the alkyl, aralkyl, heteroaryl, heteroarylalkyl and heterocyclyl groups are optionally substituted. More suitably, each R x is independently selected from H, C -6 alkyl, phenyl, C 7-i2 aralkyl groups, C 5-9 heteroaryl, C 6-I5 heteroarylalkyl, C 3-i2 heterocyclyl; wherein the alkyl, aralkyl, heteroaryl, heteroarylalkyl and heterocyclyl groups are optionally substituted.

More suitably, each R x is independently selected from H, C -6 alkyl, C 3-i2 heterocyclyl, N-methylpyrrolyl, furanyl, thiophenyl, imidazolyl, N-methylimidazolyl, oxazolyl, thiazolyl, pyridyl, pyrimidinyl, uracilyl, tetrahydropyridinyl, indolyl, N-methylindolyl, benzofuranyl, benzothiophenyl, benzimidazolyl, N-methylbenzo-imidazolyl, benzooxazolyl, benzothiazolyl, pyrrol-3-ylmethyl, pyrrol-4-ylmethyl, imidazol-2- ylmethyl, imidazol-4-ylmethyl, thiophen-3-ylmethyl, furan-3-ylmethyl, phenyl, benzyl and phenethyl; wherein each of these groups may be optionally substituted.

More suitably, each R x is independently selected from H, C -6 alkyl, phenyl and

(CH 2 ) -6 -phenyl; wherein the alkyl, phenyl and (CH 2 ) - 6 -phenyl groups are optionally substituted.

Suitably, each R x group is optionally substituted with 1, 2 or 3 optional groups independently selected from OH, C - i2 alkyl, 0C - i2 alkyl, R A , C(=0)-NH-C 6 H 4 -(CH 2 ) j - R 2I , C 5-6 heterocyclyl, -S(0) 2 -(C I-6 alkyl), 0-(CH 2 )k-0-(C - 6 alkyl), (CH 2 )k-0-(C - 6 alkyl), CN, NCO, C(0)-NH-(CH 2 ) j -Cy, C(0)-Cy, C 2-7 alkenyl, C 2-7 alkynyl, C 5-20 aryl, C 3-i0 cycloalkenyl, C 3-i0 cycloalkynyl, C 3-20 heterocyclyl, C 3-20 heteroaryl, acetal, acyl, acylamido, acyloxy, amidino, amido, amino, aminocarbonyloxy, azido, carboxy, cyano, ether, formyl, guanidino, halo, hemiacetal, hemiketal, hydroxamic acid, imidic acid, imino, ketal, nitro, nitroso, oxo, oxycarbonyl, oxycarboyloxy, sulfamino, sulfamyl, sulfate, sulfhydryl, sulfinamino, sulfinate, sulfino, sulfinyl, sulfinyloxy, sulfo, sulfonamido, sulfonamino, sulfonate, sulfonyl, sulfonyloxy, uredio and

groups,

wherein Cy is independently selected from a C 5-6 heterocyclyl or C 5-6 heteroaryl group, wherein the heterocyclyl or heteroaryl groups are optionally substituted with an R A group. In some aspects, suitably, each R x group is substituted with 1, 2 or 3 of the optional groups. More suitably, each R x group is substituted with 1 of the optional groups.

More suitably, each R x group is optionally substituted with 1, 2 or 3 optional groups independently selected from OH, Ci-i 2 alkyl, OC alkyl, R A , C(=0)-NH-C 6 H 4 -(CH 2 )j- R , C 5-6 heterocyclyl, -S(0) 2 -(C I-6 alkyl), 0-(CH 2 )k-0-(Ci- 6 alkyl), (CH 2 )k-0-(Ci- 6 alkyl), CN, NCO, C(0)-NH-(CH 2 ) j -Cy, C(0)-Cy, C 2-7 alkenyl, C 2-7 alkynyl, C 5-20 aryl, C 3-i0 cycloalkenyl, C 3-i0 cycloalkynyl, C 3-20 heterocyclyl, C 3-20 heteroaryl, -CHC(0R 32 )(0R X2 ),

g p ,

wherein each R 2I is independently selected from H and C - i2 alkyl; and each R 32 is independently selected from H, C - i2 alkyl and phenyl; and Cy is independently selected from a C 5-6 heterocyclyl or C 5-6 heteroaryl group, wherein the heterocyclyl or heteroaryl groups are optionally substituted with an R A group.

More suitably, each R x group is optionally substituted with 1, 2 or 3 optional groups independently selected from OH, C - i2 alkyl, 0C - i2 alkyl, R A , and halo. More suitably, each R x group is optionally substituted with 1, 2 or 3 optional groups independently selected from OH, C - i2 alkyl, 0C - i2 alkyl and halo; more suitably from C - i2 alkyl and OC - 12 alkyl.

In some aspects, suitably, each R x is independently selected from:

wherein X’ is N, CH or CR’”;

X” is O, NH, N-(C I-6 alkyl) or S; and each R” and R’” are independently selected from H, OH, C - 2 alkyl, OC 1-12 alkyl, R A , halo, S(0) 2 -(C 1-6 alkyl), 0-(CH 2 ) k -0-(C 1-6 alkyl), (CH 2 )j-NR 26 R 27 , NR 26 NH 2 ,

alkyl), CN, NCO, Cy,

wherein Cy is independently selected from a C 5-6 heterocyclyl or C 5-6 heteroaryl group, wherein the heterocyclyl or heteroaryl groups are optionally substituted with an R A group.

In some embodiments, R x is selected from:

In some aspects, suitably, f is o.

More suitably, in other aspects, f is 1.

I

In one aspect, suitably, a j is selected from 1, 2, 3, 4, 5 or 6.

Suitably, each j is independently selected from 0, 1, 2 or 3.

More suitably, in some aspects, j is 1.

More suitably, in other aspects, j is o. k

Suitably, each k is independently selected from l, 2 or 3.

More suitably, k is 1 or 2.

More suitably, in some aspects, k is 1. m

In one aspect, suitably, m is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12.

Suitably m is o, 1, 2, 3, 4, 5 or 6. Suitably, m is o, 1, 2 or 3. Suitably, m is 1, 2 or 3.

More suitably, in some aspects, m is 1.

More suitably, in other aspects, m is o. n

In one aspect, suitably, n is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12.

Suitably n is o, 1, 2, 3, 4, 5 or 6. Suitably, n is o, 1, 2 or 3. Suitably, n is 1, 2 or 3.

More suitably, in some aspects, n is 1.

More suitably, in other aspects, n is o.

U

Suitably, in some aspects, p is 1.

Suitably, when p is 1, then H represents a single bond.

More suitably, when p is 1, then H is a C 5 heteroaryl group optionally substituted with 1 or 2 optional substituent groups independently selected from OH, C - 2 alkyl, OC 1-12 alkyl and RA.

Suitably, in other aspects, p is o.

Stereochemistry

The compounds of formula (I) have a chiral center at the carbon where the B-ring and C-ring are fused together. Suitably, in any of the previous aspects of the invention, the compound of formula (I) comprises, or consists essentially of, or consists of a racemic mixture comprising both the (R)- and (S)-confrguration at the carbon where the B-ring and C-ring are fused together. Alternatively, suitably, in any of the previous aspects, the compound of formula (I) comprises, or consists essentially of, or consists of the (R)-configuration at the carbon where the B-ring and C-ring are fused together. Thus, the compound of formula (I) is a compound of formula (IR):

Alternatively, more suitably, in any of the previous aspects, the compound of formula (I) comprises, or consists essentially of, or consists of the (S)-configuration at the carbon where the B-ring and C-ring are fused together. Thus, the compound of formula (I) is a compound of formula (IS):

Suitably for compounds of formulas (IR) and (IS) f is 1.

Compounds

Suitably, the compound of formula (I) is:

(a) (S)-AT-(4-Aminophenyl)-4-(4-(4-(4-((2-methoxy-i2-oxo-5,6,6a, 7,8,9,io,i2- octahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i-methyl-iif- pyrrole-2-carboxamido)phenyl)-i-methyl-iff-pyrrole-2-carboxa mide (16);

(b) Methyl (S)-5-(4-(4-((2-methoxy-i2-oxo-5,6,6a,7,8,9,io,i2- octahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i-methyl-iff- pyrrole-2-carboxamido)benzo[b]thiophene-2-carboxylate (38);

(c) Methyl (S)-5-(4-(4-((2-methoxy-i2-oxo-5,6,6a,7,8,9,io,i2- octahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i-methyl-iff- imidazole-2-carboxamido)benzo[b]thiophene-2-carboxylate (43);

(d) Methyl (S)-4-(4-(4-(4-((2-methoxy-i2-oxo-5,6,6a,7,8,9,io,i2- octahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i-methyl-iff- pyrrole-2-carboxamido)phenyl)-i-methyl-iff-pyrrole-2-carboxy late (45);

(e) V-(4-((S)-2-((S)-2-(6-(2,5-Dioxo-2,5-dihydro-i/f-pyrrol-i-yl )hexanamido)-3- methylbutanamido)propanamido)phenyl)-4-(4-(4-(4-(((S')-2-met hoxy-i2-oxo-5,6,6a,7, 8,9,io,i2-octahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)o xy)butanamido)-i- methyl-iff-pyrrole-2-carboxamido)phenyl)-i-methyl-iff-pyrrol e-2-carboxamide (47); (f) A/-(4-((S , )-2-((S')-2-amino-3-methylbutanamido)propanamido)pheny l)-4-(4-(4-(4- (((S)-2-methoxy-i2-oxo-5,6,6a,7,8,9,io,i2-octahydrobenzo[e]p yrido[i,2- <3][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iii-pyrrol e-2-carboxamido)phenyl)-i- methyl-iii-pyrrole-2-carboxamide (51);

(g) (S)-4-(4-((2-Methoxy-i2-oxo-5,6,6a,7,8,9,io,i2-octahydrobenz o[e]pyrido[i,2- <3][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-A/-(4-(i-m ethyl-5-(p-tolylcarbamoyl)- ifi-pyrrol-3-yl)phenyl)-ifi-pyrrole-2-carboxamide (54);

(h) (S)-V-(4-Aminophenyl)-4-(4-((2-methoxy-i2-oxo-5,6,6a,7,8,9,i o,i2- octahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i-methyl-ifi- pyrrole-2-carboxamide (58);

(i) Methyl GS)-5-(4-(4-((2-methoxy-i2-oxo-5,6,6a,7,8,9,io,i2- octahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i-methyl-if/- imidazole-2-carboxamido)benzo[d]oxazole-2-carboxylate (61);

(j) Methyl (S)-5-(4-(4-((2-methoxy-i2-oxo-5,6,6a,7,8,9,io,i2- octahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i-methyl-i/7- imidazole-2-carboxamido)-i//-benzo[d]imidazole-2-carboxylate (64);

(k) (S)-4-(4-((2-Methoxy-i2-oxo-5,6,6a,7,8,9,io,i2-octahydrobenz o[e]pyrido-[i,2- <3][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-A/-(4-(i-m ethyl-5-(phenylcarbamoyl)- i//-pyrrol-3-yl)phenyl)-i//-pyrrole-2-carboxamide (67);

(l) (S)-V-(4-Acetamidophenyl)-4-(4-(4-(4-((2-methoxy-i2-oxo-5,6, 6a,7,8,9,io,i2- octahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i-methyl-i/7- pyrrole-2-carboxamido)phenyl)-i-methyl-iii-pyrrole-2-carboxa mide (70);

(m) (S)-4-(4-((2-Methoxy-i2-oxo-5,6,6a,7,8,9,io,i2-octahydrobenz o[e]pyrido-[i,2- <3][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-A/-(4-(i-m ethyl-5-(pyridin-4- ylcarbamoyl)-iii-pyrrol-3-yl)phenyl)-iii-pyrrole-2-carboxami de (73);

(n) Methyl (S)-4-(4-(4-(4-(4-((2-methoxy-i2-oxo-5,6,6a,7,8,9,io,i2- octahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i-methyl-i/7- pyrrole-2-carboxamido)phenyl)-i-methyl-i/7-pyrrole-2-carboxa mido)-i-methyl-i/7- pyrrole-2-carboxylate (76);

(0) (S)-4-(4-((2-Methoxy-i2-oxo-5,6,6a,7,8,9,io,i2-octahydrobenz o[e]pyrido[i,2- <3][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-A/-(4-(i-m ethyl-5-(pyridin-3- ylcarbamoyl)-iii-pyrrol-3-yl)phenyl)-iii-pyrrole-2-carboxami de (79);

(p) Methyl (S)-4-(4-(4-(4-(4-((2-methoxy-i2-oxo-5,6,6a,7,8,9,io,i2- octahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i-methyl-i/7- pyrrole-2-carboxamido)phenyl)-i-methyl-iii-pyrrole-2-carboxa mido)benzoate (82); (q) Methyl (S')-4-(4-(4-(4-((7-methoxy-5-oxo-2,3,5,io,ii,na-hexahydro-i ii- benzo[e]pyrrolo[i,2-a][i,4]diazepin-8-yl)oxy)butanamido)-i-m ethyl-iii-pyrrole-2- carboxamido)phenyl)-i-methyl-ii/-pyrrole-2-carboxylate (91);

(r) (S')-A/-(4-Aminophenyl)-4-(4-(4-(4-((8-methoxy-6-oxo-n,i2,i2 a,i3-tetrahydro-6fi- benzo[5,6][i,4]diazepino[i,2-a]indol-9-yl)oxy)butanamido)-i- methyl-iii-pyrrole-2- carboxamido)phenyl)-i-methyl-ii/-pyrrole-2-carboxamide (106);

(s) (S')-A/-(4-Aminophenyl)-4-(4-(4-(4-((2-methoxy-i4-oxo-5,6,6a ,7,i2,i4- hexahydrobenzo[5,6][i,4]diazepino[i,2-b]isoquinolin-3-yl)oxy )butanamido)-i-methyl- Li/-pyrrole-2-carboxamido)phenyl)-i-methyl-ii/-pyrrole-2-car boxamide (119);

(t) Methyl (S')-4-(4-(4-(4-((2-methoxy-i4-oxo-5,6,6a,7,i2,i4- hexahydrobenzo[5,6][i,4]diazepino[i,2-b]isoquinolin-3-yl)oxy )butanamido)-i-methyl- l//-pyrrole-2-carboxamido)phenyl)-l-methyl-l//-pyrrole-2-car boxylate (121);

(u) (S)-4-(4-((2-Methoxy-i2-oxo-5,6,6a,7,8,9,io,i2-octahydrobenz o[e]pyrido[i,2- <2][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-A/-(4-(i-m ethyl-5-(phenyl carbamoyl)- l//-pyrrol-3-yl)phenyl)-l//-imidazole-2-carboxamide (126);

(v) (S)-4-(4-((2-Methoxy-i2-oxo-5,6,6a,7,8,9,io,i2-octahydrobenz o[e]pyrido[i,2- <3][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-A/-(4-(i-m ethyl-5-(pyridin-4- ylcarbamoyl)-i//-pyrrol-3-yl)phenyl)-i//-imidazole-2-carboxa mide (129);

(w) (S)-V-(4-(5-((4-Acetamidophenyl)carbamoyl)-i-methyl-i/7-pyrr ol-3-yl)phenyl)-4- (4-((2-methoxy-i2-oxo-5,6,6a,7,8,9,io,i2-octahydrobenzo[e]py rido[i,2- <3][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iii-imidaz ole-2-carboxamide (132); or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof.

Other features

Suitably, there is a proviso that when K I -R 33 is present, there is only one K I -R 33 group present.

In some embodiments, K I -R 33 is absent from the compound of formula (I).

Suitably, the compound of formula (I) contains only one primary or secondary amine.

Suitably, the compound of formula (I) contains only one primary amine, secondary amine or K I -R 33 group. Suitably, there is a proviso that when p is l and when H represents a single bond then f is o. More suitably, there is a proviso that when p is l and H represents a single bond then f is o and T is (iv) OH, 0C - 2 alkyl and R A .

Suitably, there is a proviso that when f is o, that T is (iv) OH, 0C - 2 alkyl and R A . More suitably, there is a proviso that when f is o, that T is R A .

Applications

The compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, or a pharmaceutical compositions comprising such compounds of formula (I) find application as a medicament.

The invention finds application in the treatment of a proliferative disease, a bacterial infection, a malarial infection and inflammation.

In certain aspects a method of treating a disease or condition selected from a proliferative disease, a bacterial infection, a malarial infection and inflammation is provided, the method comprising administering to a subject a therapeutically effective amount of a compound of the formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof or a composition comprising a compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof.

In certain aspects a method of treating a disease or condition selected from

proliferative diseases, bacterial infections, malaria and inflammation is provided, the method comprising administering to a subject a therapeutically effective amount of a targeted conjugate comprising a compound of the formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof.

In certain aspects a method of treating a proliferative disease is provided, the method comprising administering to a subject a therapeutically effective amount of an antibody-drug conjugate comprising a compound of the formula (I) or

pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof. The term“proliferative disease” refers to an unwanted or uncontrolled cellular proliferation of excessive or abnormal cells which is undesired, such as, neoplastic or hyperplastic growth, whether in vitro or in vivo. Examples of proliferative conditions include, but are not limited to, benign, pre-malignant, and malignant cellular proliferation, including but not limited to, neoplasms and tumours (e.g. histocytoma, glioma, astrocyoma, osteoma), cancers (e.g. lung cancer, small cell lung cancer, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, bowel cancer, colon cancer, hepatoma, breast cancer, glioblastoma, cervical cancer, ovarian cancer, oesophageal [or esophageal] cancer, oral cancer, prostate cancer, testicular cancer, liver cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, uterine cancer, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, head and neck cancer, bladder cancer, pancreas cancer, brain cancer, sarcoma, osteosarcoma, Kaposi’s sarcoma, melanoma), leukemias, psoriasis, bone diseases, fibroproliferative disorders (e.g. of connective tissues), and atherosclerosis. Suitably the proliferative disease is selected from bladder cancer, bone cancer, bowel cancer, brain cancer, breast cancer, cervical cancer, colon cancer, head and neck cancer, leukemia, liver cancer, lung cancer, lymphoma, melanoma, oesophageal cancer, oral cancer, ovarian cancer, pancreatic cancer, prostate cancer, rectal cancer, renal cancer, retinoblastoma, sarcoma, skin cancer, stomach cancer, testicular cancer, thyroid cancer and uterine cancer. Suitably the proliferative disease is selected from breast cancer and cervical cancer.

Suitably, the proliferative disease is selected from bladder cancer, bone cancer, bowel cancer, brain cancer, breast cancer, cervical cancer, colon cancer, head and neck cancer, leukemia, liver cancer, lung cancer, lymphoma, melanoma, oesophageal cancer, oral cancer, ovarian cancer, pancreatic cancer, prostate cancer, rectal cancer, renal cancer, retinoblastoma, sarcoma, skin cancer, stomach cancer, testicular cancer, thyroid cancer and uterine cancer.

Any type of cell may be treated, including but not limited to, bone, eye, head and neck, lung, gastrointestinal (including, e.g. mouth, oesophagus, bowel, colon), breast

(mammary), cervix, ovarian, uterus, prostate, liver (hepatic), kidney (renal), bladder, pancreas, brain, and skin.

A skilled person is readily able to determine whether or not a candidate compound treats a proliferative condition for any particular cell type.

8o Suitably subjects are human, livestock animals and companion animals.

In a further aspect, the compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, may be linked, either directly or indirectly, to a targeting agent (e.g., antibody, antibody fragment, hormone, etc.) to provide a targeted conjugate. The target conjugates of the present disclosure may contain one or multiple compounds of formula (I) (or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof). A variety of target conjugates are known in the art and may be used with a compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof. For example, in a particular aspect the target conjugate is an antibody-drug conjugate, wherein one or more compounds of formula (I) are linked, directly or indirectly, to the antibody. Therefore, the compound of formula (I) or

pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, maybe used as a payload on a targeted conjugate.

Suitably, a compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, for use as a drug in a targeted conjugate is prepared by attaching a compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof to a targeting agent, either directly or via an optional linker group. Suitably, the compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, is attached to a targeting agent via a linker group. Suitably, the targeted conjugate is for use in the treatment of a disease, more specifically of a proliferative disease. Suitably, the drug may be attached by any suitable functional group that it contains to the targeting agent either directly or via a linker group. Typically, the drug contains, or can be modified to contain, one or more functional groups such as amine, hydroxyl or carboxylic acid groups for attaching the drug to the targeting agent either directly or via a linker group. In some aspects, one or more atoms or groups of the compound of formula (I) may be eliminated during the attachment of the drug to the antibody. In some aspects, the targeting agent binds to a cell surface receptor or a tumor-associated antigen. In some aspects, the targeting agent is an antibody. In some aspects, the targeting agent is a hormone. In some aspects, the targeting agent is a protein. In some aspects, the targeting agent is a polypeptide. In some aspects, the targeting agent is a small molecule (for example, folic acid). Suitably, the present invention relates to a compound of formula (I) or

pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, for use in preparing a targeting conjugate (e.g. an antibody-drug conjugate). Suitably, a compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, maybe used directly to prepare a targeting conjugate when a compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, contains one or more functional groups such as amine, hydroxyl or carboxylic acid groups for attaching the drug to the targeting agent either directly or via a linker group. Suitably, a compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, maybe used in preparing a targeting conjugate by being modified to contain one or more functional groups such as amine, hydroxyl or carboxylic acid groups for attaching the drug to the targeting agent either directly or via a linker group. Suitably, a compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, may be used in preparing a targeting conjugate by being modified to contain one or more linker groups, wherein the targeting agent (such as an antibody) is attached to the drug through one or more linker groups. Therefore, the present invention provides for a compound of formula (I) further comprising one or more linker groups or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof. Suitably, a compound of formula (I) further comprises 1, 2 or 3 linker groups. Suitably, a compound of formula (I) further comprises 1 or 2 linker groups. Suitably, a compound of formula (I) further compries 1 linker group. In some aspects, one or more atoms or groups (such as H atoms or hydroxyl groups) of the compound of formula (I) may be eliminated during the attachment of the drug to the targeting agent (such as an antibody) or the attachment of the linker to the drug or the modification of the drug to contain one or more functional groups (such as amine, hydroxyl or carboxylic acid groups) for attaching the drug to the antibody either directly or via a linker group. In some aspects, where the compound of formula (I) further comprises a linker group that is attached to the rest of the compound of formula (I) by eliminating one or more atoms or groups (such as H atom or atoms or hydroxyl groups) from an R A group or by eliminating the R 7 group from a N-R 7 group.

Suitably such linker groups may comprise from 1-200 non-hydrogen atoms selected from C, N, O, S or halogen and may be branched, cyclic and/or unsaturated and, optionally, such linker groups may incorporate ether, oxo, carboxamidyl, urethanyl, heterocyclyl, aiyl, heteroaiyl, azide, alkyne, bisulfone, carbohydrazide, hydrazine, hydroxylamine, iodoacetamide, isothiocyanate, maleimide, phosphine,

pyrridopyridazine, R A , semihydrazide, succinimidyl ester, sulfodichlorophenol ester, sulfonyl halide, sulfosuccinimidyl ester, 4-sulfotetrafluorophenyl ester,

tetrafluorophenyl ester and thiazole moieties.

The compounds of formula (I) find application as payloads for antibodies or antibody fragments. The compounds of formula (I) readily allow conjugation to antibodies or antibody fragments.

In some aspects, the present invention relates to the treatment of a bacterial infection in a subject.

In some aspects, the compounds of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, are broad spectrum agents capable of treating a bacterial infection caused by Gram-positive bacteria and/or Gram- negative bacteria and/ or atypical bacteria.

Suitably the bacterial infection is caused by at least one bacterium selected from the genera Enterococcus, Staphylococcus, Streptococcus, Bacillus, Acinetobacter,

Burkholderia, Coxiella, Francisella, Yersina, Klebsiella, Escherichia, Enterobacter and Pseudomonas.

Suitably the bacterial infection is caused by at least one bacterium selected from the genera Enterococcus, Staphylococcus, Acinetobacter, Burkholderia, Klebsiella, Escherichia, Enterobacter and Pseudomonas.

Suitably the bacterial infection is caused by at least one bacterium selected from

Enterococcus faeculis, Enterococcus faecium, Staphylococcus aureus, Streptococcus pyogenes, Streptococcus pneumoniae, Streptococcus agalactiae, Bacillus anthracis, Bacillus cereus, Bacillus subtilis, Haemophilus influenzae, Acinetobacter baumannii, Burkholderia multivorans, Burkholderia cenocepacia, Burkholderia cepacia,

Burkholderia mallei, Burkholderia pseudomallei, Coxiella burnetii, Citrobacter freundii, Escherichia coli, Enterobacter cloacae, Enterobacter aerogenes, Francisella tularensis, Yersina pestis, Klebsiella pneumoniae, Serratia marcesens, Salmonella typhi, Salmonella typhimurum, Stenotrophomonas maltophilia, Pseudomonas aeruginosa and Neisseria gonorrhoeae. More suitably the bacterial infection is caused by at least one bacterium selected from Enterococcus faeculis, Enterococcus faecium, Staphylococcus aureus, Acinetobacter baumannii, Burkholderia multivorans, Burkholderia cenocepacia, Burkholderia cepacia, Escherichia coli, Klebsiella pneumonia and Pseudomonas aeruginosa.

In some embodiments, the bacterial infection is caused by Gram-positive bacteria selected from Enterococcus faeculis, Enterococcus faecium, Staphylococcus aureus, Streptococcus pyogenes, Streptococcus pneumoniae, Streptococcus agalactiae, Bacillus anthracis, Bacillus cereus and Bacillus subtilis.

In some embodiments, the infection is caused by Gram-negative bacteria, such as Haemophilus influenzae, Acinetobacter baumannii, Burkholderia multivorans, Burkholderia cenocepacia, Burkholderia cepacia, Burkholderia mallei, Burkholderia pseudomallei, Coxiella burnetii, Citrobacter freundii, Escherichia coli (such as e. coli K12), Enterobacter cloacae, Enterobacter aerogenes, Francisella tularensis, Yersina pestis, Klebsiella pneumoniae, Pseudomonas aeruginosa and Neisseria gonorrhoeae.

In some embodiments, the bacterial infection is caused by drug-resistant bacteria.

Such drug-resistant bacteria are bacteria that are resistant to one or more antibacterials other than the compounds of formula (I) described herein. The language“resistance” and“antibacterial resistance”“drug-resistant” refers to bacteria that are able to survive exposure to one or more antibacterial drugs. In some embodiments, the drug-resistant bacteria include Escherichia coli, Streptococcus pyogenes, Streptococcus agalactiae, Streptococcus pneumoniae (including penicillin-resistant Streptococcus pneumoniae), Staphylococcus aureus (including vancomycin-resistant Staphylococcus aureus (VRSA)), methicillin-resistant Staphylococcus aureus (MRSA) (including hospital- acquired MRSA, community acquired MRSA, epidemic MRSA (EMRSA, e.g. EMRSA 16) and coagulase negative staphylocci), Acinetobacter baumannii, Burkholderia multivorans, Burkholderia cenocepacia, Burkholderia cepacia, Klebsiella pneumoniae (such as KP4631), Pseudomonas aeruginosa and Neisseria gonorrhoeae (including penicillin-resistant Neisseria gonorrhoeae).

In some embodiments, the drug-resistant bacteria is a multiple drug resistant bacteria. The language“multiple drug resistant bacteria” includes bacteria that is resistant to two or more of antibiotics typically used for the treatment of such bacterial infections, for example, tetracycline, penicillin, cephalosporins (e.g., ceftriazone or cefixime), glycopeptides (e.g. vancomycin), quinolones (e.g., norfloxacin, ciprofloxacin or ofloxacin), co-trimoxazole, sulfonamides, aminoglycosides (e.g., kanamycin or gentamicin) and macrolides (e.g., azithromycin).

One of ordinary skill in the art is readily able to determine whether or not a candidate compound treats a bacterial infection by, for example, assays (such as those described in the examples) which may be used to determine the activity of a particular compound.

In some aspects, the present invention relates to the treatment of malaria in a subject.

In some aspects, the present invention relates to the treatment of inflammation in a subject.

Linker Group

A linker is a bifunctional compound which can be used to link a drug and a targeting moiety (e.g., an antibody) to form a targeted drug conjugate (e.g., an antibody-drug conjugate) or targeting conjugate. Such conjugates are useful in the treatment of disease as a drug (e.g., a cytotoxic agent) maybe delivered to a cell through recognition of an antigen.

In one aspect, a second section of the linker group is introduced which has a second reactive site (e.g., an electrophilic group) that is reactive to an opposing group (e.g., a nucleophilic group) present on a targeting agent such as an antibody. Useful nucleophilic groups on an antibody include, but are not limited to, sulfhydryl, hydroxyl and amino groups. In this instance, the heteroatom of the nucleophilic group of an antibody is reactive to an electrophilic group on a linker group and forms a covalent bond to that linker group. The electrophilic group then provides a site of attachment for the linker-payload or linker-drug, and can include the disulfide bridges of the antibody (i.e., a stochastic conjugation) or a residue containing an electrophilic group (either synthetic or naturally-occurring) introduced to the antibody to allow efficient conjugation (i.e., site-specific conjugation).

In another aspect, a linker group has a reactive site which has a nucleophilic group that is reactive to an electrophilic group present on an antibody. Electrophilic groups on an antibody include, but are not limited to, aldehyde and ketone carbonyl groups. The heteroatom of a nucleophilic group of a linker group can react with an electrophilic group on an antibody and form a covalent bond to the antibody. Nucleophilic groups in this respect may include, but are not limited to, hydrazide, oxime, amino, hydrazine, thiosemicarbazone, hydrazine carboxylate, and arylhydrazide. The electrophilic group on an antibody provides a convenient site for attachment to a linker group. For a more comprehensive list of linking technologies, please see Jain, N.; Smith, S. W.; Ghone, S.; Tomczuk, B., Current ADC Linker Chemistry. Pharmaceutical Research 2015, 32 (11), 3526-3540.

Linkers can either be cleavable or non-cleavable, with cleavable linkers normally represented by combinations of amino acids. The list of cleavable linkers includes, but is not limited to, valine-citruline, valine-alanine and any combination of two to eight amino acids. A self-immolative unit (e.g., a PAB spacer) can be included to assist with clean cleavage, and optionally hydrophilic groups (e.g., PEG) can be added to increase hydrophilicity of the construct. In some aspects, more suitably, the linker group comprises a self-immolative unit. A range of self immolative units are known in the art [30] and have been described in, for example, US Patent No. 7,754,681, European Patent Publication No. 0624377.

A variety of suitable linker groups are known in the art and may be used as described herein. For example, the maleimide methodology is routinely used as a method to attach antibodies to drug compounds by providing a linker attached to the drug with a terminal maleimide group. In addition, methodologies using diarylcyclooctyne moieties (such as, but not limited to, DBCO, dibenzylcyclooctyne) are known in the art. Diarylcyclooctynes react with stable azides to provide attachment via the formation of stable triazoles. Diarylcyclooctynes are thermostable with very narrow and specific reactivity toward azides, resulting in almost quantitative yields of stable triazoles. Furthermore, the reaction does not require a cytotoxic Cu(I) catalyst (that is toxic to most organisms) and thus, prevents its use in many biological systems. Still further, alkoxyamine methodologies are also alternatives in the art. For site-specific

conjugation of the drug to the antibody, the antibodies may comprise a“tag” (which may be proprietary) that will react with a diarylcyclooctyne (for example DBCO), an alkyoxyamine and/ or maleimide group to attach the antibody to the drug. The tag in some instances may be a mutated amino acid. Suitably linker groups incorporating the various groups described above are available in the art.

Antibody Drug Conjugates

Antibody therapy has been established for the targeted treatment of patients with cancer, immunological and angiogenic disorders (Carter, P. (2006) Nature Reviews Immunology 6:343-357). The use of antibody-drug conjugates (ADC), i.e. immunoconjugates, for the local delivery of cytotoxic or cytostatic agents, i.e. drugs to kill or inhibit tumor cells in the treatment of cancer, targets delivery of the drug moiety to tumors, and intracellular accumulation therein, whereas systemic administration of these unconjugated drug agents may result in unacceptable levels of toxicity to normal cells (Xie et al (2006) Expert. Opin. Biol. Ther. 6(3)1281 -291 ; Kovtun ef a/ (2006) Cancer Res. 66(6):3214-3121 ; Law et al (2006) CancerRes. 66(4):2328-2337; Wu et al (2005) Nature Biotech. 23(9): 1 137-1 145; Lambert J. (2005) Current Opin. in

Pharmacol. 5:543-549; Hamann P. (2005) Expert Opin. Ther. Patents 15(9): 1087-1 103; Payne, G. (2003) Cancer Cell 3:207-212; Trail ef a/ (2003) Cancer Immunol. Immunother. 52:328-337; Syrigos and Epenetos (1999) Anticancer Research 19:605- 614).

Maximal efficacy with minimal toxicity is sought thereby. Efforts to design and refine ADC have focused on the selectivity of monoclonal antibodies (mAbs) as well as drug mechanism of action, drug-linking, drug/antibody ratio (loading), and drug-releasing properties (Junutula, et al., 2008b Nature Biotech., 26(8):925-932; Doman et al., (2009) Blood U4(i3):272i -2729; US 7521541 ; US 7723485; WO2009/052249;

McDonagh (2006) Protein Eng. Design & Sel. 19(7): 299-307; Doronina et al., (2006) Bioconj. Chem. 17:114-124; Erickson et al., (2006) CancerRes. 66(8): 1-8; et al., (2005) Clin. CancerRes. 1 1 :843-852; Jeffrey et al., (2005) J. Med. Chem. 48:1344-1358;

Hamblett et al., (2004) Clin. Cancer Res. 10:7063- 7070). Drug moieties may impart their cytotoxic and cytostatic effects by mechanisms including tubulin binding, DNA binding, proteasome and/ or topoisomerase inhibition. Some cytotoxic drugs tend to be inactive or less active when conjugated to large antibodies or protein receptor ligands.

In some aspects, the present invention relates to a compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, for use as a drug in an antibody-drug conjugate. Suitably, a compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, for use as a drug in an antibody-drug conjugate is prepared by attaching a compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof to an antibody, either directly or via an optional linker group. Suitably, the compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, is attached to an antibody via a linker group. Suitably, the antibody-drug conjugate is for use in for treatment of a disease, more specifically of a proliferative disease. Suitably, the drug may be attached by any suitable functional group that it contains to the antibody either directly or via a linker group. Typically, the drug contains, or can be modified to contain, one or more functional groups such as amine, hydroxyl or carboxylic acid groups for attaching the drug to the antibody either directly or via a linker group. In some aspects, the antibody of the antibody drug conjugate is an antibody fragment, such as, but not limited to a single chain antibody. In some aspects, one or more atoms or groups of the compound of formula (I) may be eliminated during the attachment of the drug to the antibody. In some aspects, the antibody binds to a cell surface receptor or a tumor-associated antigen.

In some aspects, the present invention relates to the use of a compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, as a drug in an antibody-drug conjugate. Suitably, the use of a compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof, as a drug in an antibody-drug conjugate is accomplished by attaching a compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof to an antibody, either directly or via an optional linker group. Suitably, the compound of formula (I) or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers or mixtures thereof , is attached to an antibody via a linker group. Suitably, the antibody-drug conjugate is for use in for treatment of a disease, more specifically of a proliferative disease. Suitably, the drug maybe attached by any suitable functional group that it contains to the antibody either directly or via a linker group. Typically, the drug contains, or can be modified to contain, one or more functional groups such as amine, hydroxyl or carboxylic acid groups for attaching the drug to the antibody either directly or via a linker group. In some aspects, the antibody of the antibody drug conjugate is an antibody fragment, such as, but not limited to a single chain antibody. In some aspects, one or more atoms or groups of the compound of formula (I) may be eliminated during the attachment of the drug to the antibody. In some aspects, the antibody binds to a cell surface receptor or a tumor-associated antigen.

The substituent groups of the compounds of formula (I) may interact with DNA sequences and may be selected so as to target specific sequences. In particular, the following groups in compounds of formula (I): maybe selected to target specific sequences. Hence, when the substituent groups are tailored in this way, the compounds of formula (I) find application in targeted chemotherapy.

Antibody and antibody fragments

The term“antibody” specifically covers monoclonal antibodies, polyclonal antibodies, dimers, multimers, multispecific antibodies (e.g., bispecific antibodies), intact antibodies and antibody fragments, so long as they exhibit the desired biological activity, for example, the ability to bind a desired antigen on a target cell or tissue. Antibodies may be murine, human, humanized, chimeric, or derived from other species. An antibody is a protein generated by the immune system that is capable of recognizing and binding to a specific antigen. (Janeway, C, Travers, P., Walport, M., Shlomchik (2001) Immuno Biology, 5th Ed., Garland Publishing, New York). A target antigen generally has numerous binding sites, also called epitopes, recognized by CDRs on the antibody. Each antibody that specifically binds to a different epitope has a different structure. Thus, one antigen may have more than one corresponding antibody. An antibody includes a full-length immunoglobulin molecule or an immunologically active portion of a full-length immunoglobulin molecule, i.e., a molecule that contains an antigen binding site that immunospecifically binds an antigen of a target of interest or part thereof, such targets including but not limited to, cancer cell or cells that produce autoimmune antibodies associated with an

autoimmune disease. The immunoglobulin can be of any type (e.g. IgG, IgE, IgM, IgD, and IgA), class (e.g. lgGi , lgG2, lgG3, lgG4, lgAi and lgA2) or subclass, or allotype (e.g. human Gi mi , Gi m2, Gi m3, non-Gi mi [that, is any allotype other than Gi mi], Gi mi7, G2m23, G3m2i , G31T128, G3mi 1 , G31T15, G3mi3, G3mi4, G3mio, G3mi5, G31TL16, G3m6, G31T124, G31TL26, G31T127, A2mi , A2m2, Kmi , Km2 and Km3) of immunoglobulin molecule. The immunoglobulins can be derived from any species, including human, murine, or rabbit origin.

As used herein,“binds an epitope” is used to mean the antibody binds an epitope with a higher affinity than a non-specific partner such as Bovine Serum Albumin (BSA, Genbank accession no. CAA76847, version no. CAA76847.1 01:3336842, record update date: Jan 7, 201 1 02:30 PM). In some embodiments the antibody binds an epitope with an association constant (Ka) at least 2, 3, 4, 5, 10, 20, 50, 100, 200, 500, 1000, 2000, 5000, 10 4 , 10 5 or io 6 -fold higher than the antibody's association constant for BSA, when measured at physiological conditions. The term“antibody fragment” refers to a portion of a full length antibody, for example, the antigen binding or variable region thereof. Examples of antibody fragments include Fab, Fab', F(ab')2, and scFv fragments; diabodies; linear antibodies; fragments produced by a Fab expression library, anti-idiotypic (anti-id) antibodies, CDR

(complementary determining region), single-chain antibody molecules; and

multispecific antibodies formed from antibody fragments and epitope-binding fragments of any of the above which immunospecifically bind to target antigens, such as, for example, cancer cell antigens, viral antigens or microbial antigens,. The term “monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e. the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to polyclonal antibody preparations which include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant or epitope on the antigen. In addition to their specificity, the monoclonal antibodies are advantageous in that they may be synthesized uncontaminated by other antibodies.

The modifier“monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al (1975) Nature 256:495, or may be made by recombinant DNA methods (see, US 4816567). The monoclonal antibodies may also be isolated from phage antibody libraries using the techniques described in Clackson et al (1991 ) Nature, 352:624-628; Marks et al (1991) J. Mol.

Biol., 222:581-597 or from transgenic mice carrying a fully human immunoglobulin system (Lonberg (2008) Curr. Opinion 20(4):450-459).

The antibodies, including monoclonal antibodies, herein specifically include“chimeric” antibodies in which a portion of the antibody structure, for example the heavy and/ or light chain, is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (US 4816567; and Morrison et al (1984) Proc. Natl. Acad. Sci. USA, 81 :685i -6855). Chimeric antibodies include“primatized” antibodies comprising variable domain antigen-binding sequences derived from a non- human primate (e.g. Old World Monkey or Ape) and human constant region sequences. An“intact antibody” herein is one comprising VL and VH domains, as well as a light chain constant domain (CL) and heavy chain constant domains, CHi , CH2 and CH3. The constant domains maybe native sequence constant domains (e.g. human native sequence constant domains) or amino acid sequence variant thereof. The intact antibody may have one or more“effector functions” which refer to those biological activities attributable to the Fc region (a native sequence Fc region or amino acid sequence variant Fc region) of an antibody. Examples of antibody effector functions include Cl q binding; complement dependent cytotoxicity; Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; and down regulation of cell surface receptors such as B cell receptor and BCR.

Depending on the amino acid sequence of the constant domain of their heavy chains, intact antibodies can be assigned to different“classes.” There are hve major classes of intact antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into "subclasses" (isotypes), e.g., lgGi , lgG2, lgG3, lgG4, IgA, and lgA2. The heavy-chain constant domains that correspond to the different classes of antibodies are called a, d, e, g, and m, respectively. The subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.

The antibodies disclosed herein maybe modified. For example, to make them less immunogenic to a human subject. This may be achieved using any of a number of techniques familiar to the person skilled in the art, such as humanisation.

TUMOR- ASSOCIATED ANTIGENS:

(1) BMPRiB (bone morphogenetic protein receptor-type IB, Genbank accession no. NM_o 01203)

ten Dijke,P., et al Science 264 (5155): 101-104 (1994), Oncogene 14 (11): 1377- 1382 (1997); WO2004063362 (Claim 2); WO2003042661 (Claim 12); US2003134790-A1 (Page 38-39); WO2002102235 (Claim 13; Page 296); WO2003055443 (Page 91-92); WO200299122 (Example 2; Page 528-530); WO2003029421 (Claim 6);

WO2003024392 (Claim 2; Fig 112); WO200298358 (Claim 1; Page 183);

WO200254940 (Page 100-101); W0200259377(Page 349- 350); WO200230268 (Claim 27; Page 376); WO200148204 (Example; Fig 4) NP_ooii94 bone morphogenetic protein receptor, type IB /pid=NP_oo 1194.1 - Cross-references:

MIM:6q3248; NP_ooii94.i; AY065994

(2) E16 (LATi, SLC7A5, Genbank accession no. NM_003486)

Biochem. Biophys. Res. Commun. 255 (2), 283-288 (1999), Nature 395 (6699):288- 291 (1998), Gaugitsch, H.W., et al (1992) J. Biol. Chem. 267 (16): 11267-11273);

WO2004048938 (Example 2); WO2004032842 (Example TV); WO2003042661 (Claim 12); WO2003016475 (Claim 1); WO200278524 (Example 2); WO200299074 (Claim 19; Page 127-129); WO200286443 (Claim 27; Pages 222, 393); WO2003003906 (Claim 10; Page 293); WO200264798 (Claim 33; Page 93-95); WO200014228 (Claim 5; Page 133-136); US2003224454 (Fig 3); WO2003025138 (Claim 12; Page 150);

NP_003477 solute carrier family 7 (cationic amino acid transporter, y+ system), member 5 /pid=NP_003477-3 - Homo sapiens; Cross-references: MIM:6OOI82;

NP_003477-3; NM_oi5923; NM_003486_I

(3) STEAPi (six transmembrane epithelial antigen of prostate, Genbank accession no. NM_o 12449)

Cancer Res. 61 (15), 5857-5860 (2001), Hubert, R.S., et al (1999) Proc. Natl. Acad. Sci. U.SA. 96 (25): 14523-14528); WO2004065577 (Claim 6); WO2004027049 (Fig lL); EP1394274 (Example 11); WO2004016225 (Claim 2); WO2003042661 (Claim 12); US2003157089 (Example 5); US2003185830 (Example 5); US2003064397 (Fig 2); WO200289747 (Example 5; Page 618-619); WO2003022995 (Example 9; Fig 13A, Example 53; Page 173, Example 2; Fig 2A); NP 036581 six transmembrane epithelial antigen of the prostate; Cross-references: MIM:6q44ΐ5; NP 036581.1; NM_oi2449_i

(4) 0772P (CA125, MUC16, Genbank accession no. AF361486)

J. Biol. Chem. 276 (29):2737i-27375 (2001)); WO2004045553 (Claim 14);

WO200292836 (Claim 6; Fig 12); WO200283866 (Claim 15; Page 116-121);

US2003124140 (Example 16); US 798959; Cross-references: GL34501467;

AAK74120.3; AF36i486_i

(5) MPF (MPF, MSLN, SMR, megakaryocyte potentiating factor, mesothelin, Genbank accession no. NM_oos823) Yamaguchi, N., et al Biol. Chem. 269 (2), 805-808 (1994), Proc. Natl. Acad. Sci. U.SA. 96 (20): 11531-11536 (1999), Proc. Natl. Acad. Sci. U.SA. 93 (1): 136-140 (1996), J. Biol. Chem. 270 (37):21984-21990 (1995)); WO2003101283 (Claim 14); (WO2002102235 (Claim 13; Page 287-288); WO2002101075 (Claim 4; Page 308-309); WO200271928 (Page 320-321); WO9410312 (Page 52-57); Cross- references: MIM:6oi05i; NP_OOS8I4.2; NM_OOS823_I

(6) Napi2b (Napi3b, NAPI-3B, NPTIIb, SLC34A2, solute carrier family 34

(sodium phosphate), member 2, type II sodium-dependent phosphate transporter 3b,Genbank accession no. NM_oo6424) J. Biol. Chem. 277 (22): 19665-19672 (2002), Genomics 62 (2)1281-284 (1999), Feild, J.A., et al (1999) Biochem. Biophys. Res. Commun. 258 (3):578-582); WO2004022778 (Claim 2); EP1394274 (Example 11); WO2002102235 (Claim 13; Page 326); EP875569 (Claim 1; Page 17-19); WO200157188 (Claim 20; Page 329); WO2004032842 (Example IV); WO200175177 (Claim 24; Page 139-140); Cross-references: MIM:6q42ΐ7; NR_oo64ΐ5·ΐ; NM_OO6424_I

(7) Serna 5b (FU 10372, KIAA1445, Mm.42015, SEMA5B, SEMAG, Semaphorin 5b Hlog, sema domain, seven thrombospondin repeats (type 1 and type l-like), transmembrane domain (TM) and short cytoplasmic domain, (semaphorin) 5B, Genbank accession no. AB040878) Nagase T., et al (2000) DNA Res. 7 (2): 143-150); WO2004000997 (Claim 1); WO2003003984 (Claim 1); WO200206339 (Claim 1; Page 50); WO200188133 (Claim 1; Page 41-43, 48-58); WO2003054152 (Claim 20);

WO2003101400 (Claim 11); Accession: Q9P283; EMBL; AB040878; BAA95969.1. Genew; HGNC: 10737;

(8) PSCA hlg (2700050Ci2Rik, C5300080i6Rik, RIKEN cDNA 2700050C12, RIKEN cDNA 2700050C12 gene, Genbank accession no. AY358628); Ross et al (2002) Cancer Res. 62:2546-2553; US2003129192 (Claim 2); US2004044180 (Claim 12);

US2004044179 (Claim 11); US2003096961 (Claim 11); US2003232056 (Example 5); WO2003105758 (Claim 12); US2003206918 (Example 5); EP1347046 (Claim 1);

WO2003025148 (Claim 20); Cross-references: GL37182378; AAQ88991.1;

AY358628_I

(9) ETBR (Endothelin type B receptor, Genbank accession no. AY275463);

Nakamuta M., et al Biochem. Biophys. Res. Commun. 177, 34-39, 1991; Ogawa Y., et al Biochem. Biophys. Res. Commun. 178, 248-255, 1991; Arai H., et al Jpn. Circ. J. 56, 1303- 1307, 1992; Arai H., et al J. Biol. Chem. 268, 3463-3470, 1993; Sakamoto A., Yanagisawa M., et al Biochem. Biophys. Res. Commun. 178, 656-663, 1991;

Elshourbagy N. A., et al J. Biol. Chem. 268, 3873-3879, 1993; Haendler B., et al J. Cardiovasc. Pharmacol. 20, S1-S4, 1992; Tsutsumi M., et al Gene 228, 43-49, 1999; Strausberg R.L., et al Proc. Natl. Acad. Sci. U.SA. 99, 16899-16903, 2002; Bourgeois C, et al J. Clin. Endocrinol. Metab. 82, 3116- 3123, 1997; Okamoto Y., et al Biol. Chem. 272, 21589-21596, 1997; Verheij J.B., et alAm. J. Med. Genet. 108, 223-225, 2002; Hofstra R.M.W., et al Eur. J. Hum. Genet. 5, 180-185, 1997; Puffenberger E.G., et al Cell 79, 1257-1266, 1994; Attie T., et al, Hum. Mol. Genet. 4, 2407-2409, 1995;

Auricchio A., et al Hum. Mol. Genet. 5:351-354, 1996; Amiel J., et al Hum. Mol. Genet. 5, 355-357, 1996; Hofstra R.M.W., et al Nat. Genet. 12, 445-447, 1996; Svensson PJ., et al Hum. Genet. 103, 145-148, 1998; Fuchs S., et al Mol. Med. 7, 115-124, 2001; Pingault V., et al (2002) Hum. Genet. 111, 198-206; WO2004045516 (Claim 1); WO2004048938 (Example 2); WO2004040000 (Claim 151); WO2003087768 (Claim 1);

WO2003016475 (Claim 1); WO2003016475 (Claim 1); WO200261087 (Fig 1);

WO2003016494 (Fig 6); WO2003025138 (Claim 12; Page 144); WO200198351 (Claim 1; Page 124-125); EP522868 (Claim 8; Fig 2); WO200177172 (Claim 1; Page 297-299); US2003109676; US6518404 (Fig 3); US5773223 (Claim la; Col 31-34);

WO2004001004;

(10) MSG783 (RNF124, hypothetical protein FLJ20315, Genbank accession no.

NM_oi7703);

WO2003104275 (Claim 1); WO2004046342 (Example 2); WO2003042661 (Claim 12); WO2003083074 (Claim 14; Page 61); WO2003018621 (Claim 1); WO2003024392 (Claim 2; Fig 93); WO200166689 (Example 6); Cross-references: LocusID: 54894; NR_o6q233·2; NM_017703_I

(11) STEAP2 (HGNC_8639, IPCA-I, PCANAPi, STAMPi, STEAP2, STMP, prostate cancer associated gene 1, prostate cancer associated protein 1, six transmembrane epithelial antigen of prostate 2, six transmembrane prostate protein, Genbank accession no. AF455138)

Lab. Invest. 82 (11): 1573-1582 (2002); WO2003087306; US2003064397 (Claim 1; Fig 1); WO200272596 (Claim 13; Page 54-55); WO200172962 (Claim 1; Fig 4B);

WO2003104270 (Claim 11); WO2003104270 (Claim 16); US2004005598 (Claim 22); WO2003042661 (Claim 12); US2003060612 (Claim 12; Fig 10); WO200226822 (Claim 23; Fig 2); WO200216429 (Claim 12; Fig 10); Cross-references: GL22655488;

AAN04080.1; AF455138_I

(12) TrpM4 (BR22450, FU20041, TRPM4, TRPM4B, transient receptor potential cation channel, subfamily M, member 4, Genbank accession no. NM_oi7036)

Xu, X.Z., et al Proc. Natl. Acad. Sci. U.SA. 98 (19): 10692-10697 (2001), Cell 109 (3):397- 407 (2002), J. Biol. Chem. 278 (33):3q8ΐ3-3q82q (2003); US2003143557 (Claim 4); WO200040614 (Claim 14; Page 100-103); WO200210382 (Claim 1; Fig 9A); WO2003042661 (Claim 12); WO200230268 (Claim 27; Page 391); US2003219806 (Claim 4); WO200162794 (Claim 14; Fig lA-D); Cross-references: MIM:6o6936;

NP_O6OIO6.2; NM_017036_I

(13) CRIPTO (CR, CRi, CRGF, CRIPTO, TDGFi, teratocarcinoma-de rived growth factor, Genbank accession no. NP_003203 or NM_003212)

Ciccodicola, A., et al EMBO J. 8 (7): 1987-1991 (1989), Am. J. Hum. Genet. 49 (3)1555- 565 (1991); US2003224411 (Claim 1); WO2003083041 (Example 1); WO2003034984 (Claim 12); WO200288170 (Claim 2; Page 52-53); WO2003024392 (Claim 2; Fig 58); WO200216413 (Claim 1; Page 94-95, 105); WO200222808 (Claim 2; Fig 1);

US5854399 (Example 2; Col 17-18); US5792616 (Fig 2); Cross-references: MIM:

187395; NP_003203.I; NM _ 003212 _ 1

(14) CD21 (CR2 (Complement receptor 2) or C3DR (C3d/Epstein Barr virus receptor) or Hs.73792 Genbank accession no. M26004)

Fujisaku et al (1989) J. Biol. Chem. 264 (4):2ii8-2i25); Weis J.J., et al J. Exp. Med. 167, 1047-1066, 1988; Moore M., et al Proc. Natl. Acad. Sd. U.SA. 84, 9194-9198, 1987; Barel M., et al Mol. Immunol. 35, 1025-1031, 1998; Weis J. J., et al Proc. Natl. Acad. Sci. U.SA. 83, 5639-5643, 1986; Sinha S.K., et al (1993) J. Immunol. 150, 5311- 5320; WO2004045520 (Example 4); US2004005538 (Example 1); WO2003062401 (Claim 9); WO2004045520 (Example 4); WO9102536 (Fig 9.1-9.9); WO2004020595 (Claim 1); Accession: P20023; Q13866; Q14212; EMBL; M26004; AAA35786.1.

(15) CD79b (CD79B, CD79 , IGb (immunoglobulin-associated beta), B29, Genbank accession no. NM_ooo626 or 11038674)

Proc. Natl. Acad. Sd. U.SA. (2003) 100 (7):4126-4131, Blood (2002) 100 (9):3q68- 3076, Muller et al (1992) Eur. J. Immunol. 22 (6): 1621-1625); WO2004016225 (claim 2, Fig 140); WO2003087768, US2004101874 (claim 1, page 102); WO2003062401 (claim 9); WO200278524 (Example 2); US2002150573 (claim 5, page 15); US5644033; WO2003048202 (claim 1, pages 306 and 309); WO 99/558658, US6534482 (claim 13, Fig 17A/B); WO200055351 (claim 11, pages 1145-1146); Cross-references: MIM:

147245; NP_ooo6i7-i; NM_OOO626_I

(16) FCRH2 (IFGP4, IRTA4, SPAPiA (SH2 domain containing phosphatase anchor protein la), SPAPiB, SPAPiC, Genbank accession no. NM_030704, AY358130) Genome Res. 13 ( 10) 2265-2270 (2003), Immunogenetics 54 (2)187-95 (2002), Blood 99 (8):2662-2669 (2002), Proc. Natl. Acad. Sci. U.SA. 98 (17) 19772-9777 (2001), Xu, M.J., et al (2001) Biochem. Biophys. Res. Commun. 280 (3):768-775; WO2004016225 (Claim 2); WO2003077836; WO200138490 (Claim 5; Fig 18D-1-18D-2);

WO2003097803 (Claim 12); WO2003089624 (Claim 25); Cross-references:

MIM:6O6509; NP_IIO391.2; NM_030704_I

(17) HER2 (ErbB2, Genbank accession no. M11730)

Coussens L., et al Science (1985) 230(4730): 1132-1139); Yamamoto T., et al Nature 319, 230-234, 1986; Semba K., et al Proc. Natl. Acad. Sci. U.SA. 82, 6497- 6501, 1985; Swiercz J.M., et al J. Cell Biol. 165, 869-880, 2004; Kuhns J.J., et al J. Biol. Chem. 274, 36422-36427, 1999; Cho H.-S., et al Nature 421, 756-760, 2003; Ehsani A, et al (1993) Genomics 15, 426-429; WO2004048938 (Example 2); WO2004027049 (Fig ll);

WO2004009622; WO2003081210; WO2003089904 (Claim 9); WO2003016475 (Claim 1); US2003118592; WO2003008537 (Claim 1); WO2003055439 (Claim 29; Fig 1 A-B); WO2003025228 (Claim 37; Fig 5C); WO200222636 (Example 13; Page 95- 107); WO200212341 (Claim 68; Fig 7); WO200213847 (Page 71-74); WO200214503 (Page 114-117); WO200153463 (Claim 2; Page 41-46); WO200141787 (Page 15);

WO200044899 (Claim 52; Fig 7); WO200020579 (Claim 3; Fig 2); US5869445 (Claim 3; Col 31-38); WO9630514 (Claim 2; Page 56-61); EP1439393 (Claim 7);

WO2004043361 (Claim 7); WO2004022709; WO200100244 (Example 3; Fig 4);

Accession: P04626; EMBL; M11767; AAA35808.1. EMBL; M11761; AAA35808.1.

(18) NCA (CEACAM6, Genbank accession no. M18728);

Barnett T., et al Genomics 3, 59-66, 1988; Tawaragi Y., et al Biochem. Biophys. Res. Commun. 150, 89-96, 1988; Strausberg R.L., et al Proc. Natl. Acad. Sci. U.SA. 99: 16899- 16903, 2002; WO2004063709; EP 1439393 (Claim 7); WO2004044178 (Example 4); WO2004031238; WO2003042661 (Claim 12); WO200278524 (Example 2); WO200286443 (Claim 27; Page 427); WO200260317 (Claim 2); Accession: P40199; Q14920; EMBL; M29541; AAA59915.1. EMBL; M18728;

(19) MDP (DPEPi, Genbank accession no. BC017023)

Proc. Natl. Acad. Sci. U.SA. 99 (26): 16899-16903 (2002); WO2003016475 (Claim 1); WO200264798 (Claim 33; Page 85-87); JP05003790 (Fig 6-8); W09946284 (Fig 9); Cross-references: MIM: 179780; AAH17023.1; BCoi7023_i

(20) IL20RCX (IL2oRa, ZCYTOR7, Genbank accession no. AF 184971); Clark H.F., et al Genome Res. 13, 2265-2270, 2003; Mungall A.J., et al Nature 425, 805-811, 2003; Blumberg H., et al Cell 104, 9-19, 2001; Dumoutier L., et al J.

Immunol. 167, 3545-3549, 2001; Parrish-Novak J., et al J. Biol. Chem. 277, 47517- 47523, 2002; Pletnev S., et al (2003) Biochemistry 42: 12617-12624; Sheikh F., et al (2004) J. Immunol. 172, 2006-2010; EP1394274 (Example 11); US2004005320 (Example 5); WO2003029262 (Page 74-75); WO2003002717 (Claim 2; Page 63);

WO200222153 (Page 45-47); US2002042366 (Page 20-21); WO200146261 (Page 57- 59); WO200146232 (Page 63-65); W09837193 (Claim 1; Page 55-59); Accession:

Q9UHF4; Q6UWA9; Q96SH8; EMBL; AF 184971; AAF01320.1.

(21) Brevican (BCAN, BEHAB, Genbank accession no. AF229053)

Gary S.C., et al Gene 256, 139-147, 2000; Clark H.F., et al Genome Res. 13, 2265- 2270, 2003; Strausberg R.L., et al Proc. Natl. Acad. Sci. U.SA. 99, 16899-16903, 2002;

US2003186372 (Claim 11); US2003186373 (Claim 11); US2003119131 (Claim 1; Fig 52); US2003119122 (Claim 1; Fig 52); US2003119126 (Claim 1); US2003119121 (Claim 1; Fig 52); US2003119129 (Claim 1); US2003119130 (Claim 1); US2003119128 (Claim 1; Fig 52); US2003119125 (Claim 1); WO2003016475 (Claim 1); WO200202634 (Claim 1);

(22) EphB2R (DRT, ERK, Heks, EPHT3, Tyros, Genbank accession no. NM_004442) Chan,J. and Watt, V.M., Oncogene 6 (6), 1057-1061 (1991) Oncogene 10 (5)1897-905 (1995), Annu. Rev. Neurosci. 21 1309-345 (1998J, Int. Rev. Cytol. 196: 177-244 (2000); WO2003042661 (Claim 12); WO200053216 (Claim 1; Page 41); WO2004065576 (Claim 1); WO2004020583 (Claim 9); WO2003004529 (Page 128-132);

WO200053216 (Claim 1; Page 42); Cross-references: MIM: 600997; NP_004433.2; NM_004442_I

(23) ASLG659 (B7h, Genbank accession no. AX092328)

US20040101899 (Claim 2); WO2003104399 (Claim 11); WO2004000221 (Fig 3); US2003165504 (Claim 1); US2003124140 (Example 2); US2003065143 (Fig 60);

WO2002102235 (Claim 13; Page 299); US2003091580 (Example 2); WO200210187 (Claim 6; Fig 10); WO200194641 (Claim 12; Fig 7b); WO200202624 (Claim 13; Fig lA- lB); US2002034749 (Claim 54; Page 45-46); WO200206317 (Example 2; Page 320- 321, Claim 34; Page 321-322); WO200271928 (Page 468-469); WO200202587

(Example 1; Fig 1); WO200140269 (Example 3; Pages 190-192); WO200036107 (Example 2; Page 205-207); WO2004053079 (Claim 12); WO2003004989 (Claim 1); WO200271928 (Page 233-234, 452-453); WO 0116318; (24) PSCA (Prostate stem cell antigen precursor, Genbank accession no. AJ297436) Reiter R.E., et al Proc. Natl. Acad. Sci. U.SA. 95, 1735-1740, 1998; Gu Z., et al

Oncogene 19, 1288-1296, 2000; Biochem. Biophys. Res. Commun. (2000) 275(3)1783- 788; WO2004022709; EP1394274 (Example 11); US2004018553 (Claim 17);

WO2003008537 (Claim 1); WO200281646 (Claim 1; Page 164); WO2003003906 (Claim 10; Page 288); WO200140309 (Example 1; Fig 17); US2001055751 (Example 1; Fig lb); WO200032752 (Claim 18; Fig 1); WO9851805 (Claim 17; Page 97);

W09851824 (Claim 10; Page 94); WO9840403 (Claim 2; Fig lB); Accession: 043653; EMBL; AF043498; AAC39607.1.

(25) GEDA (Genbank accession No. AY260763);

AAP14954 lipoma HMGIC fusion-partner-like protein /pid=AAPi4954.i - Homo sapiens Species: Homo sapiens (human)

WO2003054152 (Claim 20); WO2003000842 (Claim 1); WO2003023013 (Example 3, Claim 20); US2003194704 (Claim 45); Cross-references: GL30102449; AAP14954.1; AU26q703_i

(26) BAFF-R (B cell -activating factor receptor, BLyS receptor 3, BR3, Genbank accession No. AF116456); BAFF receptor /pid=NP_443i77.i - Homo sapiens

Thompson, J.S., et al Science 293 (5537), 2108-2111 (2001); WO2004058309;

WO2004011611; WO2003045422 (Example; Page 32-33); WO2003014294 (Claim 35; Fig 6B); WO2003035846 (Claim 70; Page 615-616); WO200294852 (Col 136-137); WO200238766 (Claim 3; Page 133); WO200224909 (Example 3; Fig 3); Cross- references: MIM:6O6209; NP_443177.I; NM_052945_I; AF132600

(27) CD22 (B-cell receptor CD22-B isoform, BL-CAM, Lyb-8, Lyb8, SIGLEC-2, FLJ22814, Genbank accession No. AK026467);

Wilson et al (1991) J. Exp. Med. 173 : 137-146; WO2003072036 (Claim 1; Fig 1); Cross- references: MIM: 107266; NP 001762.1; NM_00l77l_l

(28) CD79a (CD79A, CD79CX, immunoglobulin-associated alpha, a B cell-specific protein that covalently interacts with Ig beta (CD79B) and forms a complex on the surface with Ig M molecules, transduces a signal involved in B-cell differentiation), pi: 4.84, MW: 25028 TM: 2 [P] Gene Chromosome: 19413.2, Genbank accession No. NP_ooi774-io)

WO2003088808, US20030228319; WO2003062401 (claim 9); US2002150573 (claim 4, pages 13-14); W09958658 (claim 13, Fig 16); WO9207574 (Fig 1); US5644033; Ha et al (1992) J. Immunol. 148(5): 1526-1531; Mueller et al (1992) Eur. J. Biochem. 22: 1621-1625; Hashimoto et al (1994) Immunogenetics 40(4)1287-295; Preud'homme et al (1992) Clin. Exp. Immunol. 90(1): 141-146; Yu et al (1992) J. Immunol. 148(2) 633-637; Sakaguchi et al (1988) EMBOJ. 7(n):3457-3464;

(29) CXCR5 (Burkitt's lymphoma receptor 1, a G protein-coupled receptor that is activated by the CXCL13 chemokine, functions in lymphocyte migration and humoral defense, plays a role in HIV-2 infection and perhaps development of AIDS, lymphoma, myeloma, and leukemia); 372 aa, pi: 8.54 MW: 41959 TM: 7 [P] Gene Chromosome: 1 iq23 3, Genbank accession No. NP_ooi707.i)

WO2004040000; WO2004015426; US2003105292 (Example 2); US6555339

(Example 2); WO200261087 (Fig 1); WO200157188 (Claim 20, page 269);

WO200172830 (pages 12- 13); WO200022129 (Example 1, pages 152-153, Example 2, pages 254-256); W09928468 (claim 1, page 38); US5440021 (Example 2, col 49-52); W09428931 (pages 56-58); W09217497 (claim 7, Fig 5); Dobner et al (1992) Eur. J. Immunol. 22:2795-2799; Barella et al (1995) Biochem. J. 309:773-779;

(30) HLA-DOB (Beta subunit of MHC class II molecule (la antigen) that binds peptides and presents them to CD4+ T lymphocytes); 273 aa, pi: 6.56 MW: 30820 TM: 1 [P] Gene Chromosome: 6p2i.3, Genbank accession No. NP_002in.i)

Tonnelle et al (1985) EMBO J. 4(n):2839-2847; Jonsson et al (1989) Immunogenetics 29(6):4ii-4i3; Beck et al (1992) J. Mol. Biol. 228:433-441; Strausberg et al (2002)

Proc. Natl. Acad. Sci USA 99: 16899-16903; Servenius et al (1987) J. Biol. Chem.

262:8759-8766; Beck et al (1996) J. Mol. Biol. 255: 1-13; Naruse et al (2002) Tissue Antigens 59:512-519; W09958658 (claim 13, Fig 15); US6153408 (Col 35-38);

US5976551 (col 168-170); US6011146 (col 145-146); Kasahara et al (1989)

Immunogenetics 3q(i):66-68; Larhammar et al (1985) J. Biol. Chem. 260(26): 14111- 14119;

(31) P2X5 (Purinergic receptor P2X ligand-gated ion channel 5, an ion channel gated by extracellular ATP, may be involved in synaptic transmission and neurogenesis, deficiency may contribute to the pathophysiology of idiopathic detrusor instability);

422 aa), pi: 7.63, MW: 47206 TM: 1 [P] Gene Chromosome: 17P13.3, Genbank accession No. NP_002552.2)

Le et al (1997) FEBSLett. 418(1-2): 195-199; WO2004047749; WO2003072035 (claim 10); Touchman et al (2000) Genome Res. 10: 165-173; WO200222660 (claim 20); WO2003093444 (claim 1); WO2003087768 (claim 1); WO2003029277 (page 82); (32) CD72 (B-cell differentiation antigen CD72, Lyb-2) PROTEIN SEQUENCE Full maeaity...tafrfpd (1..359; 359 aa), pi: 8.66, MW: 40225 TM: 1 [P] Gene Chromosome: 9R13·3 > Genbank accession No. NP_ooi773.i)

WO2004042346 (claim 65); WO2003026493 (pages 51-52, 57-58); WO200075655 (pages 105-106); Von Hoegen et al (1990) J. Immunol. i44(i2):4870-4877; Strausberg et al (2002) Proc. Natl. Acad. Sci USA 99: 16899-16903;

(33) LY64 (Lymphocyte antigen 64 (RP105), type I membrane protein of the leucine rich repeat (LRR) family, regulates B-cell activation and apoptosis, loss of function is associated with increased disease activity in patients with systemic lupus

erythematosis); 661 aa, pi: 6.20, MW: 74147 TM: 1 [P] Gene Chromosome: 5qi2, Genbank accession No. NP_005573.I)

US2002193567; WO9707198 (claim 11, pages 39-42); Miura et al (1996) Genomics 38(3):299-304; Miura et al (1998) Blood 92:2815-2822; WO2003083047; W09744452 (claim 8, pages 57-61); WO200012130 (pages 24-26);

(34) FcRHi (Fc receptor-like protein 1, a putative receptor for the immunoglobulin Fc domain that contains C2 type Ig-like and ITAM domains, may have a role in B- lymphocyte differentiation); 429 aa, pi: 5.28, MW: 46925 TM: 1 [P] Gene

Chromosome: iq2i-iq22, Genbank accession No. NP_443170.I)

WO2003077836; WO200138490 (claim 6, Fig 18E-1-18-E-2); Davis et al (2001) Proc. Natl. Acad. Sci USA 98(i7):9772-9777; WO2003089624 (claim 8); EP1347046 (claim 1); WO2003089624 (claim 7);

(35) IRTA2 (Immunoglobulin superfamily receptor translocation associated 2, a putative immunoreceptor with possible roles in B cell development and

lymphomagenesis; deregulation of the gene by translocation occurs in some B cell malignancies); 977 aa, pi: 6.88 MW: 106468 TM: 1 [P] Gene Chromosome: iq2i, Genbank accession No. Human: AF343662, AF343663, AF343664, AF343665, AF369794, AF397453, AK090423, AK090475, AL834187, AY358085; Mouse:

AK089756, AY158090, AY506558; NP_ii257i.i WO2003024392 (claim 2, Fig 97); Nakayama et al (2000) Biochem. Biophys. Res. Commun. 277(1): 124-127;

WO2003077836; WO200138490 (claim 3, Fig 18B-1-18B-2);

(36) TENB2 (TMEFF2, tomoregulin, TPEF, HPPi, TR, putative transmembrane proteoglycan, related to the EGF/heregulin family of growth factors and follistatin); 374 aa, NCBI Accession: AAD55776, AAF91397, AAG49451, NCBI RefSeq: NP_057276; NCBI Gene: 23671; OMIM: 605734; SwissProt Q9UIK5; Genbank accession No.

AF179274; AY358907, CAF85723, CQ782436

WO2004074320 (SEQ ID NO 810); JP2004113151 (SEQ ID NOS 2, 4, 8);

WO2003042661 (SEQ ID NO 580); WO2003009814 (SEQ ID NO 411); EP1295944 (pages 69-70); WO200230268 (page 329); WO200190304 (SEQ ID NO 2706);

US2004249130; US2004022727; WO2004063355; US2004197325; US2003232350; US2004005563; US2003124579; Horie et al (2000) Genomics 67: 146-152; Uchida et al (1999) Biochem. Biophys. Res. Comrnun. 266:593-602; Liang et al (2000) Cancer Res. 60:4907-12; Glynne- Jones et al (2001) IntJ Cancer. Oct 15594(2): 178-84;

(37) PMEL17 (silver homolog; SILV; D12S53E; PMEL17; SI; SIL); ME20; gpioo) BC001414; BT007202; M32295; M77348; NM_OO6928; McGlinchey, R.P. et al (2009) Proc. Natl. Acad. Sci. U.SA. 106 (33), 13731-13736; Rummer, M.P. et al (2009) J. Biol. Chem. 284 (4), 2296-2306;

(38) TMEFFi (transmembrane protein with EGF-like and two follistatin-like domains 1; Tomoregulin-i); H7365; C9orf2; C9ORF2; U19878; X83961; NM_o8o655;

NM_003092; Harms, P.W. (2003) Genes Dev. 17 (21), 2624-2629; Gery, S. et al (2003) Oncogene 22 (I8):2723-2727;

(39) GDNF-Rai (GDNF family receptor alpha 1; GFRAi; GDNFR; GDNFRA; RETLi; TRNRi; RETiL; GDNFR-alphai; GFR-ALPHA-i) ; U95847; BC014962; NM_145793 NM_005204; Kim, M.H. et al (2009) Mol. Cell. Biol. 29 (8), 2264-2277; Treanor, J.J. et al (1996) Nature 382 (6586):8o-83;

(40) Ly6E (lymphocyte antigen 6 complex, locus E, Ly67,RIG-E,SCA-2,TSA-l);

NP_002337.I; NM_002346.2; de Nooij-van Dalen, A G. et al (2003) Int. J. Cancer 103 (6), 768-774; Zammit, D.J. et al (2002) Mol. Cell. Biol. 22 (3):946-952; WO

2013/17705;

(41) TMEM46 (shisa homolog 2 (Xenopus laevis); SHISA2); NP_001007539.I;

NM_001007538.I; Furushima, K. et al (2007) Dev. Biol. 306 (2), 480-492; Clark, H.F. et al (2003) Genome Res. 13 (io):2205-2270; (42) Ly6G6D (lymphocyte antigen 6 complex, locus G6D; Ly6-D, MEGTl);

NP_O67079.2; NM_021246.2; Mallya, M. et al (2002) Genomics 80 (1): 113-123;

Ribas, G. et al (1999) J. Immunol. 163 (I):278-287;

(43) LGR5 (leucine-rich repeat-containing G protein-coupled receptor 5; GPR49, GPR67); NP_003058.I; NM_OO3667.2; Salanti, G. et al (2009 )Am. J. Epidemiol. 170 (5):537-545; Yamamoto, Y. et al (2003) Hepatology 37 (3):528-533;

(44) RET (ret proto-oncogene; MEN2A; HSCRi; MEN2B; MTCi; PTC; CDHF12;

Hs.168114; RET51; RET-ELEi); NP_o66i24.i; NM_020975-4; Tsukamoto, H. et al (2009) Cancer Sci. 100 (10): 1895-1901; Narita, N. et al (2009) Oncogene 28

(34):3058-3068;

(45) LY6K (lymphocyte antigen 6 complex, locus K; LY6K; HSJ001348; FLJ35226); NR_059997·3; NM_oΐ7527·3; Ishikawa, N. et al (2007) Cancer Res. 67 (24): 11601- 11611; de Nooij-van Dalen, A G. et al (2003) Int. J. Cancer 103 (6):768-774;

(46) GPR19 (G protein-coupled receptor 19; Mm.4787); NP_oo6i34.i; NM_oo6i43.2; Montpetit, A. and Sinnett, D. (1999) Hum. Genet. 105 (1-2): 162-164; O'Dowd, B.F. et al (1996) FEBSLett. 394 (3):325 329;

(47) GPR54 (KISSi receptor; KISSiR; GPR54; HOT7T175; AXOR12); NP_ii5940.2;

NM 032551.4; Navenot, J.M. et al (2009) Mol. Pharmacol. 75 (6): 1300-1306; Hata, K. et al (2009) Anticancer Res. 29 (2):6i7-623;

(48) ASPHDi (aspartate beta-hydroxylase domain containing 1; LOC253982);

NP_859O69.2; NM_i8i7i8.3; Gerhard, D.S. et al (2004) Genome Res. 14 (IOB):2121- 2127;

(49) Tyrosinase (TYR; OCALA; OCAiA; tyrosinase; SHEP3); NP_000363.i;

NM_000372.4; Bishop, D.T. et al (2009) Nat. Genet. 41 (8)1920-925; Nan, H. et al (2009) Int. J. Cancer 125 (4): 909-917;

(50) TMEM118 (ring finger protein, transmembrane 2; RNFT2; FLJ14627);

NP_OOIIO3373.I; NM 001109903.1; Clark, H.F. et al (2003) Genome Res. 13

(io):2205- 2270; Scherer, S.E. et al (2006) Nature 440 (7q82):346-35ΐ (51) GPR172A (G protein-coupled receptor 172A; GPCR41; FLJ11856; Di5Ertd747e); NP_078807.I; NM_024531.3; Ericsson, T.A. et al (2003) Proc. Natl. Acad. Sci. U.SA. too (1:0:6759-6764; Takeda, S. et al (2002) FEBS Lett. 520 (i-3):97-ioi.

(52) CD33, a member of the sialic acid binding, immunoglobulin-like lectin family, is a 67- kDa glycosylated transmembrane protein. CD33 is expressed on most myeloid and monocytic leukemia cells in addition to committed myelomonocytic and erythroid progenitor cells. It is not seen on the earliest pluripotent stem cells, mature

granulocytes, lymphoid cells, or nonhematopoietic cells (Sabbath et al., (1985) J. Clin. Invest. 75:756-56; Andrews et al, (1986) Blood 68: 1030-5). CD33 contains two tyrosine residues on its cytoplasmic tail, each of which is followed by hydrophobic residues similar to the immunoreceptor tyrosine-based inhibitory motif (ITIM) seen in many inhibitory receptors.

(53) CLL-i (CLEC12A, MICL, and DCAL2), encodes a member of the C-type lectin/C- type lectin-like domain (CTL/CTLD) superfamily. Members of this family share a common protein fold and have diverse functions, such as cell adhesion, cell-cell signalling, glycoprotein turnover, and roles in inflammation and immune response. The protein encoded by this gene is a negative regulator of granulocyte and monocyte function. Several alternatively spliced transcript variants of this gene have been described, but the full-length nature of some of these variants has not been determined. This gene is closely linked to other CTL/ CTLD superfamily members in the natural killer gene complex region on chromosome I2pi3 (Drickamer K (1999) Curr. Opin. Struct. Biol. 9 (53:585-90; van Rhenen A, et al, (2007) Blood 110 (7): 2659-66; Chen CH, et al. (2006) Blood 107 (4): 1459-67; Marshall AS, et al. (2006) Eur. J. Immunol.

36 (83:2159-69; Bakker AB, et al (2005) Cancer Res. 64 (223:8443-50; Marshall AS, et al (2004) J. Biol. Chem. 279 (15): 14792-802). CLL-i has been shown to be a type II transmembrane receptor comprising a single C-type lectin-like domain (which is not predicted to bind either calcium or sugar), a stalk region, a transmembrane domain and a short cytoplasmic tail containing an ITIM motif.

Anti-CD22 Antibodies

In certain embodiments, the anti-CD22 antibodies of an ADC comprises three light chain hypervariable regions (HVR-Li, HVR-L2 and HVR-L3) and three heavy chain hypervariable regions (HVR-Hi, HVR-H2 and HVR-H3), according to US 8226945:

HVR-Li RSSQSIVHSVGNTFLE (SEQ ID NO: 1) HVR-L2 KVSNRFS (SEQ ID NO: 2)

HVR-L3 FQGSQFPYT (SEQ ID NO: 3)

HVR-Hl GYEFSRSWMN (SEQ ID NO: 4)

HVR-H2 GRIYPGDGDTNYSGKFKG (SEQ ID NO: 5)

HVR-H3 DGSSWDWYFDV (SEQ ID NO: 6)

Anti-Ly6E Antibodies

In certain embodiments, an ADC comprises anti-Ly6E antibodies. Lymphocyte antigen 6 complex, locus E (Ly6E), also known as retinoic acid induced gene E (RIG-E) and stem cell antigen 2 (SCA-2). It is a GPI linked, 131 amino acid length, ~8.4kDa protein of unknown function with no known binding partners. It was initially identified as a transcript expressed in immature thymocyte, thymic medullary epithelial cells in mice (Mao, et al. (1996) Proc. Natl. Acad. Sci. U.SA. 93 :59io-59i4). In some embodiments, the invention provides an immunoconjugate comprising an anti-Ly6E antibody described in PCT Publication No. WO 2013/177055.

In some embodiments, the invention provides an antibody-drug conjugate comprising an anti-Ly6E antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-Hl comprising the amino acid sequence of SEQ ID NO: 12; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 13; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 14; (d) HVR-Li comprising the amino acid sequence of SEQ ID NO: 9; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 10; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 11.

In one aspect, the invention provides an antibody-drug conjugate comprising an antibody that comprises at least one, at least two, or all three VH HVR sequences selected from (a) HVR-Hl comprising the amino acid sequence of SEQ ID NO: 12; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 13; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 14. In a further embodiment, the antibody comprises (a) HVR-Hl comprising the amino acid sequence of SEQ ID NO: 12; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 13; and (c) HVR- H3 comprising the amino acid sequence of SEQ ID NO: 14. In another aspect, the invention provides an antibody-drug conjugate comprising an antibody that comprises at least one, at least two, or all three VL HVR sequences selected from (a) HVR-Li comprising the amino acid sequence of SEQ ID NO: 9; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 10; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 11. In one embodiment, the antibody comprises (a) HVR-Li comprising the amino acid sequence of SEQ ID NO: 9; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 10; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 11.

In another aspect, an antibody-drug conjugate of the invention comprises an antibody comprising (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 12, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 13, and (iii) HVR-H3 comprising an amino acid sequence selected from SEQ ID NO: 14; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-Li comprising the amino acid sequence of SEQ ID NO: 9, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 10, and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 11.

In another aspect, the invention provides an antibody-drug conjugate comprising an antibody that comprises (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 12; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 13; (c) HVR- H3 comprising the amino acid sequence of SEQ ID NO: 14; (d) HVR-Li comprising the amino acid sequence of SEQ ID NO: 9; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 10; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 11.

In any of the above embodiments, an anti-Ly6E antibody of an antibody-drug conjugate is humanized. In one embodiment, an anti-Ly6E antibody comprises HVRs as in any of the above embodiments, and further comprises a human acceptor framework, e.g. a human immunoglobulin framework or a human consensus framework.

In another aspect, an anti-Ly6E antibody of an antibody-drug conjugate comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 8. In certain embodiments, a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 8 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-Ly6E antibody comprising that sequence retains the ability to bind to Ly6E. In certain embodiments, a total of l to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 8. In certain embodiments, a total of l to 5 amino acids have been substituted, inserted and/ or deleted in SEQ ID NO: 8. In certain embodiments, substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti-Ly6E antibody comprises the VH sequence of SEQ ID NO: 8, including post-translational modifications of that sequence. In a particular embodiment, the VH comprises one, two or three HVRs selected from: (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 12, (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 13, and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 14.

In another aspect, an anti-Ly6E antibody of an antibody-drug conjugate is provided, wherein the antibody comprises a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 7. In certain embodiments, a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO:7 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-Ly6E antibody comprising that sequence retains the ability to bind to Ly6E. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 7. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 7. In certain embodiments, the substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti-Ly6E antibody comprises the VL sequence of SEQ ID NO: 7, including post- translational modifications of that sequence. In a particular embodiment, the VL comprises one, two or three HVRs selected from (a) HVR-Li comprising the amino acid sequence of SEQ ID NO: 9; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 10; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 11.

In another aspect, an antibody-drug conjugate comprising an anti-Ly6E antibody is provided, wherein the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above.

In one embodiment, an antibody-drug conjugate is provided, wherein the antibody comprises the VH and VL sequences in SEQ ID NO: 8 and SEQ ID NO: 7, respectively, including post-translational modifications of those sequences. In a further aspect, provided herein are antibody-drug conjugate comprising antibodies that bind to the same epitope as an anti-Ly6E antibody provided herein. For example, in certain embodiments, an immunoconjugate is provided comprising an antibody that binds to the same epitope as an anti-Ly6E antibody comprising a VH sequence of SEQ ID NO: 8 and a VL sequence of SEQ ID NO: 7, respectively.

In a further aspect of the invention, an anti-Ly6E antibody of an antibody-drug conjugate according to any of the above embodiments is a monoclonal antibody, including a human antibody. In one embodiment, an anti-Ly6E antibody of an antibody-drug conjugate is an antibody fragment, e.g., a Fv, Fab, Fab’, scFv, diabody, or F(ab’) 2 fragment. In another embodiment, the antibody is a substantially full length antibody, e.g., an IgGl antibody, IgG2a antibody or other antibody class or isotype as defined herein. In some embodiments, an immunconjugate (ADC) comprises an anti- Ly6E antibody comprising a heavy chain and a light chain comprising the amino acid sequences of SEQ ID NO: 16 and 15, respectively.

Table of Ly6E AntibodySequences

1q8

Anti-HER2 Antibodies

In certain embodiments, an ADC comprises anti-HER2 antibodies. In one embodiment of the invention, an anti-HER2 antibody of an ADC of the invention comprises a humanized anti-HER2 antibody, e.g., huMAb4D5-i, huMAb4D5-2, huMAb4D5-3, huMAb4D5-4, huMAb4D5-5, huMAb4D5-6, huMAb4D5-7 and huMAb4D5- 8, as described in Table 3 of US 5821337, which is specifically incorporated by reference herein. Those antibodies contain human framework regions with the complementarity- determining regions of a murine antibody (4D5) that binds to HER2. The humanized antibody huMAb4D5-8 is also referred to as trastuzumab, commercially available under the tradename HERCEPTIN®. In another embodiment of the invention, an anti- HER2 antibody of an ADC of the invention comprises a humanized anti-HER2 antibody, e.g., humanized 2C4, as described in US7862817. An exemplary humanized 2C4 antibody is pertuzumab, commercially available under the tradename PERJETA® .

In another embodiment of the invention, an anti-HER2 antibody of an ADC of the invention comprises a humanized 7C2 anti-HER2 antibody. A humanized 7C2 antibody is an anti-HER2 antibody.

In some embodiments, the invention provides an antibody-drug conjugate comprising an anti-HER2 antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 22; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, 27, or 28; (c) HVR- H3 comprising the amino acid sequence of SEQ ID NO: 24 or 29; (d) HVR-Li comprising the amino acid sequence of SEQ ID NO: 19; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21. In some embodiments, the invention provides an antibody-drug conjugate comprising an anti-HER2 antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 22; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24; (d) HVR-Li comprising the amino acid sequence of SEQ ID NO: 19; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21.

In one aspect, the invention provides an antibody-drug conjugate comprising an antibody that comprises at least one, at least two, or all three VH HVR sequences selected from (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 22; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, 27, or 28; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24 or 29. In one aspect, the invention provides an immunoconjugate comprising an antibody that comprises at least one, at least two, or all three VH HVR sequences selected from (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 22; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24. In a further embodiment, the antibody comprises (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 22; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, 27, or 28; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24 or 29. In a further embodiment, the antibody comprises (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 22; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24.

In another aspect, the invention provides an antibody-drug conjugate comprising an antibody that comprises at least one, at least two, or all three VL HVR sequences selected from (a) HVR-Li comprising the amino acid sequence of SEQ ID NO: 19; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21. In one embodiment, the antibody comprises (a) HVR-Li comprising the amino acid sequence of SEQ ID NO: 19; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO : 21.

In another aspect, an antibody-drug conjugate of the invention comprises an antibody comprising (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 22, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, 27, or 28, and (iii) HVR-H3 comprising an amino acid sequence selected from SEQ ID NO: 24 or 29; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-Li comprising the amino acid sequence of SEQ ID NO: 19, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20, and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21. In another aspect, an antibody-drug conjugate of the invention comprises an antibody comprising (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 22, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, and (iii) HVR-H3 comprising an amino acid sequence selected from SEQ ID NO: 24; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-Li comprising the amino acid sequence of SEQ ID NO: 19, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20, and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21.

In another aspect, the invention provides an antibody-drug conjugate comprising an antibody that comprises (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 22; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, 27, or 28;

(c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24 or 29; (d) HVR-Li comprising the amino acid sequence of SEQ ID NO: 19; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21. In another aspect, the invention provides an antibody-drug conjugate comprising an antibody that comprises (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 22; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24;

(d) HVR-Li comprising the amino acid sequence of SEQ ID NO: 19; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21.

In any of the above embodiments, an anti-HER2 antibody of an antibody-drug conjugate is humanized. In one embodiment, an anti-HER2 antibody of an antibody- drug conjugate comprises HVRs as in any of the above embodiments, and further comprises a human acceptor framework, e.g. a human immunoglobulin framework or a human consensus framework.

In another aspect, an anti-HER2 antibody of an antibody-drug conjugate comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 18. In certain embodiments, a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 18 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-HER2 antibody comprising that sequence retains the ability to bind to HER2. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 18. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 18. In certain embodiments, substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti- HER2 antibody comprises the VH sequence of SEQ ID NO: 18, including post- translational modifications of that sequence. In a particular embodiment, the VH comprises one, two or three HVRs selected from: (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 22, (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24

In another aspect, an anti-HER2 antibody of an antibody-drug conjugate is provided, wherein the antibody comprises a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 17. In certain embodiments, a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 17 contains substitutions (e.g., conservative

substitutions), insertions, or deletions relative to the reference sequence, but an anti- HER2 antibody comprising that sequence retains the ability to bind to HER2. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 17. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 17. In certain

embodiments, the substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti-HER2 antibody comprises the VL sequence of SEQ ID NO: 17, including post-translational modifications of that sequence. In a particular embodiment, the VL comprises one, two or three HVRs selected from (a) HVR-Li comprising the amino acid sequence of SEQ ID NO: 19; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21.

In another aspect, an antibody-drug conjugate comprising an anti-HER2 antibody is provided, wherein the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above. In one embodiment, an antibody-drug conjugate comprising an antibody is provided, wherein the antibody comprises the VH and VL sequences in SEQ ID NO: 18 and SEQ ID NO: 17, respectively, including post-translational modifications of those sequences.

In one embodiment, an antibody-drug conjugate comprising an antibody is provided, wherein the antibody comprises the humanized 7C2.V2.2.LA (hu7C2) K149C kappa light chain sequence of SEQ ID NO: 30

In one embodiment, an antibody-drug conjugate comprising an antibody is provided, wherein the antibody comprises the HU7C2 A118C IgGi heavy chain sequence of SEQ ID NO: 31

In a further aspect, provided herein are antibody-drug conjugates comprising antibodies that bind to the same epitope as an anti-HER2 antibody provided herein. For example, in certain embodiments, an immunoconjugate is provided, comprising an antibody that binds to the same epitope as an anti-HER2 antibody comprising a VH sequence of SEQ ID NO: 18 and a VL sequence of SEQ ID NO: 17, respectively.

In a further aspect of the invention, an anti-HER2 antibody of an antibody-drug conjugate according to any of the above embodiments is a monoclonal antibody, including a human antibody. In one embodiment, an anti-HER2 antibody of an immunoconjugate is an antibody fragment, e.g., a Fv, Fab, Fab’, scFv, diabody, or F(ab’) 2 fragment. In another embodiment, an immunoconjugate comprises an antibody that is a substantially full length antibody, e.g., an IgGl antibody, IgG2a antibody or other antibody class or isotype as defined herein.

Table of humanized 7C2 anti-HER2 antibody sequences

Anti-MUCi6 Antibodies In certain embodiments, an ADC comprises anti-MUCi6 antibodies.

In some embodiments, the invention provides an antibody-drug conjugate comprising an anti-MUCi6 antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 35; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 36; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 37; (d) HVR-Li comprising the amino acid sequence of SEQ ID NO: 32; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 33 and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 34-

In one aspect, the invention provides an antibody-drug conjugate comprising an antibody that comprises at least one, at least two, or all three VH HVR sequences selected from (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 35; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 36; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 37. In a further embodiment, the antibody comprises (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO:

35; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 36; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 37.

In another aspect, the invention provides an antibody-drug conjugate comprising an antibody that comprises at least one, at least two, or all three VL HVR sequences selected from (a) HVR-Li comprising the amino acid sequence of SEQ ID NO: 32; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 33; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 34. In one embodiment, the antibody comprises (a) HVR-Li comprising the amino acid sequence of SEQ ID NO:

32; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 33; and (c) HVR- L3 comprising the amino acid sequence of SEQ ID NO: 34.

In another aspect, an antibody-drug conjugate of the invention comprises an antibody comprising (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 35, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 36, and (iii) HVR-H3 comprising an amino acid sequence selected from SEQ ID NO: 37; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-Li comprising the amino acid sequence of SEQ ID NO: 32, (ii) HVR-L2 ll6 comprising the amino acid sequence of SEQ ID NO: 33, and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 34.

In another aspect, the invention provides an antibody-drug conjugate comprising an antibody that comprises (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 35 (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 36; (c) HVR- H3 comprising the amino acid sequence of SEQ ID NO: 37; (d) HVR-Li comprising the amino acid sequence of SEQ ID NO: 32; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 33; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 34-

In any of the above embodiments, an anti-MUCi6 antibody of an antibody-drug conjugate is humanized. In one embodiment, an anti-MUCi6 antibody comprises HVRs as in any of the above embodiments, and further comprises a human acceptor framework, e.g. a human immunoglobulin framework or a human consensus framework.

In another aspect, an anti-MUCi6 antibody of an antibody-drug conjugate comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 39. In certain embodiments, a VH sequence having at least 90%, 91%,

92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 39 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-MUCi6 antibody comprising that sequence retains the ability to bind to MUC16. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 39. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 39. In certain embodiments, substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti- MUC16 antibody comprises the VH sequence of SEQ ID NO: 39, including post- translational modifications of that sequence. In a particular embodiment, the VH comprises one, two or three HVRs selected from: (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 35, (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 36, and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 37- In another aspect, an anti-MUCi6 antibody of an antibody-drug conjugate is provided, wherein the antibody comprises a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 38. In certain embodiments, a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO:38 contains substitutions (e.g., conservative

substitutions), insertions, or deletions relative to the reference sequence, but an anti- MUC16 antibody comprising that sequence retains the ability to bind to MUC16. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 38. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 38. In certain

embodiments, the substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti-MUCi6 antibody comprises the VL sequence of SEQ ID NO: 38, including post-translational modifications of that sequence. In a particular embodiment, the VL comprises one, two or three HVRs selected from (a) HVR-Li comprising the amino acid sequence of SEQ ID NO: 32; (b) HVR- L2 comprising the amino acid sequence of SEQ ID NO: 33; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 34.

In another aspect, an antibody-drug conjugate comprising an anti-MUCi6 antibody is provided, wherein the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above.

In one embodiment, an antibody-drug conjugate is provided, wherein the antibody comprises the VH and VL sequences in SEQ ID NO: 39 and SEQ ID NO: 38, respectively, including post-translational modifications of those sequences.

In a further aspect, provided herein are antibody-drug conjugate comprising antibodies that bind to the same epitope as an anti-MUCi6 antibody provided herein. For example, in certain embodiments, an immunoconjugate is provided comprising an antibody that binds to the same epitope as an anti-MUCi6 antibody comprising a VH sequence of SEQ ID NO: 39 and a VL sequence of SEQ ID NO: 38, respectively.

In a further aspect of the invention, an anti-MUCi6 antibody of an antibody-drug conjugate according to any of the above embodiments is a monoclonal antibody, including a human antibody. In one embodiment, an anti-MUCi6 antibody of an antibody-drug conjugate is an antibody fragment, e.g., a Fv, Fab, Fab’, scFv, diabody, or F(ab’) 2 fragment. In another embodiment, the antibody is a substantially full length antibody, e.g., an IgGi antibody, IgG2a antibody or other antibody class or isotype as defined herein. Table of MUC16 Antibody Sequences

Anti-STEAP-i Antibodies In certain embodiments, an ADC comprises anti-STEAP-i antibodies.

In some embodiments, the invention provides an antibody-drug conjugate comprising an anti-STEAP-i antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 40; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 41; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 42; (d) HVR-Li comprising the amino acid sequence of SEQ ID NO: 43; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 44 and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 45-

In one aspect, the invention provides an antibody-drug conjugate comprising an antibody that comprises at least one, at least two, or all three VH HVR sequences selected from (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 40; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 41; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 42. In a further embodiment, the antibody comprises (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO:

40; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 41; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 42.

In another aspect, the invention provides an antibody-drug conjugate comprising an antibody that comprises at least one, at least two, or all three VL HVR sequences selected from (a) HVR-Li comprising the amino acid sequence of SEQ ID NO: 43; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 44; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 45. In one embodiment, the antibody comprises (a) HVR-Li comprising the amino acid sequence of SEQ ID NO:

43; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 44; and (c) HVR- L3 comprising the amino acid sequence of SEQ ID NO: 45.

In another aspect, an antibody-drug conjugate of the invention comprises an antibody comprising (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 40, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 41, and (iii) HVR-H3 comprising an amino acid sequence selected from SEQ ID NO: 42; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-Li comprising the amino acid sequence of SEQ ID NO: 43, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 44, and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 45.

In another aspect, the invention provides an antibody-drug conjugate comprising an antibody that comprises (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 40 (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 41; (c) HVR- H3 comprising the amino acid sequence of SEQ ID NO: 42; (d) HVR-Li comprising the amino acid sequence of SEQ ID NO: 43; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 44; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 45-

In any of the above embodiments, an anti-STEAP-i antibody of an antibody-drug conjugate is humanized. In one embodiment, an anti-STEAP-i antibody comprises HVRs as in any of the above embodiments, and further comprises a human acceptor framework, e.g. a human immunoglobulin framework or a human consensus framework.

In another aspect, an anti-STEAP-i antibody of an antibody-drug conjugate comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 46. In certain embodiments, a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 46 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-STEAP-i antibody comprising that sequence retains the ability to bind to STEAP-i. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 46.

In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 46. In certain embodiments, substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti- STEAP-i antibody comprises the VH sequence of SEQ ID NO: 46, including post- translational modifications of that sequence. In a particular embodiment, the VH comprises one, two or three HVRs selected from: (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 40, (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 41, and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 42. In another aspect, an anti-STEAP-i antibody of an antibody-drug conjugate is provided, wherein the antibody comprises a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 47. In certain embodiments, a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 47 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti- STEAP-i antibody comprising that sequence retains the ability to bind to STEAP-i. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 47 In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 47. In certain

embodiments, the substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti-STEAP-i antibody comprises the VL sequence of SEQ ID NO: 47, including post-translational modifications of that sequence. In a particular embodiment, the VL comprises one, two or three HVRs selected from (a) HVR-Li comprising the amino acid sequence of SEQ ID NO: 43; (b) HVR- L2 comprising the amino acid sequence of SEQ ID NO: 44; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 45.

In another aspect, an antibody-drug conjugate comprising an anti-STEAP-i antibody is provided, wherein the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above.

In one embodiment, an antibody-drug conjugate is provided, wherein the antibody comprises the VH and VL sequences in SEQ ID NO: 46 and SEQ ID NO: 47, respectively, including post-translational modifications of those sequences.

In a further aspect, provided herein are antibody-drug conjugate comprising antibodies that bind to the same epitope as an anti-STEAP-i antibody provided herein. For example, in certain embodiments, an immunoconjugate is provided comprising an antibody that binds to the same epitope as an anti-STEAP-i antibody comprising a VH sequence of SEQ ID NO: 46 and a VL sequence of SEQ ID NO: 47, respectively.

In a further aspect of the invention, an anti-STEAP-i antibody of an antibody-drug conjugate according to any of the above embodiments is a monoclonal antibody, including a human antibody. In one embodiment, an anti-STEAP-i antibody of an antibody-drug conjugate is an antibody fragment, e.g., a Fv, Fab, Fab’, scFv, diabody, or F(ab’) 2 fragment. In another embodiment, the antibody is a substantially full length antibody, e.g., an IgGl antibody, IgG2a antibody or other antibody class or isotype as defined herein. Table of STEAP Antibody Sequences

Anti-NaPi2b Antibodies In certain embodiments, an ADC comprises anti-NaPi2b antibodies. In some embodiments, the invention provides an antibody-drug conjugate comprising an anti- NaPi2b antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 48; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 49; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 50; (d) HVR-Li comprising the amino acid sequence of SEQ ID NO: 51; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 52 and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 53.

In one aspect, the invention provides an antibody-drug conjugate comprising an antibody that comprises at least one, at least two, or all three VH HVR sequences selected from (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 48; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 49; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 50. In a further embodiment, the antibody comprises (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO:

48; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 49; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 50.

In another aspect, the invention provides an antibody-drug conjugate comprising an antibody that comprises at least one, at least two, or all three VL HVR sequences selected from (a) HVR-Li comprising the amino acid sequence of SEQ ID NO: 51; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 52; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 53. In one embodiment, the antibody comprises (a) HVR-Li comprising the amino acid sequence of SEQ ID NO: 51; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 52; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 53.

In another aspect, an antibody-drug conjugate of the invention comprises an antibody comprising (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 48, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 49, and (iii) HVR-H3 comprising an amino acid sequence selected from SEQ ID NO: 50; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-Li comprising the amino acid sequence of SEQ ID NO: 51, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 52, and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 53.

In another aspect, the invention provides an antibody-drug conjugate comprising an antibody that comprises (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 48 (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 49; (c) HVR- H3 comprising the amino acid sequence of SEQ ID NO: 50; (d) HVR-Li comprising the amino acid sequence of SEQ ID NO: 51; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 52; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 53-

In any of the above embodiments, an anti-NaPi2b antibody of an antibody-drug conjugate is humanized. In one embodiment, an anti-NaPi2b antibody comprises HVRs as in any of the above embodiments, and further comprises a human acceptor framework, e.g. a human immunoglobulin framework or a human consensus framework.

In another aspect, an anti-NaPi2b antibody of an antibody-drug conjugate comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 54. In certain embodiments, a VH sequence having at least 90%, 91%,

92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 54 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-NaPi2b antibody comprising that sequence retains the ability to bind to NaPi2b. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 54. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 54. In certain embodiments, substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti- NaPi2b antibody comprises the VH sequence of SEQ ID NO: 54, including post- translational modifications of that sequence. In a particular embodiment, the VH comprises one, two or three HVRs selected from: (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 48, (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 49, and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 50.

In another aspect, an anti-NaPi2b antibody of an antibody-drug conjugate is provided, wherein the antibody comprises a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 55. In certain embodiments, a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 55 contains substitutions (e.g., conservative

substitutions), insertions, or deletions relative to the reference sequence, but an anti- NaPi2b antibody comprising that sequence retains the ability to bind to anti-NaPi2b. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 55. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 55. In certain

embodiments, the substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti-NaPi2b antibody comprises the VL sequence of SEQ ID NO: 55, including post-translational modifications of that sequence. In a particular embodiment, the VL comprises one, two or three HVRs selected from (a) HVR-Li comprising the amino acid sequence of SEQ ID NO: 51; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 52; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 53.

In another aspect, an antibody-drug conjugate comprising an anti-NaPi2b antibody is provided, wherein the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above.

In one embodiment, an antibody-drug conjugate is provided, wherein the antibody comprises the VH and VL sequences in SEQ ID NO: 54 and SEQ ID NO: 55, respectively, including post-translational modifications of those sequences.

In a further aspect, provided herein are antibody-drug conjugate comprising antibodies that bind to the same epitope as an anti-NaPi2b antibody provided herein. For example, in certain embodiments, an immunoconjugate is provided comprising an antibody that binds to the same epitope as an anti-NaPi2b antibody comprising a VH sequence of SEQ ID NO: 54 and a VL sequence of SEQ ID NO: 55, respectively.

In a further aspect of the invention, an anti-NaPi2b antibody of an antibody-drug conjugate according to any of the above embodiments is a monoclonal antibody, including a human antibody. In one embodiment, an anti-NaPi2b antibody of an antibody-drug conjugate is an antibody fragment, e.g., a Fv, Fab, Fab’, scFv, diabody, or F(ab’) 2 fragment. In another embodiment, the antibody is a substantially full length antibody, e.g., an IgGl antibody, IgG2a antibody or other antibody class or isotype as defined herein.

Table of NaPi2b Antibody Sequences

Anti-CD79b Antibodies

In certain embodiments, an ADC comprises anti-CD79b antibodies. In some embodiments, the invention provides an antibody-drug conjugate comprising an anti- CD79I) antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 58; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 59; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 60; (d) HVR-Li comprising the amino acid sequence of SEQ ID NO: 61; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 62; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 63.

In one aspect, the invention provides an antibody-drug conjugate comprising an antibody that comprises at least one, at least two, or all three VH HVR sequences selected from (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 58; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 59; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 60. In a further embodiment, the antibody comprises (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 58; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 59; and (c) HVR- H3 comprising the amino acid sequence of SEQ ID NO: 60.

In another aspect, the invention provides an antibody-drug conjugate comprising an antibody that comprises at least one, at least two, or all three VL HVR sequences selected from (a) HVR-Li comprising the amino acid sequence of SEQ ID NO: 61; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 62; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 63. In one embodiment, the antibody comprises (a) HVR-Li comprising the amino acid sequence of SEQ ID NO: 61; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 62; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 63.

In another aspect, an antibody-drug conjugate of the invention comprises an antibody comprising (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 58, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 59, and (iii) HVR-H3 comprising an amino acid sequence selected from SEQ ID NO: 60; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-Li comprising the amino acid sequence of SEQ ID NO: 61, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 62, and (iii) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 63.

In another aspect, the invention provides an antibody-drug conjugate comprising an antibody that comprises (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 58; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 59; (c) HVR- H3 comprising the amino acid sequence of SEQ ID NO: 60; (d) HVR-Li comprising the amino acid sequence of SEQ ID NO: 61; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 62; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 63.

In any of the above embodiments, an anti-CD79b antibody of an antibody-drug conjugate is humanized. In one embodiment, an anti-CD79b antibody comprises HVRs as in any of the above embodiments, and further comprises a human acceptor framework, e.g. a human immunoglobulin framework or a human consensus framework. In another aspect, an anti-CD79b antibody of an antibody-drug conjugate comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 56. In certain embodiments, a VH sequence having at least 90%, 91%,

92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 56 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-CD79b antibody comprising that sequence retains the ability to bind to CD79b. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 56. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 56. In certain embodiments, substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti- CD79b antibody comprises the VH sequence of SEQ ID NO: 8, including post- translational modifications of that sequence. In a particular embodiment, the VH comprises one, two or three HVRs selected from: (a) HVR-Hi comprising the amino acid sequence of SEQ ID NO: 58, (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 59, and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 60.

In another aspect, an anti-CD79b antibody of an antibody-drug conjugate is provided, wherein the antibody comprises a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 57. In certain embodiments, a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 57 contains substitutions (e.g., conservative

substitutions), insertions, or deletions relative to the reference sequence, but an anti- CD79b antibody comprising that sequence retains the ability to bind to CD79b. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 57. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 57. In certain

embodiments, the substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti-CD79b antibody comprises the VL sequence of SEQ ID NO: 57, including post-translational modifications of that sequence. In a particular embodiment, the VL comprises one, two or three HVRs selected from (a) HVR-Li comprising the amino acid sequence of SEQ ID NO: 61; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 62; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 63. In another aspect, an antibody-drug conjugate comprising an anti-CD79b antibody is provided, wherein the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above.

In one embodiment, an antibody-drug conjugate is provided, wherein the antibody comprises the VH and VL sequences in SEQ ID NO: 56 and SEQ ID NO: 57,

respectively, including post-translational modifications of those sequences. In a further aspect, provided herein are antibody-drug conjugate comprising antibodies that bind to the same epitope as an anti-CD79b antibody provided herein. For example, in certain embodiments, an immunoconjugate is provided comprising an antibody that binds to the same epitope as an anti-CD79b antibody comprising a VH sequence of SEQ ID NO: 56 and a VL sequence of SEQ ID NO: 57, respectively.

In a further aspect of the invention, an anti-CD79b antibody of an antibody-drug conjugate according to any of the above embodiments is a monoclonal antibody, including a human antibody. In one embodiment, an anti-CD79b antibody of an antibody-drug conjugate is an antibody fragment, e.g., a Fv, Fab, Fab’, scFv, diabody, or F(ab’) 2 fragment. In another embodiment, the antibody is a substantially full length antibody, e.g., an IgGl antibody, IgG2a antibody or other antibody class or isotype as defined herein.

Table of CD79b Antibody Sequences

Human HER2 Precursor Protein

Details of an exemplary human HER2 precursor protein with signal sequences is provided below

Antibody Affinity

In certain embodiments, an antibody provided herein has a dissociation constant (Kd) of < ΐmM, < too nM, < 50 nM, < 10 nM, < 5 nM, < 1 nM, < 0.1 nM, < 0.01 nM, or < 0.001 nM, and optionally is ³ 10 13 M. (e.g. io -8 M or less, e.g. from io -8 M to 10 13 M, e.g., from io -9 M to 10 13 M). In one embodiment, Kd is measured by a radiolabeled antigen binding assay (RIA) performed with the Fab version of an antibody of interest and its antigen as described by the following assay. Solution binding affinity of Fabs for antigen is measured by equilibrating Fab with a minimal concentration of ( 125 I)-labeled antigen in the presence of a titration series of unlabeled antigen, then capturing bound antigen with an anti- Fab antibody-coated plate (see, e.g., Chen et ah, J. Mol. Biol. 293 :865-88i(i999)). To establish conditions for the assay, MICROTITER® multi-well plates (Thermo

Scientific) are coated overnight with 5 pg/ml of a capturing anti-Fab antibody (Cappel Labs) in 50 mM sodium carbonate (pH 9.6), and subsequently blocked with 2% (w/v) bovine serum albumin in PBS for two to five hours at room temperature

(approximately 23°C). In a non-adsorbent plate (Nunc #269620), too pM or 26 pM [ 125 I]-antigen are mixed with serial dilutions of a Fab of interest (e.g., consistent with assessment of the anti-VEGF antibody, Fab-12, in Presta et ah, Cancer Res. 57:4593- 4599 (1997)). The Fab of interest is then incubated overnight; however, the incubation may continue for a longer period (e.g., about 65 hours) to ensure that equilibrium is reached. Thereafter, the mixtures are transferred to the capture plate for incubation at room temperature (e.g., for one hour). The solution is then removed and the plate washed eight times with 0.1% polysorbate 20 (TWEEN-20®) in PBS. When the plates have dried, 150 pl/well of scintillant (MICROSCF T-20™; Packard) is added, and the plates are counted on a TOPCOUNT™ gamma counter (Packard) for ten minutes. Concentrations of each Fab that give less than or equal to 20% of maximal binding are chosen for use in competitive binding assays.

According to another embodiment, Kd is measured using surface plasmon resonance assays using a BIACORE®-2000 or a BIACORE®-3000 (BIAcore, Inc., Piscataway, NJ) at 25°C with immobilized antigen CM5 chips at ~io response units (RU). Briefly, carboxymethylated dextran biosensor chips (CM5, BIACORE, Inc.) are activated with N- ethyl-V-(3-di methyl-ami nopropyl)-carbodiimide hydrochloride (EDC) and N- hydroxysuccinimide (NHS) according to the supplier's instructions. Antigen is diluted with 10 mM sodium acetate, pH 4.8, to 5 pg/ml (-0.2 pM) before injection at a flow rate of 5 pl/minute to achieve approximately 10 response units (RU) of coupled protein. Following the injection of antigen, 1 M ethanolamine is injected to block unreacted groups. For kinetics measurements, two-fold serial dilutions of Fab (0.78 nM to 500 nM) are injected in PBS with 0.05% polysorbate 20 (TWEEN-20™) surfactant (PBST) at 25°C at a flow rate of approximately 25 pl/min. Association rates (k on ) and dissociation rates (k 0ff ) are calculated using a simple one-to-one Langmuir binding model (BIACORE® Evaluation Software version 3.2) by simultaneously fitting the association and dissociation sensorgrams. The equilibrium dissociation constant (Kd) is calculated as the ratio k 0ff /k 0n , See, e.g., Chen et ah, J. Mol. Biol. 293 :86s-88i (1999). If the on-rate exceeds 10 6 M 1 s 1 by the surface plasmon resonance assay above, then the on-rate can be determined by using a fluorescent quenching technique that measures the increase or decrease in fluorescence emission intensity (excitation = 295 nm; emission = 340 nm, 16 nm band-pass) at 25°C of a 20 nM anti-antigen antibody (Fab form) in PBS, pH 7.2, in the presence of increasing concentrations of antigen as measured in a spectrometer, such as a stop-flow equipped spectrophotometer (Aviv Instalments) or a 8000-series SLM-AMINCO spectrophotometer (ThermoSpectronic) with a stirred cuvette.

Antibody Fragments

In certain embodiments, an antibody provided herein is an antibody fragment.

Antibody fragments include, but are not limited to, Fab, Fab’, Fab’-SH, F(ab’) 2 , Fv, and scFv fragments, and other fragments described below. For a review of certain antibody fragments, see Hudson et al. Nat. Med. 9: 129-134 (2003). For a review of scFv fragments, see, e.g., Pluckthun, in The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., (Springer- Verlag, New York), pp. 269-315 (1994); see also WO 93/ 16185; and U.S. Patent Nos. 5,571,894 and 5,587,458. For discussion of Fab and F(ab’) 2 fragments comprising salvage receptor binding epitope residues and having increased in vivo half-life, see U.S. Patent No. 5,869,046.

Diabodies are antibody fragments with two antigen-binding sites that may be bivalent or bispecific. See, for example, EP 404,097; WO 1993/01161; Hudson etal, Nat. Med. 9: 129-134 (2003); and Hollinger et al, Proc. Natl. Acad. Sci. USA 90: 6444-6448 (1993)· Triabodies and tetrabodies are also described in Hudson et al, Nat. Med. 9: 129-134 (2003).

Single-domain antibodies are antibody fragments comprising all or a portion of the heavy chain variable domain or ah or a portion of the light chain variable domain of an antibody. In certain embodiments, a single-domain antibody is a human single-domain antibody (Domantis, Inc., Waltham, MA; see, e.g., U.S. Patent No. 6,248,516). Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as production by recombinant host cells (e.g. E. coli or phage), as described herein.

Chimeric and Humanized Antibodies

In certain embodiments, an antibody provided herein is a chimeric antibody. Certain chimeric antibodies are described, e.g., in U.S. Patent No. 4,816,567; and Morrison et ah, Proc. Natl. Acad. Sci. USA, 81 :685i-6855 (1984)). In one example, a chimeric antibody comprises a non-human variable region (e.g., a variable region derived from a mouse, rat, hamster, rabbit, or non-human primate, such as a monkey) and a human constant region. In a further example, a chimeric antibody is a“class switched” antibody in which the class or subclass has been changed from that of the parent antibody. Chimeric antibodies include antigen-binding fragments thereof.

In certain embodiments, a chimeric antibody is a humanized antibody. Typically, a non-human antibody is humanized to reduce immunogenicity to humans, while retaining the specificity and affinity of the parental non-human antibody. Generally, a humanized antibody comprises one or more variable domains in which HVRs, e.g., CDRs, (or portions thereof) are derived from a non-human antibody, and FRs (or portions thereof) are derived from human antibody sequences. A humanized antibody optionally will also comprise at least a portion of a human constant region. In some embodiments, some FR residues in a humanized antibody are substituted with corresponding residues from a non-human antibody (e.g., the antibody from which the HVR residues are derived), e.g., to restore or improve antibody specificity or affinity.

Humanized antibodies and methods of making them are reviewed, e.g., in Almagro and Fransson, Front. Biosci. 13 : 1619-1633 (2008), and are further described, e.g., in Riechmann etah, Nature 332:323-329 (1988); Queen et ah, Proc. Nat’lAcad. Sci. USA 86: 10029-10033 (1989); US Patent Nos. 5,821,337, 7,527,791, 6,982,321, and

7,087,409; Kashmiri et al, Methods 36:25-34 (2005) (describing SDR (a-CDR) grafting); Padlan, Mol. Immunol. 28:489-498 (1991) (describing“resurfacing”);

Dall'Acqua et ah, Methods 36:43-60 (2005) (describing“FR shuffling”); and Osbourn et ah, Methods 36:61-68 (2005) and Klimka et ah, Br. J. Cancer, 83 :252-26o (2000) (describing the“guided selection” approach to FR shuffling). Human framework regions that may be used for humanization include but are not limited to: framework regions selected using the“best-fit” method (see, e.g., Sims et al. J. Immunol. 151 12296 (1993)); framework regions derived from the consensus sequence of human antibodies of a particular subgroup of light or heavy chain variable regions (see, e.g., Carter etal. Proc. Natl. Acad. Sci. USA, 89:4285 (1992); and Presta et al. J. Immunol, 151 :2023 (1993)); human mature (somatically mutated) framework regions or human germline framework regions (see, e.g., Almagro and Fransson, Front. Biosci. 13 : 1619-1633 (2008)); and framework regions derived from screening FR libraries (see, e.g., Baca et al, J. Biol. Chem. 272: 10678-10684 (1997) and Rosok et al, J. Biol. Chem. 271 :226II-226I8 (1996)).

Human Antibodies

In certain embodiments, an antibody provided herein is a human antibody. Human antibodies can be produced using various techniques known in the art. Human antibodies are described generally in van Dijk and van de Winkel, Curr. Opin.

Pharmacol. 5: 368-74 (2001) and Lonberg, Curr. Opin. Immunol. 20:450-459 (2008).

Human antibodies may be prepared by administering an immunogen to a transgenic animal that has been modified to produce intact human antibodies or intact antibodies with human variable regions in response to antigenic challenge. Such animals typically contain all or a portion of the human immunoglobulin loci, which replace the endogenous immunoglobulin loci, or which are present extrachromosomally or integrated randomly into the animal's chromosomes. In such transgenic mice, the endogenous immunoglobulin loci have generally been inactivated. For review of methods for obtaining human antibodies from transgenic animals, see Lonberg, Nat. Biotech. 23 : 1117-1125 (2005). See also, e.g., U.S. Patent Nos. 6,075,181 and 6,150,584 describing XENOMOUSE™ technology; U.S. Patent No. 5,770,429 describing

HuMAB® technology; U.S. Patent No. 7,041,870 describing K-M MOUSE®

technology, and U.S. Patent Application Publication No. US 2007/0061900, describing VELOCIMOUSE® technology). Human variable regions from intact antibodies generated by such animals may be further modified, e.g., by combining with a different human constant region.

Human antibodies can also be made by hybridoma-based methods. Human myeloma and mouse-human heteromyeloma cell lines for the production of human monoclonal antibodies have been described. (See, e.g., Kozbor J. Immunol, 133 : 3001 (1984); Brodeur et al, Monoclonal Antibody Production Techniques and Applications, pp. 51- 63 (Marcel Dekker, Inc., New York, 1987); and Boerner etal, J. Immunol., 147: 86 (1991).) Human antibodies generated via human B-cell hybridoma technology are also described in Li et al, Proc. Natl. Acad. Sci. USA, 103 .3557-3562 (2006). Additional methods include those described, for example, in U.S. Patent No. 7,189,826 (describing production of monoclonal human IgM antibodies from hybridoma cell lines) and Ni, Xiandai Mianyixue, 26(4):205-268 (2qq6) (describing human-human hybridomas). Human hybridoma technology (Trioma technology) is also described in Vollmers and Brandlein, Histology and Histopathology, 20(3)1927-937 (2005) and Vollmers and Brandlein, Methods and Findings in Experimental and Clinical Pharmacology, 27(3): 185-91 (2005).

Human antibodies may also be generated by isolating Fv clone variable domain sequences selected from human-derived phage display libraries. Such variable domain sequences may then be combined with a desired human constant domain. Techniques for selecting human antibodies from antibody libraries are described below.

Library-Derived Antibodies

Antibodies of the invention may be isolated by screening combinatorial libraries for antibodies with the desired activity or activities. For example, a variety of methods are known in the art for generating phage display libraries and screening such libraries for antibodies possessing the desired binding characteristics. Such methods are reviewed, e.g., in Hoogenboom et al. Methods in Molecular Biology 178: 1-37 (O'Brien et al, ed., Human Press, Totowa, NJ, 2001) and further described, e.g., in the McCafferty et al, Nature 348:552-554; Clackson etal, Nature 352: 624-628 (1991); Marks et al, J. Mol. Biol. 222: 581-597 (1992); Marks and Bradbury, Methods in Molecular Biology 248: 161-175 (Lo, ed., Human Press, Totowa, NJ, 2003); Sidhu et al, J. Mol. Biol. 338(2): 299-310 (2004); Lee et al, J. Mol. Biol. 340(5): 1073-1093 (2004); Fellouse, Proc.

Natl. Acad. Sci. USA 101(34): 12467-12472 (2004); and Lee et al, J. Immunol.

Methods 284(1-2): 119-132(2004).

In certain phage display methods, repertoires of VH and VL genes are separately cloned by polymerase chain reaction (PCR) and recombined randomly in phage libraries, which can then be screened for antigen-binding phage as described in Winter et al, Ann. Rev. Immunol, 12: 433-455 (1994). Phage typically display antibody fragments, either as single- chain Fv (scFv) fragments or as Fab fragments. Libraries from immunized sources provide high-affinity antibodies to the immunogen without the requirement of constructing hybridomas. Alternatively, the naive repertoire can be cloned (e.g., from human) to provide a single source of antibodies to a wide range of non-self and also self antigens without any immunization as described by Griffiths et al, EMBOJ 12: 725-734 (1993). Finally, naive libraries can also be made synthetically by cloning unrearranged V-gene segments from stem cells, and using PCR primers containing random sequence to encode the highly variable CDR3 regions and to accomplish rearrangement in vitro, as described by Hoogenboom and Winter, J. Mol. Biol, 227: 381-388 (1992). Patent publications describing human antibody phage libraries include, for example: US Patent No. 5,750,373, and US Patent Publication Nos. 2005/0079574, 2005/0119455, 2005/0266000, 2007/0117126, 2007/0160598, 2007/0237764, 2007/0292936, and 2009/0002360.

Antibodies or antibody fragments isolated from human antibody libraries are considered human antibodies or human antibody fragments herein.

Multispecific Antibodies

In certain embodiments, an antibody provided herein is a multispecific antibody, e.g. a bispecific antibody. Multispecific antibodies are monoclonal antibodies that have binding specificities for at least two different sites. In certain embodiments, bispecific antibodies may bind to two different epitopes of the same target. Bispecific antibodies may also be used to localize cytotoxic agents to cells which express the target. Bispecific antibodies can be prepared as full length antibodies or antibody fragments.

Techniques for making multispecific antibodies include, but are not limited to, recombinant co-expression of two immunoglobulin heavy chain-light chain pairs having different specificities (see Milstein and Cuello, Nature 305: 537 (1983)), WO 93/08829, and Traunecker et al, EMBOJ. 10: 3655 (1991)), and“knob-in-hole” engineering (see, e.g., U.S. Patent No. 5,731,168). The term“knob-into-hole” or“KnH” technology as used herein refers to the technology directing the pairing of two polypeptides together in vitro or in vivo by introducing a protuberance (knob) into one polypeptide and a cavity (hole) into the other polypeptide at an interface in which they interact. For example, KnHs have been introduced in the Fc:Fc binding interfaces, CL:CHi interfaces or VH/VL interfaces of antibodies (see, e.g., US 2011/0287009, US2007/0178552, WO 96/027011, WO 98/050431, Zhu et al, 1997, Protein Science 6:781-788, and WO2012/106587). In some embodiments, KnHs drive the pairing of two different heavy chains together during the manufacture of multispecific antibodies. For example, multispecific antibodies having KnH in their Fc regions can further comprise single variable domains linked to each Fc region, or further comprise different heavy chain variable domains that pair with similar or different light chain variable domains. KnH technology can be also be used to pair two different receptor extracellular domains together or any other polypeptide sequences that comprises different target recognition sequences (e.g., including affibodies, peptibodies and other Fc fusions).

The term“knob mutation” as used herein refers to a mutation that introduces a protuberance (knob) into a polypeptide at an interface in which the polypeptide interacts with another polypeptide. In some embodiments, the other polypeptide has a hole mutation.

The term“hole mutation” as used herein refers to a mutation that introduces a cavity (hole) into a polypeptide at an interface in which the polypeptide interacts with another polypeptide. In some embodiments, the other polypeptide has a knob mutation.

A brief nonlimiting discussion is provided below.

A“protuberance” refers to at least one amino acid side chain which projects from the interface of a first polypeptide and is therefore positionable in a compensatory cavity in the adjacent interface (i.e. the interface of a second polypeptide) so as to stabilize the heteromultimer, and thereby favor heteromultimer formation over homomultimer formation, for example. The protuberance may exist in the original interface or may be introduced synthetically (e.g., by altering nucleic acid encoding the interface). In some embodiments, nucleic acid encoding the interface of the first polypeptide is altered to encode the protuberance. To achieve this, the nucleic acid encoding at least one “original” amino acid residue in the interface of the first polypeptide is replaced with nucleic acid encoding at least one“import” amino acid residue which has a larger side chain volume than the original amino acid residue. It will be appreciated that there can be more than one original and corresponding import residue. The side chain volumes of the various amino residues are shown, for example, in Table 1 of US2011/0287009. A mutation to introduce a“protuberance” maybe referred to as a“knob mutation.”

In some embodiments, import residues for the formation of a protuberance are naturally occurring amino acid residues selected from arginine (R), phenylalanine (F), tyrosine (Y) and tryptophan (W). In some embodiments, an import residue is tryptophan or tyrosine. In some embodiment, the original residue for the formation of the protuberance has a small side chain volume, such as alanine, asparagine, aspartic acid, glycine, serine, threonine or valine.

A“cavity” refers to at least one amino acid side chain which is recessed from the interface of a second polypeptide and therefore accommodates a corresponding protuberance on the adjacent interface of a first polypeptide. The cavity may exist in the original interface or may be introduced synthetically (e.g. by altering nucleic acid encoding the interface). In some embodiments, nucleic acid encoding the interface of the second polypeptide is altered to encode the cavity. To achieve this, the nucleic acid encoding at least one“original” amino acid residue in the interface of the second polypeptide is replaced with DNA encoding at least one“import” amino acid residue which has a smaller side chain volume than the original amino acid residue. It will be appreciated that there can be more than one original and corresponding import residue. In some embodiments, import residues for the formation of a cavity are naturally occurring amino acid residues selected from alanine (A), serine (S), threonine (T) and valine (V). In some embodiments, an import residue is serine, alanine or threonine. In some embodiments, the original residue for the formation of the cavity has a large side chain volume, such as tyrosine, arginine, phenylalanine or tryptophan. A mutation to introduce a“cavity” may be referred to as a“hole mutation.”

The protuberance is“positionable” in the cavity which means that the spatial location of the protuberance and cavity on the interface of a first polypeptide and second polypeptide respectively and the sizes of the protuberance and cavity are such that the protuberance can be located in the cavity without significantly perturbing the normal association of the first and second polypeptides at the interface. Since protuberances such as Tyr, Phe and Trp do not typically extend perpendicularly from the axis of the interface and have preferred conformations, the alignment of a protuberance with a corresponding cavity may, in some instances, rely on modeling the protuberance/cavity pair based upon a three-dimensional structure such as that obtained by X-ray crystallography or nuclear magnetic resonance (NMR). This can be achieved using widely accepted techniques in the art.

In some embodiments, a knob mutation in an IgGl constant region is T366W (EU numbering). In some embodiments, a hole mutation in an IgGl constant region comprises one or more mutations selected from T366S, L368A and Y407V (EU numbering). In some embodiments, a hole mutation in an IgGl constant region comprises T366S, L368A andY407V (EU numbering).

In some embodiments, a knob mutation in an IgG4 constant region is T366W (EU numbering). In some embodiments, a hole mutation in an IgG4 constant region comprises one or more mutations selected from T366S, L368A, and Y407V (EU numbering). In some embodiments, a hole mutation in an IgG4 constant region comprises T366S, L368A, and Y407V (EU numbering).

Multi-specific antibodies may also be made by engineering electrostatic steering effects for making antibody Fc-heterodimeric molecules (WO 2009/089004A1); cross-linking two or more antibodies or fragments (see, e.g., US Patent No. 4,676,980, and Brennan et al, Science, 229: 81 (1985)); using leucine zippers to produce bi-specific antibodies (see, e.g., Kostelny etal, J. Immunol, 148(5): 1547-1553 (1992)); using“diabody” technology for making bispecific antibody fragments (see, e.g., Hollinger et al, Proc. Natl Acad. Sci. USA, 90:6444-6448 (1993)); and using single-chain Fv (sFv) dimers (see, e.g. Gruber et al, J. Immunol, 152:5368 (1994)); and preparing trispecific antibodies as described, e.g., in Tutt et al J. Immunol 147: 60 (1991).

Engineered antibodies with three or more functional antigen binding sites, including “Octopus antibodies,” are also included herein (see, e.g. US 2006/0025576A1).

The antibody or fragment herein also includes a“Dual Acting FAb” or“DAF” comprising an antigen binding site that binds to the target as well as another, different antigen (see, US 2008/0069820, for example).

Antibody Variants

In certain embodiments, amino acid sequence variants of the antibodies provided herein are contemplated. For example, it may be desirable to improve the binding affinity and/or other biological properties of the antibody. Amino acid sequence variants of an antibody may be prepared by introducing appropriate modifications into the nucleotide sequence encoding the antibody, or by peptide synthesis. Such modifications include, for example, deletions from, and/or insertions into and/or substitutions of residues within the amino acid sequences of the antibody. Any combination of deletion, insertion, and substitution can be made to arrive at the final construct, provided that the final construct possesses the desired characteristics, e.g., antigen-binding.

Substitution, Insertion, and Deletion Variants

In certain embodiments, antibody variants having one or more amino acid

substitutions are provided. Sites of interest for substitutional mutagenesis include the HVRs and FRs. Conservative substitutions are shown below in a Table of conservative substitutions under the heading of“preferred substitutions.” More substantial changes are provided in the Table under the heading of“exemplary substitutions,” and as further described below in reference to amino acid side chain classes. Amino acid substitutions may be introduced into an antibody of interest and the products screened for a desired activity, e.g., retained/improved antigen binding, decreased

immunogenicity, or improved ADCC or CDC.

Table of conservative substitutions

Amino acids maybe grouped according to common side-chain properties:

(1) hydrophobic: Norleucine, Met, Ala, Val, Leu, Ile;

(2) neutral hydrophilic: Cys, Ser, Thr, Asn, Gin;

(3) acidic: Asp, Glu;

(4) basic: His, Lys, Arg;

(5) residues that influence chain orientation: Gly, Pro;

(6) aromatic: Trp, Tyr, Phe.

Non-conservative substitutions will entail exchanging a member of one of these classes for another class.

One type of substitutional variant involves substituting one or more hypervariable region residues of a parent antibody (e.g. a humanized or human antibody). Generally, the resulting variant(s) selected for further study will have modifications (e.g., improvements) in certain biological properties (e.g., increased affinity, reduced immunogenicity) relative to the parent antibody and/or will have substantially retained certain biological properties of the parent antibody. An exemplary substitutional variant is an affinity matured antibody, which maybe conveniently generated, e.g., using phage display-based affinity maturation techniques such as those described herein. Briefly, one or more HVR residues are mutated and the variant antibodies displayed on phage and screened for a particular biological activity (e.g. binding affinity).

Alterations (e.g., substitutions) maybe made in HVRs, e.g., to improve antibody affinity. Such alterations maybe made in HVR“hotspots,” i.e., residues encoded by codons that undergo mutation at high frequency during the somatic maturation process (see, e.g., Chowdhury, Methods Mol. Biol. 207: 179-196 (2008)), and/or SDRs (a- CDRs), with the resulting variant VH or VL being tested for binding affinity. Affinity maturation by constructing and reselecting from secondary libraries has been described, e.g., in Hoogenboom et al. in Methods in Molecular Biology 178: 1-37 (O'Brien et al, ed., Human Press, Totowa, NJ, (2001).) In some embodiments of affinity maturation, diversity is introduced into the variable genes chosen for maturation by any of a variety of methods (e.g., error-prone PCR, chain shuffling, or oligonucleotide-directed mutagenesis). A secondary library is then created. The library is then screened to identify any antibody variants with the desired affinity. Another method to introduce diversity involves HVR-directed approaches, in which several HVR residues (e.g., 4-6 residues at a time) are randomized. HVR residues involved in antigen binding may be specifically identified, e.g., using alanine scanning mutagenesis or modeling. CDR-H3 and CDR-L3 in particular are often targeted.

In certain embodiments, substitutions, insertions, or deletions may occur within one or more HVRs so long as such alterations do not substantially reduce the ability of the antibody to bind antigen. For example, conservative alterations (e.g., conservative substitutions as provided herein) that do not substantially reduce binding affinity may be made in HVRs. Such alterations may be outside of HVR“hotspots” or SDRs. In certain embodiments of the variant VH and VL sequences provided above, each HVR either is unaltered, or contains no more than one, two or three amino acid

substitutions.

A useful method for identification of residues or regions of an antibody that may be targeted for mutagenesis is called“alanine scanning mutagenesis” as described by Cunningham and Wells (1989) Science, 244: 1081-1085. In this method, a residue or group of target residues (e.g., charged residues such as arg, asp, his, lys, and glu) are identified and replaced by a neutral or negatively charged amino acid (e.g., alanine or polyalanine) to determine whether the interaction of the antibody with antigen is affected. Further substitutions may be introduced at the amino acid locations demonstrating functional sensitivity to the initial substitutions. Alternatively, or additionally, a crystal structure of an antigen-antibody complex is used to identify contact points between the antibody and antigen. Such contact residues and

neighboring residues may be targeted or eliminated as candidates for substitution. Variants may be screened to determine whether they contain the desired properties.

Amino acid sequence insertions include amino- and/or carboxyl -terminal fusions ranging in length from one residue to polypeptides containing a hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues. Examples of terminal insertions include an antibody with an N-terminal methionyl residue. Other insertional variants of the antibody molecule include the fusion to the N- or C-terminus of the antibody to an enzyme (e.g. for ADEPT) or a polypeptide which increases the serum half-life of the antibody. Glycosylation variants

In certain embodiments, an antibody provided herein is altered to increase or decrease the extent to which the antibody is glycosylated. Addition or deletion of glycosylation sites to an antibody may be conveniently accomplished by altering the amino acid sequence such that one or more glycosylation sites is created or removed.

Where the antibody comprises an Fc region, the carbohydrate attached thereto maybe altered. Native antibodies produced by mammalian cells typically comprise a branched, biantennary oligosaccharide that is generally attached by an N-linkage to Asn297 of the CH2 domain of the Fc region. See, e.g., Wright et al. TIBTECH 15:26-32 (1997). The oligosaccharide may include various carbohydrates, e.g., mannose, N-acetyl

glucosamine (GlcNAc), galactose, and sialic acid, as well as a fucose attached to a GlcNAc in the“stem” of the biantennary oligosaccharide structure. In some

embodiments, modifications of the oligosaccharide in an antibody of the invention may be made in order to create antibody variants with certain improved properties.

In one embodiment, antibody variants are provided having a carbohydrate structure that lacks fucose attached (directly or indirectly) to an Fc region. For example, the amount of fucose in such antibody maybe from 1% to 80%, from 1% to 65%, from 5% to 65% or from 20% to 40%). The amount of fucose is determined by calculating the average amount of fucose within the sugar chain at Asn297, relative to the sum of all glycostructures attached to Asn 297 (e. g. complex, hybrid and high mannose structures) as measured by MALDI-TOF mass spectrometry, as described in WO 2008/077546, for example. Asn297 refers to the asparagine residue located at about position 297 in the Fc region (Eu numbering of Fc region residues); however, Asn297 may also be located about ± 3 amino acids upstream or downstream of position 297, i.e., between positions 294 and 300, due to minor sequence variations in antibodies. Such fucosylation variants may have improved ADCC function. See, e.g., US Patent Publication Nos. US 2003/0157108; US 2004/0093621. Examples of publications related to“defucosylated” or“fucose- deficient” antibody variants include: US

2003/0157108; WO 2000/61739; WO 2001/29246; US 2003/0115614; US

2002/0164328; US 2004/0093621; US 2004/0132140; US 2004/0110704; US

2004/0110282; US 2004/0109865; WO 2003/085119; WO 2003/084570; wo

2005/035586; WO 2005/035778; WO2005/053742; WO2002/031140; Okazaki et al. J. Mol. Biol. 336: 1239-1249 (2004); Yamane-Ohnuki et al. Biotech. Bioeng. 87: 614 (2004). Examples of cell lines capable of producing defucosylated antibodies include Led 3 CHO cells deficient in protein fucosylation (Ripka et al. Arch. Biochem. Biophys. 249:533-545 (1986); US Pat Appl No US 2003/0157108; and WO 2004/056312, especially at Example 11), and knockout cell lines, such as alpha-1,6- fucosyl transferase gene, FUT8, knockout CHO cells (see, e.g., Yamane-Ohnuki et al. Biotech. Bioeng. 87: 614 (2004); Kanda, Y. et al., Biotechnol. Bioeng., 94(4):68o-688 (2006); and

WO2003/085107).

Antibodies variants are further provided with bisected oligosaccharides, e.g., in which a biantennary oligosaccharide attached to the Fc region of the antibody is bisected by GlcNAc. Such antibody variants may have reduced fucosylation and/or improved ADCC function. Examples of such antibody variants are described, e.g., in WO 2003/011878 (Jean- Mairet et al); US Patent No. 6,602,684 (Umana et al); and US 2005/0123546 (Umana et al). Antibody variants with at least one galactose residue in the

oligosaccharide attached to the Fc region are also provided. Such antibody variants may have improved CDC function. Such antibody variants are described, e.g., in WO 1997/30087 (Patel et al); WO 1998/58964 (Raju, S.); and WO 1999/22764 (Raju, S.).

Fc region variants

In certain embodiments, one or more amino acid modifications may be introduced into the Fc region of an antibody provided herein, thereby generating an Fc region variant. The Fc region variant may comprise a human Fc region sequence (e.g., a human IgGl, IgG2, IgG3 or IgG4 Fc region) comprising an amino acid modification (e.g. a substitution) at one or more amino acid positions.

In certain embodiments, the invention contemplates an antibody variant that possesses some but not all effector functions, which make it a desirable candidate for applications in which the half life of the antibody in vivo is important yet certain effector functions (such as complement and ADCC) are unnecessary or deleterious. In vitro and/ or in vivo cytotoxicity assays can be conducted to confirm the reduction/depletion of CDC and/or ADCC activities. For example, Fc receptor (FcR) binding assays can be conducted to ensure that the antibody lacks FcyR binding (hence likely lacking ADCC activity), but retains FcRn binding ability. The primary cells for mediating ADCC, NK cells, express FcyRIII only, whereas monocytes express FcyRI, FcyRII and FcyRIII. FcR expression on hematopoietic cells is summarized in Table 3 on page 464 of Ravetch and Kinet, Annu. Rev. Immunol. 9:457-492 (1991). Non- limiting examples of in vitro assays to assess ADCC activity of a molecule of interest is described in U.S. Patent No. 5,500,362 (see, e.g. Hellstrom, I. et al. Proc. Natl Acad. Sci. USA 83 17059-7063 (1986)) and Hellstrom, I et ah, Proc. Nat'lAcad. Sci. USA 82: 1499-1502 (1985);

5,821,337 (see Bruggemann, M. et ah, J. Exp. Med. 166: 1351-1361 (1987)).

Alternatively, non-radioactive assays methods may be employed (see, for example, ACTI™ non-radioactive cytotoxicity assay for flow cytometry (CellTechnology, Inc. Mountain View, CA; and CytoTox 96® non-radioactive cytotoxicity assay (Promega, Madison, WI). Useful effector cells for such assays include peripheral blood

mononuclear cells (PBMC) and Natural Killer (NK) cells. Alternatively, or additionally, ADCC activity of the molecule of interest may be assessed in vivo, e.g., in an animal model such as that disclosed in Clynes et al. Proc. Nat'lAcad. Sci. USA 95:652-656 (1998). Ciq binding assays may also be carried out to confirm that the antibody is unable to bind Ciq and hence lacks CDC activity. See, e.g., Ciq and C3C binding ELISA in WO 2006/029879 and WO 2005/100402. To assess complement activation, a CDC assay may be performed (see, for example, Gazzano-Santoro et ah, J. Immunol.

Methods 202: 163 (1996); Cragg, M.S. et al., Blood 101 : 1045-1052 (2003); and Cragg, M.S. and M.J. Glennie, Blood 103 12738-2743 (2004)). FcRn binding and in vivo clearance/half life determinations can also be performed using methods known in the art (see, e.g., Petkova, S.B. et al., Int Ί. Immunol. 18(12): 1759-1769 (2006)).

In some embodiments, one or more amino acid modifications may be introduced into the Fc portion of the antibody provided herein in order to increase IgG binding to the neonatal Fc receptor. In certain embodiments, the antibody comprises the following three mutations according to EU numbering: M252Y, S254T, and T256E (the“YTE mutation”) (US Patent No. 8,697,650; see also Dall'Acqua et ah, Journal of Biological Chemistry 28I(33):23514-23524 (2006). In certain embodiments, the YTE mutation does not affect the ability of the antibody to bind to its cognate antigen. In certain embodiments, the YTE mutation increases the antibody's serum half-life compared to the native (i.e., non-YTE mutant) antibody. In some embodiments, the YTE mutation increases the serum half-life of the antibody by 3-fold compared to the native (i.e., non- YTE mutant) antibody. In some embodiments, the YTE mutation increases the serum half-life of the antibody by 2-fold compared to the native (i.e., non-YTE mutant) antibody. In some embodiments, the YTE mutation increases the serum half-life of the antibody by 4-fold compared to the native (i.e., non-YTE mutant) antibody. In some embodiments, the YTE mutation increases the serum half-life of the antibody by at least 5-fold compared to the native (i.e., non-YTE mutant) antibody. In some embodiments, the YTE mutation increases the serum half-life of the antibody by at least 10-fold compared to the native (i.e., non-YTE mutant) antibody. See, e.g., US Patent No. 8,697,650; see also Dall'Acqua et al., Journal of Biological Chemistry 28I(33):23514-23524 (2006).

In certain embodiments, the YTE mutant provides a means to modulate antibody- dependent cell-mediated cytotoxicity (ADCC) activity of the antibody. In certain embodiments, the YTEO mutant provides a means to modulate ADCC activity of a humanized IgG antibody directed against a human antigen. See, e.g., US Patent No. 8,697,650; see also Dall'Acqua et al., Journal of Biological Chemistry 28i(33):235i4- 23524 (2006).

In certain embodiments, the YTE mutant allows the simultaneous modulation of serum half-life, tissue distribution, and antibody activity (e.g., the ADCC activity of an IgG antibody). See, e.g., US Patent No. 8,697,650; see also Dall'Acqua etal, Journal of Biological Chemistry 28I(33):23514-23524 (2006).

Antibodies with reduced effector function include those with substitution of one or more of Fc region residues 238, 265, 269, 270, 297, 327 and 329 (U.S. Patent No.

6,737,056). Such Fc mutants include Fc mutants with substitutions at two or more of amino acid positions 265, 269, 270, 297 and 327, including the so-called“DANA” Fc mutant with substitution of residues 265 and 297 to alanine (US Patent No. 7,332,581).

In certain embodiments, the proline at position 329 (EU numbering ) (P329) of a wild- type human Fc region is substituted with glycine or arginine or an amino acid residue large enough to destroy the proline sandwich within the Fc/Fc gamma receptor interface, that is formed between the P329 of the Fc and tryptophane residues W87 and WHO of FcgRIII (Sondermann etal, Nature 406, 267-273 (20 July 2000)). In a further embodiment, at least one further amino acid substitution in the Fc variant is S228P, E233P, L234A, L235A, L235E, N297A, N297D, or P331S and still in another embodiment said at least one further amino acid substitution is L234A and L235A of the human IgGl Fc region or S228P and L235E of the human IgG4 Fc region, all according to EU numbering (U.S. Patent No. 8,969,526 which is incorporated by reference in its entirety).

In certain embodiments, a polypeptide comprises the Fc variant of a wild-type human IgG Fc region wherein the polypeptide has P329 of the human IgG Fc region substituted with glycine and wherein the Fc variant comprises at least two further amino acid substitutions at L234A and L235A of the human IgGl Fc region or S228P and L235E of the human IgG4 Fc region, and wherein the residues are numbered according to the EU numbering (U.S. Patent No. 8,969,526 which is incorporated by reference in its entirety). In certain embodiments, the polypeptide comprising the P329G, L234A and L235A (EU numbering) substitutions exhibit a reduced affinity to the human FCYRIILA and FCYRILA, for down-modulation of ADCC to at least 20% of the ADCC induced by the polypeptide comprising the wildtype human IgG Fc region, and/or for down-modulation of ADCP (U.S. Patent No. 8,969,526 which is

incorporated by reference in its entirety).

In a specific embodiment the polypeptide comprising an Fc variant of a wildtype human Fc polypeptide comprises a triple mutation: an amino acid substitution at position Pro329, a L234A and a L235A mutation according to EU numbering (P329 / LALA) (U.S. Patent No. 8,969,526 which is incorporated by reference in its entirety). In specific embodiments, the polypeptide comprises the following amino acid

substitutions: P329G, L234A, and L235A according to EU numbering.

Certain antibody variants with improved or diminished binding to FcRs are described. (See, e.g., U.S. Patent No. 6,737,056; WO 2004/056312, and Shields et ah, J. Biol. Chem. 9(2): 6591-6604 (2001).)

In certain embodiments, an antibody variant comprises an Fc region with one or more amino acid substitutions which improve ADCC, e.g., substitutions at positions 298,

333, and/or 334 of the Fc region (EU numbering of residues).

In some embodiments, alterations are made in the Fc region that result in altered (i.e., either improved or diminished) Ciq binding and/ or Complement Dependent

Cytotoxicity (CDC), e.g., as described in US Patent No. 6,194,551, WO 99/51642, and Idusogie etal. J. Immunol. 164: 4178-4184 (2000).

Antibodies with increased half lives and improved binding to the neonatal Fc receptor (FcRn), which is responsible for the transfer of maternal IgGs to the fetus (Guyer et ah, J. Immunol. 117:587 (1976) and Kim etal., J. Immunol. 24:249 (1994)), are described in US2005/0014934A1 (Hinton et ah). Those antibodies comprise an Fc region with one or more substitutions therein which improve binding of the Fc region to FcRn.

Such Fc variants include those with substitutions at one or more of Fc region residues: 238, 256, 265, 272, 286, 303, 305, 307, 3ii, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 413, 424 or 434, e.g., substitution of Fc region residue 434 (US Patent No.

7,371,826). See also Duncan & Winter, Nature 322:738-40 (1988); U.S. Patent No. 5,648,260; U.S. Patent No. 5,624,821; and WO 94/29351 concerning other examples of Fc region variants.

Cysteine engineered antibody variants

In certain embodiments, it may be desirable to create cysteine engineered antibodies, e.g., a“THIOMAB™” or TDC, in which one or more residues of an antibody are substituted with cysteine residues. In particular embodiments, the substituted residues occur at sites of the antibody that are available for conjugation. By substituting those residues with cysteine, reactive thiol groups are thereby positioned at accessible sites of the antibody and may be used to conjugate the antibody to other moieties, such as drug moieties or linker-drug moieties, to create an immunoconjugate, as described further herein. In certain embodiments, any one or more of the following residues maybe substituted with cysteine: K149 (Rabat numbering) of the light chain; V205 (Rabat numbering) of the light chain; A118 (EU numbering) of the heavy chain; A140 (EU numbering) of the heavy chain; L174 (EU numbering) of the heavy chain; Y373 (EU numbering) of the heavy chain; and S400 (EU numbering) of the heavy chain Fc region. In specific embodiments, the antibodies described herein comprise the HC- A140C (EU numbering) cysteine substitution. In specific embodiments, the antibodies described herein comprise the LC-R149C (Rabat numbering) cysteine substitution. In specific embodiments, the antibodies described herein comprise the HC-A118C (EU numbering) cysteine substitution. Cysteine engineered antibodies may be generated as described, e.g., in U.S. Patent No. 7,521,541.

In certain embodiments, the antibody comprises one of the following heavy chain cysteine substitutions:

In certain embodiments, the antibody comprises one of the following light chain cysteine substitutions:

A nonlimiting exemplary I1U7C2.V2.2.LA light chain (LC) K149C TΉIOMAB™ has the heavy chain and light chain amino acid sequences of SEQ ID NOs: 26 and 30, respectively. A nonlimiting exemplary hu7C2.v2.2.LA heavy chain (HC) A118C

THIOMAB™ has the heavy chain and light chain amino acid sequences of SEQ ID NOs: 31 and 25, respectively.

Antibody Derivatives In certain embodiments, an antibody provided herein may be further modified to contain additional nonproteinaceous moieties that are known in the art and readily available. The moieties suitable for derivatization of the antibody include but are not limited to water soluble polymers. Non-limiting examples of water soluble polymers include, but are not limited to, polyethylene glycol (PEG), copolymers of ethylene glycol/propylene glycol, carboxymethylcellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone, poly-i, 3- dioxolane, poly-i, 3, 6-trioxane, ethylene/maleic anhydride copolymer, polyaminoacids (either homopolymers or random copolymers), and dextran or poly(n-vinyl pyrrolidone)polyethylene glycol, propylene glycol

homopolymers, polypropylene oxide/ethylene oxide co-polymers, polyoxyethylated polyols (e.g., glycerol), polyvinyl alcohol, and mixtures thereof. Polyethylene glycol propionaldehyde may have advantages in manufacturing due to its stability in water. The polymer may be of any molecular weight, and may be branched or unbranched.

The number of polymers attached to the antibody may vary, and if more than one polymer is attached, they can be the same or different molecules. In general, the number and/ or type of polymers used for derivatization can be determined based on considerations including, but not limited to, the particular properties or functions of the antibody to be improved, whether the antibody derivative will be used in a therapy under defined conditions, etc.

In another embodiment, conjugates of an antibody and nonproteinaceous moiety that may be selectively heated by exposure to radiation are provided. In one embodiment, the nonproteinaceous moiety is a carbon nanotube (Kam et al., Proc. Natl. Acad. Sci. USA 102: 11600-11605 (2005)). The radiation maybe of any wavelength, and includes, but is not limited to, wavelengths that do not harm ordinary cells, but which heat the nonproteinaceous moiety to a temperature at which cells proximal to the antibody- nonproteinaceous moiety are killed.

Recombinant Methods and Compositions

Antibodies maybe produced using recombinant methods and compositions, e.g., as described in U.S. Patent No. 4,816,567. In one embodiment, isolated nucleic acid encoding an antibody described herein is provided. Such nucleic acid may encode an amino acid sequence comprising the VL and/or an amino acid sequence comprising the VH of the antibody (e.g., the light and/or heavy chains of the antibody). In a further embodiment, one or more vectors (e.g., expression vectors) comprising such nucleic acid are provided. In a further embodiment, a host cell comprising such nucleic acid is provided. In one such embodiment, a host cell comprises (e.g., has been transformed with): (1) a vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and an amino acid sequence comprising the VH of the antibody, or (2) a first vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and a second vector comprising a nucleic acid that encodes an amino acid sequence comprising the VH of the antibody. In one embodiment, the host cell is eukaryotic, e.g. a Chinese Hamster Ovary (CHO) cell or lymphoid cell (e.g., Yo, NSo Sp20 cell). In one embodiment, a method of making an antibody is provided, wherein the method comprises culturing a host cell comprising a nucleic acid encoding the antibody, as provided above, under conditions suitable for expression of the antibody, and optionally recovering the antibody from the host cell (or host cell culture medium).

For recombinant production of an antibody, nucleic acid encoding an antibody, e.g., as described above, is isolated and inserted into one or more vectors for further cloning and/or expression in a host cell. Such nucleic acid maybe readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the antibody).

Suitable host cells for cloning or expression of antibody-encoding vectors include prokaryotic or eukaryotic cells described herein. For example, antibodies may be produced in bacteria, in particular when glycosylation and Fc effector function are not needed. For expression of antibody fragments and polypeptides in bacteria, see, e.g., U.S. Patent Nos. 5,648,237, 5,789, 199, and 5,840,523. (See also Charlton, Methods in Molecular Biology, Vol. 248 (B.K.C. Lo, ed., Humana Press, Totowa, NT, 2003), pp. 245-254, describing expression of antibody fragments in E. coli.) After expression, the antibody may be isolated from the bacterial cell paste in a soluble fraction and can be further purified.

In addition to prokaryotes, eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for antibody-encoding vectors, including fungi and yeast strains whose glycosylation pathways have been“humanized,” resulting in the production of an antibody with a partially or fully human glycosylation pattern. See Gerngross, Nat. Biotech. 22: 1409-1414 (2004), and Li et al., Nat. Biotech. 24:210-215 (2006).

Suitable host cells for the expression of glycosylated antibody are also derived from multicellular organisms (invertebrates and vertebrates). Examples of invertebrate cells include plant and insect cells. Numerous baculoviral strains have been identified which maybe used in conjunction with insect cells, particularly for transfection of Spodoptera frugiperda cells.

Plant cell cultures can also be utilized as hosts. See, e.g., US Patent Nos. 5,959,177, 6,040,498, 6,420,548, 7,125,978, and 6,417,429 (describing PLANTIBODIES™ technology for producing antibodies in transgenic plants). Vertebrate cells may also be used as hosts. For example, mammalian cell lines that are adapted to grow in suspension may be useful. Other examples of useful mammalian host cell lines are monkey kidney CV 1 line transformed by SV40 (COS-7); human embryonic kidney line (293 or 293 cells as described, e.g., in Graham et al., J. Gen Virol. 36:59 (1977); baby hamster kidney cells (BHK); mouse Sertoli cells (TM4 cells as described, e.g., in Mather, Biol. Reprod. 23 1243-251 (1980); monkey kidney cells (CVi); African green monkey kidney cells (VERO-76); human cervical carcinoma cells (HELA); canine kidney cells (MDCK; buffalo rat liver cells (BRL 3A); human lung cells (W138); human liver cells (Hep G2); mouse mammary tumor (MMT 060562); TRI cells, as described, e.g., in Mather et al., Annals N.Y. Acad. Sci. 383 144-68 (1982); MRC 5 cells; and FS4 cells. Other useful mammalian host cell lines include Chinese hamster ovary (CHO) cells, including DHFR CHO cells (Urlaub et al, Proc. Natl. Acad. Sci. USA 77:4216 (1980)); and myeloma cell lines such as Yo, NSo and Sp2/o. For a review of certain mammalian host cell lines suitable for antibody production, see, e.g., Yazaki and Wu, Methods in Molecular Biology, Vol. 248 (B.K.C. Lo, ed., Humana Press, Totowa, NJ), pp. 255-268 (2003).

Administration & Dose

Compounds of formula I may be administered alone or in combination with one or another or with one or more pharmacologically active compounds which are different from the compounds of formula I.

Compounds of the invention may suitably be combined with various components to produce compositions of the invention. Suitably the compositions are combined with a pharmaceutically acceptable carrier or diluent to produce a pharmaceutical

composition (which may be for human or animal use). Suitable carriers and diluents include isotonic saline solutions, for example phosphate-buffered saline. Useful pharmaceutical compositions and methods for their preparation may be found in standard pharmaceutical texts. See, for example, Handbook for Pharmaceutical Additives, 3rd Edition (eds. M. Ash and I. Ash), 2007 (Synapse Information Resources, Inc., Endicott, New York, USA) and Remington: The Science and Practice of

Pharmacy, 21st Edition (ed. D. B. Troy) 2006 (Lippincott, Williams and Wilkins, Philadelphia, USA) which are incorporated herein by reference.

The compounds of the invention may be administered by any suitable route. Suitably the compounds of the invention will normally be administered orally or by any parenteral route, in the form of pharmaceutical preparations comprising the active ingredient, optionally in the form of a non-toxic organic, or inorganic, acid, or base, addition salt, in a pharmaceutically acceptable dosage form.

The compounds of the invention, their pharmaceutically acceptable salts, and pharmaceutically acceptable solvates of either entity can be administered alone but will generally be administered in admixture with a suitable pharmaceutical excipient diluent or carrier selected with regard to the intended route of administration and standard pharmaceutical practice.

For example, the compounds of the invention or salts or solvates thereof can be administered orally, buccally or sublingually in the form of tablets, capsules (including soft gel capsules), ovules, elixirs, solutions or suspensions, which may contain flavouring or colouring agents, for immediate-, delayed-, modified-, sustained-, controlled-release or pulsatile delivery applications. The compounds of the invention may also be administered via fast dispersing or fast dissolving dosages forms.

Such tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycollate,

croscarmellose sodium and certain complex silicates, and granulation binders such as polyvinylpyrrolidone, hydroxypropylmethyl cellulose (HPMC), hydroxypropylcellulose (HPC), sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, stearic acid, glyceryl behenate and talc may be included.

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

combinations thereof.

Modified release and pulsatile release dosage forms may contain excipients such as those detailed for immediate release dosage forms together with additional excipients that act as release rate modifiers, these being coated on and/or included in the body of the device. Release rate modifiers include, but are not exclusively limited to, hydroxypropylmethyl cellulose, methyl cellulose, sodium carboxymethylcellulose, ethyl cellulose, cellulose acetate, polyethylene oxide, Xanthan gum, Carbomer, ammonio methacrylate copolymer, hydrogenated castor oil, carnauba wax, paraffin wax, cellulose acetate phthalate, hydroxypropylmethyl cellulose phthalate, methacrylic acid copolymer and mixtures thereof. Modified release and pulsatile release dosage forms may contain one or a combination of release rate modifying excipients. Release rate modifying excipients maybe present both within the dosage form i.e. within the matrix, and/or on the dosage form i.e. upon the surface or coating.

Fast dispersing or dissolving dosage formulations (FDDFs) may contain the following ingredients: aspartame, acesulfame potassium, citric acid, croscarmellose sodium, crospovidone, diascorbic acid, ethyl acrylate, ethyl cellulose, gelatin,

hydroxypropylmethyl cellulose, magnesium stearate, mannitol, methyl methacrylate, mint flavouring, polyethylene glycol, fumed silica, silicon dioxide, sodium starch glycolate, sodium stearyl fumarate, sorbitol, xylitol.

The compounds of the invention can also be administered parenterally, for example, intravenously, intra-arterially, or they may be administered by infusion techniques.

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

Suitably formulation of the invention is optimised for the route of administration e.g. oral, intravenously, etc.

Administration may be in one dose, continuously or intermittently (e.g. in divided doses at appropriate intervals) during the course of treatment. Methods of determining the most effective means and dosage are well known to a skilled person and will vary with the formulation used for therapy, the purpose of the therapy, the target cell(s) being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and the dose regimen being selected by the treating physician, veterinarian, or clinician.

Depending upon the disorder and patient to be treated, as well as the route of administration, the compositions maybe administered at vaiying doses. For example, a typical dosage for an adult human may be too ng to 25 mg (suitably about 1 micro g to about 10 mg) per kg body weight of the subject per day.

Suitably guidance may be taken from studies in test animals when estimating an initial dose for human subjects. For example when a particular dose is identified for mice, suitably an initial test dose for humans may be approx. 0.5X to 2x the mg/Kg value given to mice.

Other Forms

Unless otherwise specified, included in the above are the well known ionic, salt, solvate, and protected forms of these substituents. For example, a reference to carboxylic acid (-COOH) also includes the anionic (carboxylate) form (-COO ), a salt or solvate thereof, as well as conventional protected forms. Similarly, a reference to an amino group includes the protonated form (-N + HIUR 2 ), a salt or solvate of the amino group, for example, a hydrochloride salt, as well as conventional protected forms of an amino group. Similarly, a reference to a hydroxyl group also includes the anionic form (-0 ), a salt or solvate thereof, as well as conventional protected forms.

Isomers. Salts and Solvates

Certain compounds may exist in one or more particular geometric, optical,

enantiomeric, diasteriomeric, epimeric, atropic, stereoisomeric, tautomeric, conformational, or anomeric forms, including but not limited to, cis- and trans-forms; E- and Z-forms; c-, t-, and r- forms; endo- and exo-forms; R-, S-, and meso-forms; D- and L-forms; d- and 1- forms; (+) and (-) forms; keto-, enol-, and enolate-forms; syn- and anti-forms; synclinal- and anticlinal-forms; alpha- and beta-forms; axial and equatorial forms; boat-, chair-, twist-, envelope-, and halfchair-forms; and

combinations thereof, hereinafter collectively referred to as“isomers” (or“isomeric forms”).

Note that, except as discussed below for tautomeric forms, specifically excluded from the term“isomers”, as used herein, are structural (or constitutional) isomers (i.e.

isomers which differ in the connections between atoms rather than merely by the position of atoms in space). For example, a reference to a methoxy group, -OCH 3 , is not to be construed as a reference to its structural isomer, a hydroxymethyl group, - CH 2 0H. A reference to a class of structures may well include structurally isomeric forms falling within that class (e.g. C -7 alkyl includes n-propyl and iso-propyl; butyl includes n-, iso-, sec-, and tert-butyl; methoxyphenyl includes ortho-, meta-, and para-methoxyphenyl).

The above exclusion does not apply to tautomeric forms, for example, keto-, enol-, and enolate-forms, as in, for example, the following tautomeric pairs: keto/enol, imine/enamine, amide/imino alcohol, amidine/amidine, nitroso/oxime,

thioketone/enethiol, N-nitroso/hyroxyazo, and nitro/aci-nitro.

Note that specifically included in the term“isomer” are compounds with one or more isotopic substitutions. For example, H maybe in any isotopic form, including Ή, 2 H (D), and 3 H (T); C may be in any isotopic form, including 12 C, 13 C, and 14 C; O may be in any isotopic form, including l6 0 and l8 0; and the like.

Unless otherwise specified, a reference to a particular compound includes all such isomeric forms, including (wholly or partially) racemic and other mixtures thereof.

Methods for the preparation (e.g. asymmetric synthesis) and separation (e.g. fractional crystallisation and chromatographic means) of such isomeric forms are either known in the art or are readily obtained by adapting the methods taught herein, or known methods, in a known manner.

Unless otherwise specified, a reference to a particular compound also includes ionic, salt, solvate, and protected forms of thereof, for example, as discussed below.

In some embodiments, the compound of formula (I) and salts and solvates thereof, comprises pharmaceutically acceptable salts of the compounds of formula (I).

Compounds of Formula (I), which include compounds specifically named above, may form pharmaceutically acceptable complexes, salts, solvates and hydrates. These salts include nontoxic acid addition salts (including di-acids) and base salts.

If the compound is cationic, or has a functional group which may be cationic (e.g. -NH 2 may be -NH 3 + ), then an acid addition salt may be formed with a suitable anion.

Examples of suitable inorganic anions include, but are not limited to, those derived from the following inorganic acids hydrochloric acid, nitric acid, nitrous acid, phosphoric acid, sulfuric acid, sulphurous acid, hydrobromic acid, hydroiodic acid, hydrofluoric acid, phosphoric acid and phosphorous acids. Examples of suitable organic anions include, but are not limited to, those derived from the following organic acids: 2-acetyoxybenzoic, acetic, ascorbic, aspartic, benzoic, camphorsulfonic, cinnamic, citric, edetic, ethanedisulfonic, ethanesulfonic, fumaric, glucheptonic, gluconic, glutamic, glycolic, hydroxymaleic, hydroxynaphthalene carboxylic, isethionic, lactic, lactobionic, lauric, maleic, malic, methanesulfonic, mucic, oleic, oxalic, palmitic, pamoic, pantothenic, phenylacetic, phenylsulfonic, propionic, pyruvic, salicylic, stearic, succinic, sulfanilic, tartaric, toluenesulfonic, and valeric. Examples of suitable polymeric organic anions include, but are not limited to, those derived from the following polymeric acids: tannic acid, carboxymethyl cellulose. Such salts include acetate, adipate, aspartate, benzoate, besylate, bicarbonate, carbonate, bisulfate, sulfate, borate, camsylate, citrate, cyclamate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate,

hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, mesylate, methylsulfonate, naphthylate, 2- napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate, hydrogen phosphate, dihydrogen phosphate, pyroglutamate, saccharate, stearate, succinate, tannate, tartrate, tosylate, trifluoroacetate and xinofoate salts.

For example, if the compound is anionic, or has a functional group which maybe anionic (e.g. -COOH may be -COO ), then a base salt may be formed with a suitable cation. Examples of suitable inorganic cations include, but are not limited to, metal cations, such as an alkali or alkaline earth metal cation, ammonium and substituted ammonium cations, as well as amines. Examples of suitable metal cations include sodium (Na + ) potassium (K + ), magnesium (Mg 2+ ), calcium (Ca 2+ ), zinc (Zn 2+ ), and aluminum (Al3 + ). Examples of suitable organic cations include, but are not limited to, ammonium ion (i.e. NH4 + ) and substituted ammonium ions (e.g. NH 3 R + , NH 2 R 2 + , NHR 3 + , NR^). Examples of some suitable substituted ammonium ions are those derived from: ethylamine, diethylamine, dicyclohexylamine, triethylamine, butylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine, benzylamine,

phenylbenzylamine, choline, meglumine, and tromethamine, as well as amino acids, such as lysine and arginine. An example of a common quaternary ammonium ion is N(CH 3 ) 4 + . Examples of suitable amines include arginine, N,N'-dibenzyl ethylene- diamine, chloroprocaine, choline, diethylamine, diethanolamine, dicyclohexylamine, ethylenediamine, glycine, lysine, N-methylglucamine, olamine, 2-amino-2- hydroxymethyl-propane-i,3-diol, and procaine. For a discussion of useful acid addition and base salts, see S. M. Berge et al., J. Pharm. Sci. (1977) 66:1-19; see also Stahl and Wermuth, Handbook of Pharmaceutical Salts: Properties, Selection, and Use (2011)

Pharmaceutically acceptable salts maybe prepared using various methods. For example, one may react a compound of Formula 1 with an appropriate acid or base to give the desired salt. One may also react a precursor of the compound of Formula 1 with an acid or base to remove an acid- or base-labile protecting group or to open a lactone or lactam group of the precursor. Additionally, one may convert a salt of the compound of Formula 1 to another salt through treatment with an appropriate acid or base or through contact with an ion exchange resin. Following reaction, one may then isolate the salt by filtration if it precipitates from solution, or by evaporation to recover the salt. The degree of ionization of the salt may vary from completely ionized to almost non-ionized.

It may be convenient or desirable to prepare, purify, and/or handle a corresponding solvate of the active compound. The term“solvate” describes a molecular complex comprising the compound and one or more pharmaceutically acceptable solvent molecules (e.g., EtOH). The term“hydrate” is a solvate in which the solvent is water. Pharmaceutically acceptable solvates include those in which the solvent may be isotopically substituted (e.g., D 2 0, acetone-d6, DMSO-d6).

A currently accepted classification system for solvates and hydrates of organic compounds is one that distinguishes between isolated site, channel, and metal-ion coordinated solvates and hydrates. See, e.g., K. R. Morris (H. G. Brittain ed.)

Polymorphism in Pharmaceutical Solids (1995). Isolated site solvates and hydrates are ones in which the solvent (e.g., water) molecules are isolated from direct contact with each other by intervening molecules of the organic compound. In channel solvates, the solvent molecules lie in lattice channels where they are next to other solvent molecules. In metal-ion coordinated solvates, the solvent molecules are bonded to the metal ion.

When the solvent or water is tightly bound, the complex will have a well-defined stoichiometry independent of humidity. When, however, the solvent or water is weakly bound, as in channel solvates and in hygroscopic compounds, the water or solvent content will depend on humidity and drying conditions.v In such cases, non- stoichiometry will typically be observed.

BRIEF DESCRIPTION OF THE DRAWINGS ΐ6q Embodiments of the present invention will now be described further, with reference to the accompanying drawings, in which:

Figure l shows fluorescently labelled DNA duplexes used in the FRET melting study to study the DNA interactivity of 15 and 16. The labels were fluorescein (F) and dabcyl (Q).

Figure 2 shows FRET Denaturation data for the two DNA sequences shown in Figure 1. Figure 3 shows FRET Denaturation data for 28 and 32.

Figure 4 shows FRET Denaturation data for 16.

Figure 5 shows DNA footprinting gels (upper panel) illustrating DNA sequence selectivity of compounds 15 (gel on the right) and 16 (gel on the left), and a schematic (lower panel) illustrating the binding pattern of both molecules as follows: 15 (blue hatched line, at the bottom of the lower panel represents 15 at 0.03 mM), 15 (blue solid line, one line up from the bottom of the lower panel represents 15 at 10.00 mM), and 16 (black solid line, two lines up from the bottom of the lower panel).

Figure 6 shows a DNA footprint (top panel) showing the interaction of 16 with a“D” DNA fragment (bottom panel). Ligand concentrations are shown at the top of the gel. Tracks labelled“GA” are markers for specific purines.

Figure 7 shows a graph illustrating transcription factor downregulation profile of 16. Figure 8 shows a size-exclusion chromatography (SEC) profile of a test IgGi antibody. This SEC profile shows peaks at 12.451 (area 438.736), 13.280 (area 883.843) and 15.256 (area 13267.9) showing 90.9% monomer.

Figure 9 shows a hydrophobic interaction chromatography (HIC) profile of a test IgGi antibody. This profile has peaks at 4.663 (area 2539.06), 6.011 (area 816.134), 6.748 (ara4389.i), 8.156 (area 1760.14), 8.733 (area 260.239) and 9.694 (area 44.5016).

Figure 10 shows free toxin linker traces of an IgGi-47 sample. No free toxin could be detected in the antibody-drug conjugate trace. The predominantly lower trace is 100 pmol N-Acetyl-Cysteine-47 (NAC-47), the predominantly higher trace is IgGi-47 after protein precipitation; the NAC-47 peak at 3.824 (with an area of 7.00543) shows residual protinaceous material.

Figure 11 shows general synthetic scheme 1. The reagents used for each step of general synthetic scheme 1 are as follows: i) K 2 C0 3 , methyl-4-bromobutyrate, DMF, r.t.; ii) KNO 3 , TFA, o - 5 °C; iii) KMn0 4 , acetone, H 2 0, reflux; iv) oxalyl chloride, (S)-piperidin- 2-ylmethanol, cat. DMF, Et 3 N, CH 2 Cl 2 , o °C - r.t.; v) H 2 , Pd/C, EtOH/EtOAc; vi) allyl chloroformate, pyridine, CH 2 Cl 2 , -10 °C - r.t.; vii) TEMPO, BAIB, CH 2 Cl 2 , r.t.; viii) pTSA, DHP, EtOAc, r.t.; ix) NaOH, dioxane, H 2 0, r.t.; x) RNH 2 , EDCI, DMAP. DMF, r.t. or RNH 2 , HATU, Et 3 N, CH 2 Cl 2 , r.t.; xi) Pd(PPh 3 ) 4 , pyrrolidine, CH 2 Cl 2 , r.t.; xii) ammonium formate, Pd/C, THF/H 2 0, 35 °C. Figure 12 shows general synthetic scheme 2. The reagents used for each step of general synthetic scheme 2 are as follows: i) Methyl 4-bromo-i-methyl-iff-pyrrole-2- carboxylate, Pd(PPh 3 ) 4 , K 2 C0 3 , MeCN, H 2 0, pW, too °C; ii) 4 M HC1,

dioxane/methanol (1:1), r.t., then 4-((tert-butoxycarbonyl)-amino)-i-methyl-iff- pyrrole-2-carboxylic acid, EDCI, DMAP, DMF, r.t.; iii) 4 M HC1, dioxane, r.t.

Figure 13 shows general synthetic scheme 3. The reagents used for each step of general synthetic scheme 3 are as follows: i) (S)-Pyrrol idi n-2-yl methanol , cat. DMF, oxalyl chloride, Et 3 N, CH 2 Cl 2 , -30 °C - r.t.; ii) H 2 , Pd/C, EtOH/EtOAc, r.t.; iii) allyl chloroformate, pyridine, CH 2 Cl 2 , -10 °C - r.t.; iv) TEMPO, BAIB, CH 2 Cl 2 , r.t.; v) pTSA, DHP, EtOAc, r.t.; vi) 1 M NaOH, dioxane, H 2 0, r.t.; vii) methyl 4-(4-(4-amino-i- methyl-iff-pyrrole-2-carboxamido)phenyl)-i-methyl-iif-pyrrol e-2-carboxylate hydrochloride, HATU, Et 3 N, DMF, r.t.; viii) Pd(PPh 3 ) 4 , pyrrolidine, CH 2 Cl 2 , r.t.; ix) ammonium formate, Pd/C, THF/H 2 0, 35 °C.

Figure 14 shows general synthetic scheme 4. The reagents used for each step of general synthetic scheme 4 are as follows: i) BnBr, K 2 C0 3 , MeOH, reflux; ii) KN0 3 , TFA, o °C; iii) HBr/AcOH, 85 °C; iv) TIPS-C1, imidazole, too °C; v) NaCl0 2 , H 2 0 2 , NaH 2 P0 4 , 45 °C; vi) (.S)-indolin-2-ylmethanol, HATU, Et 3 N, CH 2 Cl 2 ; vii) ammonium formate, Pd/C, THF/H 2 0, reflux; viii) Alloc-Cl, pyridine, -10 °C, CH 2 Cl 2 ; ix) TEMPO, PIDA, CH 2 Cl 2 , r.t.; x) DHP, pTSA, THF; xi) TBAF, THF; xii) methyl 4-bromobutanoate, K 2 C0 3 , DMF; xiii) LiOH, THF/Me0H/H 2 0 (3/1/1); xiv) RNH 2 , HATU, Et 3 N, CH 2 Cl 2 , r.t.; xv) Pd(PPh 3 ) 4 , pyrrolidine, CH 2 Cl 2 , r.t.; xvi) ammonium formate, Pd/C, THF/H 2 0, reflux. Figure 15 shows general synthetic scheme 5. The reagents used for each step of general synthetic scheme 5 are as follows: i) Methyl (S)-i,2,3,4-tetrahydroisoquinoline-3- carboxylate, (C0Cl) 2 , DMF, Et 3 N, CH 2 Cl 2 , -10 °C; ii) LiBH 4 , THF, o °C; iii) FeCl 3 .6H 2 0, NH 2 NH 2 .H 2 0, activated charcoal, MeOH, reflux; iv) allyl chloroformate, pyridine, CH 2 Cl 2 , -10 °C; v) TEMPO, PIDA, CH 2 Cl 2 , r.t.; vi) BCl 3 , CH 2 Cl 2 , r.t., then MeOH, microwave 50 °C; vii) methyl 4-bromobutanoate, K 2 C0 3 , DMF; viii) LiOH,

THF/Me0H/H 2 0 (3/1/1); ix) RNH 2 , HATU, Et 3 N, CH 2 Cl 2 , r.t.; x) Pd(PPh 3 ) 4 , pyrrolidine, CH 2 Cl 2 , r.t.; xi) ammonium formate, Pd/C, THF/H 2 0, reflux.

Figure 16A-D shows the antimicrobial activity in e.coli K12 of free payloads of compounds 16 (16A), 45 (16B) and 73 (16C) as well as antibiotic kanamycin (16D). Figure 17A-D shows the antimicrobial activity in EMRSA 16 of free payloads of compounds 16 (17A), 45 (17B) and 73 (17C) as well as antibiotic vancomycin (16D). Figure 18A-D shows the antimicrobial activity Klebsiella pneumoniae KP4631 of free payloads of compounds 16 (18A), 45 (18B) and 73 (18C) as well as antibiotic

Gentamicin (16D).

102 EXAMPLES

CHEMISTRY

General remarks

Reagents were purchased from Standard commercial suppliers. Solvents were purchased from VWR (UK). Anhydrous reactions were carried out in pre-oven-dried glassware under an inert atmosphere of argon. Anhydrous solvents were used as purchased without further drying. Thin Layer Chromatography (TLC) was performed on silica gel aluminium plates (Merck 60, F254), and flash column chromatography was carried out either manually, using silica gel (Merck 9385, 230-400 mesh ASTM, 40-63 mM) (whilst monitoring by thin layer chromatography: UV (254 nm) and an aqueous alkaline solution of potassium permanganate as stain), or using a Biotage Isolera 1 Chromatography System coupled to Dalton mass spectrometer. All NMR spectra were obtained at room temperature using a Varian Mercury Vx, Agilent 400 Hz

spectrometer, for which chemical shifts are expressed in ppm relative to the solvent and coupling constants are expressed in Hz. Microwave reactions were carried out on a Biotage Initiator microwave synthesis reactor. HRMS was performed on a Thermo Scientific-Exactive HCD Orbitrap Mass Spectrometer. Yields refer to isolated material (homogeneous by TLC or NMR) unless otherwise stated and names are assigned according to IUPAC nomenclature. All Liquid Chromatography Mass Spectroscopy (LCMS) analysis was performed on a Waters Alliance 2695 with water (A) and acetonitrile (B) comprising the mobile phases. Formic acid (0.1%) was added to both acetonitrile and water to ensure acidic conditions throughout the analysis. Function type: Diode array (535 scans). Column type: Monolithic C18 50 X 4.60 mm. Mass spectrometry data were collected using a Waters Micromass ZQ instrument coupled to a Waters 2695 HPLC with a Waters 2996 PDA. Waters Micromass ZQ parameters used were: Capillary (kV), 3.38; Cone (V), 35; Extractor (V), 3.0; Source temperature (°C), too; De-solvation Temperature (°C), 200; Cone flow rate (L/h), 50; De-solvation flow rate (L/h), 250. LCMS gradient conditions are described below (Methods A & B). Ultra- Performance Liquid Chromatography Mass Spectroscopy (UPLC-MS) analysis was performed on a Waters Acquity H-class UPLC with water (A) and acetonitrile (B) comprising the mobile phases. Trifluoracetic acid (0.1%) was added to both acetonitrile and water to ensure acidic conditions throughout the analysis. Function type: Photo Diode array (502.93 n). Column type: Acquity UPLC BEH C18 1.7pm 2.1 X 50 mm.

Mass spectrometry data were collected using a Waters SQ Detector 2 coupled to a Waters Acquity H Class UPLC with ACQ-PDA. Waters SQ Detector 2 parameters used were: Capillary (kV), 3.00; Cone (V), 30; De-solvation Temperature (°C), 600; Cone flow rate (L/h), 50; De-solvation flow rate (L/h), 600. UPLC-MS gradient conditions are described below (Methods 1, 2 & 3).

Method A (10 min): from 95% A/5% B to 50% B over 3 min. Then from 50% B to 80% B over 2 min. Then from 80% B to 95% B over 1.5 min and held constant for 1.5 min. This was then reduced to 5% B over 0.2 min and maintained to 5% B for 1.8 min. The flow rate was 0.5 mL/min, 200 pL was split via a zero dead volume T piece which passed into the mass spectrometer. The wavelength range of the UV detector was 220- 400 nm.

Method B (5 min): from 95% A/5% B to 90% B over 3 min. Then from 90% B to 95%

B over 0.5 min and held constant for 1 min. This was then reduced to 5% B over 0.5 min. The flow rate was 1.0 mL/ min, too pL was split via a zero dead volume T piece which passed into the mass spectrometer. The wavelength range of the UV detector was 220-500 nm.

Method 1 (7 min): from 90% A/ 10% B to 50% B over 1.5 min. Then from 50% B to 75% B over 1.5 min. Then from 75% B to 90% B over 1 min. This was then reduced to 10% B over 1 min. The flow rate was 0.6 mL/ min, 5 pL was split via a zero-dead volume T piece which passed into the mass spectrometer. The wavelength range of the UV detector was 230-280 nm.

Method 2 (12 min): from 90% A/ 10% B to 50% B over 3 min. Then from 50% B to 65% B over 1.5 min. Then from 65% B to 75% B over 1.5 min. Then from 75% B to 90%B over 4 2 min. This was then reduced to 10% B over 2 min. The flow rate was

0.6 mL/min, 5 pL was split via a zero-dead volume T piece which passed into the mass spectrometer. The wavelength range of the UV detector was 230-280 nm.

Method 3 (12 min): from 90% A/ 10% B to 50% B over 3 min. Then from 50% B to 59% B over 2 min. Then from 59% B to 60% B over 0.5 min. Then from 60% B to 65%B over 0.5 min. Then from 65%B to 90%B over 2 min. This was then reduced to 10% B over 2 min. The flow rate was 0.6 mL/min, 5 pL was split via a zero-dead volume T piece which passed into the mass spectrometer. The wavelength range of the UV detector was 230-280 nm.

Evidence of no presence of imine after reduction step

Extra care was taken during the transfer hydrogenation step in order to make sure there was no trace of imine precursors left in non-alkylating final compounds. To do so, very thorough and meticulous analysis of final secondary amines was carried out. A wide range of different analytical techniques and tools was used in order to achieve this. First, the mass ion for parent imine and its related carbinolamine were looked for in each LCMS chromatogram, to make sure no residual imine was present.

Then, the same principle was applied in UPLC analyses.

Finally, when using proton NMR spectroscopy, the disappearance of the C-11 imine proton (doublet at around 8 ppm) was also used to show that no residual imines were present in final secondary amines.

CHEMICAL SYNTHESIS

Methyl 4-(4-formyl-2-methoxyphenoxy)butanoate (l)

A mixture of vanillin (20.0 g, 131 mmol), methyl 4-bromobutanoate (17.5 mL, 139 mmol) and potassium carbonate (27.2 g, 197 mmol) in N,iV-dimethylformamide (too mL) was stirred at room temperature for 18 h. The reaction mixture was diluted with water (500 mL) and the title compound (30.2 g, 91%) was obtained by filtration as a white solid. The product was carried through to the next step without any further purification. Ή NMR (400 MHz, CDCl 3 ) <59.84 (s, lH), 7.46-7.37 (m, 2H), 6.98 (d, J=8.2 HZ, lH), 4.16 (t, J=6.3 HZ, 2H), 3.91 (s, 3H), 3.69 (s, 3H), 2.56 (t, J=7- 2 Hz, 2H), 2.20 (quin, J= 6.7 Hz, 2H); «C NMR (too MHz, CDCl 3 ) d 190.9, 173.4, 153-8, 149-9, 130.1, 126.8, 111.6, 109.2, 67.8, 56.0, 51.7, 30.3, 24.2; MS m/z (EIMS) = 271.9 (M+Na) + ; MS (ES+): m/z = 253 (M+H) + ; LCMS (Method A): £R = 6.48 min.

Methyl 4-(4-formyl-2-methoxy-5-nitrophenoxy)butanoate (2)

To a stirring solution of potassium nitrate (10.0 g, 98.9 mmol) in TFA (50 mL) at o °C was added dropwise a solution of methyl 4-(4-formyl-2-methoxyphenoxy)butanoate (1) (20.0 g, 79.2 mmol) in TFA (50 mL). The reaction mixture was stirred at room temperature for 1 h. It was then concentrated in vacuo and diluted with ethyl acetate (400 mL). The organic layer was washed with brine (3 x 100 mL) and a saturated aqueous solution of sodium hydrogen carbonate (2 x 80 mL), dried over sodium sulfate, filtered and concentrated to give the title compound (23.5 g, 100%) as a yellow solid. The product was carried through to the next step without any further

purification. Ή NMR (400 MHz, CDCl 3 ) d 10.42 (s, lH), 7.60 (s, lH), 7.39 (s, lH), 4.21 (t, J= 6.3 Hz, 2H), 3.98 (s, 3H), 3.70 (s, 3H), 2.61-2.53 (m, 2H), 2.22 (quin, J= 6.6 Hz, 2H); «C NMR (100 MHz, CDCl 3 ) d 187.8, 173.2, 153.5, 151-7, 143-8, 125.5, 109-9, iob.i, 68.6, 56.6, 51.8, 30.2, 24.1; MS m/z (EIMS) = 296.1 (M-H)-; MS (ES+): m/z = 298 (M+H) + ; LCMS (Method A): £ R = 6.97 min. 5-Methoxy-4-(4-methoxy-4-oxobutoxy)-2-nitrobenzoic acid (3)

To a solution of methyl 4-(4-formyl-2-methoxy-5-nitrophenoxy)butanoate (2) (23.0 g, 77.4 mmol) in acetone (600 mL) was quickly added a hot (70 °C) solution of potassium permanganate (46.0 g, 291 mmol) in water (400 mL). The reaction mixture was stirred at 70 °C for 3 h. The reaction mixture was cooled to room temperature and passed through celite. The cake of celite was washed with hot water (200 mL). A solution of sodium bisulfite in hydrochloric acid (lM, 200 mL) was added to the filtrate which was extracted with dichloromethane (2 x 400 mL). The organic layer was dried over sodium sulfate, filtered and concentrated. The resulting residue was purified by column chromatography (silica), eluting with methanol/ dichloromethane (from 0% to 50%), to give the title compound (17.0 g, 70%) as a pale yellow solid. Ή NMR (400 MHz,

MeOD) 57-47 (s, lH), 7 2 5 (s, lH), 4-13 (t, J= 6-2 Hz, 2H), 3-94 (s, 3H), 3-68 (s, 3H), 2.54 (t, J=7.2 Hz, 2H), 2.17-2.06 (m, 2H); «C NMR (100 MHz, MeOD) d 175-3, i68.6, 153.8, 151.3, 143.1, 122.8, 112.4, 109-2, 69.6, 57.0, 52.2, 31.2, 25.5; MS m/z (EIMS) = 311.9 (M-H)-; MS (ES+): m/z = 314 (M+H) + ; LCMS (Method A): t R = 6.22 min.

Methyl (,S)-4-(4-(2-(hydroxymethyl)piperidine-i-carbonyl)-2-methoxy -5- nitrophenoxy)butanoate (4)

A mixture of 5-methoxy-4-(4-methoxy-4-oxobutoxy)-2-nitrobenzoic acid (3) (8.0 g, 25.5 mmol), oxalyl chloride (6.6 mL, 77.0 mmol) and anhydrous A/N-dimethl- formamide (2 drops) in anhydrous dichloromethane (100 mL) was stirred at room temperature for 1 h. Anhydrous toluene (20 mL) was added to the reaction mixture which was then concentrated in vacuo. A solution of the resulting residue in anhydrous dichloromethane (10 mL) was added dropwise to a solution of (S)-piperidin-2- ylmethanol (3.8 g, 33.4 mmol) and triethylamine (10.7 mL, 77.0 mmol) in anhydrous dichloromethane (90 mL) at - 10 °C. The reaction mixture was stirred at room temperature for 2 h and then washed with hydrochloric acid (1 M, 50 mL) and a saturated aqueous solution of sodium chloride (50 mL), dried over sodium sulfate, filtered and concentrated. The resulting residue was purified by column

chromatography (silica), eluting with methanol/ dichloromethane (from 0% to 5%), to give the title compound (9.2 g, 73%) as a yellow oil. Ή NMR (400 MHz, CDCl 3 ) d 7.68- 7.64 (m, lH), 6.77-6.70 (m, lH), 4.16-4.07 (m, 3H), 3-93-3-89 (m, 3H), 3-83 (s, lH),

3.67 (s, 3H), 3-15 (d, « 7=1.4 Hz, lH), 3.11 (s, lH), 2.78 (s, lH), 2.56-2.50 (m, 3H), 2.21- 2.12 (m, 4H), 1.74-1-55 (m, 4H); «C NMR (100 MHz, CDCl 3 ) d 173-3, i68.i, 154.6, 148.2, 137.4, 127-6, 111.4, 108.3, 68.3, 60.6, 56.7, 53-5, 51-7, 43-3, 38.0, 34-9, 30-3, 24.1, 19.7; MS m/z (EIMS) = 411.0 (M+H) + ; LCMS (Method A): t R = 6.28 min.

Methyl (,S)-4-(5-amino-4-(2-(hydroxymethyl)piperidine-i-carbonyl)-2 - methoxyphenoxy)butanoate (5)

To a solution of methyl (S')-4-(4-(2-(hydroxymethyl)piperidine-i-carbonyl)-2-methoxy - 5-nitrophenoxy)butanoate (4) (9.2 g, 22.4 mmol) in ethanol (40 mL) and ethyl acetate (10 mL) was added palladium on activated charcoal (10% wt. basis) (920 mg). The reaction mixture was hydrogenated at 35 psi for 3 h in a Parr apparatus. The reaction mixture was filtered through celite and the resulting cake was washed with ethyl acetate. The filtrate was concentrated in vacuo to give the title compound (9.0 g, 90%) as a pink solid. The product was carried through to the next step without any further purification. Ή NMR (400 MHz, CDCl 3 ) d 6.69 (s, lH), 6.27-6.18 (m, lH), 4.03-3.94 (m, 3H), 3-94-3-82 (m, 3H), 3·8i-3·76 (m, lH), 3.74 (s, 3H), 3·73-3·68 (m, lH), 3.67- 3.65 (m, 3H), 3.56 (d, J=4.8 HZ, lH), 3.03 (s, lH), 2.51 (t, J=7- 2 Hz, 2H), 2.11 (quin,

J= 6.7 Hz, 2H), 1.68-1.59 (m, 4H), 1.55-1-40 (m, 2H); «C NMR (lOO MHz, CDCl 3 ) d 173.6, 171.2, 150.3, 141.8, 141.1, 113.2, 112.3, 102.4, 67.5, 6o.8, 60.4, 56.8, 51.6, 30.4,

25.8, 24.3, 21.0, 19.9, 14.2; MS m/z (EIMS) = 381.0 (M+H) + ; LCMS (Method A): t R = 5.52 min.

Methyl (S)-4-(5-(((allyloxy)carbonyl)amino)-4-(2-(hydroxyl- methyl)piperidine-i-carbonyl)-2-methoxyphenoxy)butanoate (6)

To a solution of methyl (S')-4-(5-amino-4-(2-(hydroxymethyl)piperidine-i-carbonyl)-2 - methoxyphenoxy)butanoate (5) (9.0 g, 23.7 mmol) and pyridine (4.4 mL, 54.4 mmol) in anhydrous dichloromethane (100 mL) at - 10 °C was added dropwise a solution of allylchloroformate (2.6 mL, 24.8 mmol) in anhydrous dichloromethane (20 mL). The reaction mixture was stirred at room temperature for 30 min. The reaction mixture was sequentially washed with a saturated aqueous solution of copper (II) sulfate (80 mL), water (80 mL) and a saturated aqueous solution of sodium hydrogen carbonate (80 mL). The organic layer was dried over sodium sulfate, filtered and concentrated. The resulting residue (2.0 g out of the 11.0 g crude) was purified by column chromatography (silica), eluting with methanol/ dichloromethane (from 0% to 1%), to give the title compound (930 mg, 47% based on the amount purified) as a yellow oil. Ή NMR (400 MHz, CDCI3) d 8.30 (br s, lH), 7.63 (br s, lH), 6.76 (br s, lH), 5.92 (ddt, =i7-2, 10.6, 5.4, 5.4 Hz, lH), 5·37-5·28 (m, lH), 5.20 (dq, J= 10.4, 1.3 Hz, lH), 4.65-4.56 (m, 2H), 4.06 (t, J= 6.2 Hz, 2H), 3.94-3·82 (m, lH), 3.79 (s, 3H), 3-66 (s, 3H), 3.62-3.54 (m, lH), 3.40 (br s, lH), 3.10-2.88 (m, lH), 2.52 (t, J=7A Hz, 2H), 2.22-2.04 (m, 3H), 1.64 (br s, 4H), 1.56-1.31 (m, 2H); 3 C NMR (100 MHz, CDCl 3 ) d 173-5, 170.6, 153.9, 149-7, 144-8, 132.6, 130.1, 117.6, 116.9, uo.8, 107.1, 106.0, 67.7, 65.6, 60.7, 56.3, 53.5, 51.6, 43.1,

30.5, 25.7, 24.4, 19.7; MS m/z (EIMS) = 465.1 (M+H) + ; LCMS (Method A): f R = 6.47 min.

Allyl (6aS')-6-hydroxy-2-methoxy-3-(4-methoxy-4-oxobutoxy)-i2-oxo- 6,6a,7,8,9,io-hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine-5 (i2Ji)- carboxylate (7)

To a solution of methyl (S)-4-(5-(((allyloxy)carbonyl)amino)-4-(2-(hydroxymethyl)- piperidine-i-carbonyl)-2-methoxyphenoxy)butanoate (6) (930 mg, 2.0 mmol) in dichloromethane (45 mL) was added TEMPO (32 mg, 0.20 mmol) and (diacetoxyiodo)- benzene (773 mg, 2.4 mmol). The reaction mixture was stirred at room temperature for 16 h, and was then sequentially washed with a saturated aqueous solution of sodium metabisulfite (20 mL), a saturated aqueous solution of sodium hydrogen carbonate (20 mL), water (20 mL) and brine (20 mL). The organic layer was then dried over sodium sulfate, filtered and concentrated. The resulting residue was purified by column chromatography (silica), eluting with methanol/ dichloromethane (from 0% to 5%), to give the title compound (825 mg, 89%) as a cream solid. MS m/z (EIMS) = 462.9 (M+H) + ; LCMS (Method A): f R = 6.30 min. Allyl (6aS , )-2-methoxy-3-(4-methoxy-4-oxobutoxy)-i2-oxo-6-((tetra hydro- 2JT-pyran-2-yl)oxy)-6,6a,7,8,9,io-hexahydrobenzo[e]pyrido-[i ,2- «][i,4]diazepine-5(i2Ji)-carboxylate (8)

A mixture of allyl (6aS')-6-hydroxy-2-methoxy-3-(4-methoxy-4-oxobutoxy)-i2-oxo- 6,6a,7,8,9,io-hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine-5 (i2ff)-carboxylate (7) (825 mg, 1.8 mmol), 3,4-dihydro-2ff-pyran (1.7 mL, 18.2 mmol) and -toluenesulfonic acid monohydrate (8.5 mg, 1% w/w) in ethyl acetate (12 mL) was stirred at room temperature for 16 h. The reaction mixture was then diluted with ethyl acetate (50 mL) and washed with a saturated aqueous solution of sodium hydrogen carbonate (20 mL) and brine (30 mL). The organic layer was dried over sodium sulfate, filtered and concentrated. The resulting residue was purified by column chromatography (silica), eluting with methanol/ dichloro methane (from 0% to 2%), to give the title compound (820 mg, 84%) as a cream solid. MS m/z (EIMS) = 546.7 (M+H) + ; LCMS (Method A): £R = 7.70 min.

4-(((6aS , )-5-((Allyloxy)carbonyl)-2-methoxy-i2-oxo-6-((tetra-hy dro-2JT- pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydrobenzo[e]pyrido[i ,2- «] [1, 4] diazepin-3-yl)oxy) butanoic acid (9)

To a solution of allyl (6aS)-2-methoxy-3-(4-methoxy-4-oxobutoxy)-i2-oxo-6- ((tetrahydro-2if-pyran-2-yl)oxy)-6,6a,7,8,9,io-hexahydrobenz o[e]pyrido[i,2- <3][i,4]diazepine-5(i2ff)-carboxylate (8) (770 mg, 1.4 mmol) in 1,4-dioxane (10 mL) was added a 0.5 M aqueous solution of sodium hydroxide (10 mL, 5.0 mmol). The reaction mixture was stirred at room temperature for 2 h and was then concentrated in vacuo, after which water (20 mL) was added and the aqueous layer was acidified to pH = 1 with a 1 M citric acid solution (5 mL). The aqueous layer was then extracted with ethyl acetate (2 x 50 mL). The combined organic extracts were then washed with brine (50 mL), dried over sodium sulfate, filtered and concentrated to give the title compound (700 mg, 93%) as a yellow oil. The product was carried through to the next step without any further purification. MS m/z (EIMS) = 532.9 (M+H) + ; LCMS

(Method A): £R = 6.98 min. Methyl 4-(4-aminophenyl)-i-methyl-iii-pyrrole-2-carboxylate (io)

A mixture of methyl 4-bromo-i-methyl-iff-pyrrole-2-carboxylate (750 mg, 3.44 mmol), 4-(4,4,5,5-tetramethyl-i,3,2-dioxaborolan-2-yl)aniline (905 mg, 4.13 mmol) and potassium carbonate (1.43 g, 10.3 mmol) in toluene/ethanol/water (9:3:1) (13 mL total) was degassed with nitrogen for 5 mins. Tetrakis(triphenylphosphine)palladium(o) (230 mg, 6 mol%) was then charged and the reaction mixture was irradiated with

microwaves at 100 °C for 15 mins. Water (10 mL) was then added to the reaction mixture, which was extracted with ethyl acetate (3 x 40 mL). The combined organic extracts were then dried over sodium sulfate, filtered and concentrated. The resulting residue was purified by column chromatography (silica), eluting with ethyl

acetate/ hexanes (from 0% to 50%), to give the title compound (145 mg, 18%) as a yellow solid. MS m/z (EIMS) = 230.9 (M+H) + ; LCMS (Method A): £R = 5.17 min.

Allyl (6aS)-2-methoxy-3-(4-((5-(methoxycarbonyl)-i-methyl-iH-pyrro l-3- yl)amino)-4-oxobutoxy)-i2-oxo-6-((tetrahydro-2H-pyran-2-yl)o xy)- 6,6a,7,8,9,io-hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine-5 (i2H)- carboxylate (11)

A solution of 4-(((6aS)-5-((allyloxy)carbonyl)-2-methoxy-i2-oxo-6-((tetrah ydro-2/7- pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydrobenzo[e]pyrido[i ,2-a][i,4]diazepin-3- yl)oxy)butanoic acid (9) (150 mg, 0.64 mmol) in N, L G - d i m e t h y 1 f 0 r m a m i d e (4 mL) was charged with i-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (98 mg, 0.51 mmol) and 4-(dimethylamino)pyridine (79 mg, 0.64 mmol). The reaction mixture was stirred at room temperature for 30 min. Methyl 4-ami no-i-methyl-i/f-pyrrole-2- carboxylate hydrochloride (49 mg, 0.26 mmol) was then added and the resulting mixture was stirred at room temperature for 16 h. This was then poured into ice-water (40 mL) and extracted with ethyl acetate (3 x 100 mL). The combined organic extracts were sequentially washed with 1 M citric acid (60 mL), a saturated aqueous solution of sodium hydrogen carbonate (70 mL), water (70 mL) and brine (70 mL). The organic layer was then dried over sodium sulfate, filtered and concentrated in vacuo to give the title compound (150 mg, 88%) as a yellow oil. The product was carried through to the next step without any further purification. MS m/z (EIMS) = 668.8 (M+H) + ; LCMS (Method A): t R = 7.42 min.

4-(4-(((6aS')-5-((Allyloxy)carbonyl)-2-methoxy-i2-oxo-6-( (tetrahydro-2Ji- pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydrobenzo[e]-pyrido[ i,2-a][i,4]- diazepin-3-yl)oxy)butanamido)-i-methyl-iii-pyrrole-2-carboxy lic acid (12)

To a solution of allyl (6aS)-2-methoxy-3-(4-((5-(methoxycarbonyl)-i-methyl-i//- pyrrol-3-yl)amino)-4-oxobutoxy)-i2-oxo-6-((tetrahydro-2ff-py ran-2-yl)oxy)- 6,6a,7,8,9,io-hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine-5 (i2ff)-carboxylate (11) (150 mg, 0.22 mmol) in 1,4-dioxane (5 mL) was added a 0.5 M aqueous solution of sodium hydroxide (5 mL, 5.0 mmol). The reaction mixture was stirred at room temperature for 2 h and was then concentrated in vacuo, after which water (20 mL) was added and the aqueous layer was acidified to pH = 1 with a 1 M citric acid solution (10 mL). The aqueous layer was then extracted with ethyl acetate (2 x 50 mL). The combined organic extracts were then washed with brine (50 mL), dried over sodium sulfate, filtered and concentrated in vacuo to give the title compound (140 mg, 90%) as a beige solid. The product was carried through to the next step without any further purification. MS m/z (EIMS) = 677.0 (M+Na) + ; LCMS (Method A): t R = 6.92 min.

Allyl (6S , ,6aS')-2-methoxy-3-(4-((5-((4-(5-(methoxycarbonyl)-i-m ethyl-iJT- pyrrol-3-yl)phenyl)carbamoyl)-i-methyl-iiT-pyrrol-3-yl)amino )-4- oxobutoxy)-i2-oxo-6-((tetrahydro-2JT-pyran-2-yl)oxy)-6,6a,7, 8,9,io- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine-5(i2H)-carboxyl ate (13)

A solution of 4-(4-(((6aS')-5-((allyloxy)carbonyl)-2-methoxy-i2-oxo-6-((te trahydro-2ff- pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydrobenzo[e]pyrido[i ,2-a][i,4]diazepin-3- yl)oxy)butanamido)-i-methyl-iff-pyrrole-2-carboxylic acid (12) (140 mg, 0.21 mmol) in /V,/V- dimethyl formamide (4 mL) was charged with i-(3-dimethylaminopropyl)-3- ethylcarbodiimide hydrochloride (74 mg, 0.39 mmol) and 4-(dimethylamino)pyridine (59 mg, 0.48 mmol). The reaction mixture was stirred at room temperature for 30 min. Methyl 4-(4-aminophenyl)-i-methyl-i//-pyrrole-2-carboxylate (10) (45 mg, 0.19 mmol) was then added and the resulting mixture was stirred at room temperature for 16 h. This was then poured into ice-water (40 mL) and extracted with ethyl acetate (3 x too mL). The combined organic extracts were sequentially washed with 1 M citric acid (60 mL), a saturated aqueous solution of sodium hydrogen carbonate (70 mL), water (70 mL) and brine (70 mL). The organic layer was then dried over sodium sulfate, filtered and concentrated. The resulting residue was then purified by column chromatography (silica), eluting with acetone/dichloromethane (0% to 50%), to give the title compound (160 mg, 95%) as a yellow solid. MS m/z (EIMS) = 867.0 (M+H) + ; LCMS (Method A): tn = 8.10 min.

Allyl (6aS)-3-(4-((5-((4-(5-((4-aminophenyl)carbamoyl)-i-methyl-ii i- pyrrol-3-yl)phenyl)carbamoyl)-i-methyl-iiT-pyrrol-3-yl)amino )-4- oxobutoxy)-2-methoxy-i2-oxo-6-((tetrahydro-2JT-pyran-2-yl)ox y)-

6,6a,7,8,9,io-hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepin e-5(i2Ji)- carboxylate

A solution of 44-(4-(4-(4-(((6aS')-5-((allyloxy)carbonyl)-2-methoxy-i2-oxo -6-

((tetrahydro-2if-pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octah ydrobenzo[e]pyrido[i,2- <3][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iff-pyrrol e-2-carboxamido)phenyl)-i- methyl-iff-pyrrole-2-carboxylic acid (46) (320 mg, 0.375 mmol) in anhydrous dichloromethane (1.5 mL) was charged with A/-[(dimethylamino)-iff-i,2,3-triazolo- [4,5-b]pyridin-i-ylmethylene]-V-methylmethanaminiiim hexafluorophosphate A-oxide (150 mg, 0.395 mmol) and anhydrous triethylamine (220 pL, 1.58 mmol). The reaction mixture was stirred at room temperature for 30 min. Benzene-i, 4-diamine (41 mg, 0.38 mmol) was then added and the resulting mixture was stirred at room temperature for 16 h. The reaction mixture was quenched with a saturated aqueous solution of sodium hydrogen carbonate (20 ruL) and extracted with dichloromethane (2 x 50 mL). The combined organic extracts were washed with water containing a few drops of acetic acid (30 mL). The organic layer was then dried over sodium sulfate, filtered and concentrated in vacuo. The resulting residue was then purified by column

chromatography (silica), eluting with methanol/dichloro methane (from 0% to 10%), to give the title compound (250 mg, 71%) as a cream solid. MS (ES+): m/z = 944 (M+H) + ; LCMS (Method B): t R = 3.45 min.

(S)-/V-(4-aminophenyl)-4-(4-(4-(4-((2-methoxy-i2-oxo-6a,7 , 8,9,10,12- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butan-a mido)-i- methyl-iJT-pyrrole-2-carboxamido)phenyl)-i-methyl-iJT-pyrrol e-2- carboxamid

To a solution of allyl (6aS)-3-(4-((5-((4-(5-((4-ami nophenyl )carbamoyl)-i-methyl-i/7- pyrrol-3-yl)phenyl)carbamoyl)-i-methyl-iif-pyrrol-3-yl)amino )-4-oxobutoxy)-2- methoxy-i2-oxo-6-((tetrahydro-2if-pyran-2-yl)oxy)-6, 6a, 7,8,9, io-hexahydrobenzo[e]- pyrido[i,2-a][i,4]diazepine-5(i2if)-carboxylate (14) (250 mg, 0.265 mmol) in dichloromethane (3 mL) was added tetrakis(triphenylphosphine)palladium(o) (15 mg, 5 mol%), triphenylphosphine (17 mg, 25 mol%) and pyrrolidine (26 pL, 0.32 mmol). The reaction mixture was stirred at room temperature for 16 h. The reaction mixture was subjected to high vacuum for 30 min until excess pyrrolidine was thoroughly removed. The resulting residue was then purified by column chromatography (silica), eluting with acetone/dichloromethane (from 0% to 100%) followed by methanol/ acetone (from 0% to 100%), to give the title compound (118 mg, 59%) as a yellow solid. Ή NMR (DMSO-de, 400 MHz) 59.88-9.96 (m, lH), 9.81 (s, 2H), 9.50 (s, lH), 8.32 (br s, 2H), 8.00 (d, J= 5.7 Hz, lH), 7·67-7·73 (m, 2H), 7.48 (d, J=8.6 Hz, 2H), 7.39 (d, J=i.8 Hz, lH), 7·3ΐ-7·35 (m, 2H), 7.30 (d, J=i.6 Hz, lH), 7.27 (s, lH), 7.22 (d, J= 1.5 Hz, lH), 6.96 (d, J=i.6 Hz, lH), 6.80 (s, lH), 6.51-6.55 (m, 2H), 4.09-4.17 (m, lH), 3-99-4-05 (m, lH), 3-90-3-97 (m, lH), 3.88 (s, 3H), 3.83 (s, 3H), 3.82 (s, 3H), 3.68-3.72 (m, lH), 3.05-3.16 (m, 2H), 2.44 (t, J=7.3 Hz, 2H), 2.02-2.07 (m, 2H), 1.81-1.91 (m, lH), 1.68- 1.78 (m, 2H), 1.56 (d, J= 4.9 Hz, 2H); «C NMR (DMSO-d 6 , lOO MHz) S 168.8, 166.3, 164.7, 159-5, 159-2, 150.2, 147.1, 144.7, 139-8, 137-0, 129.6, 128.2, 126.1, 124.6, 124.3, 122.0, 121.8, 120.4, 120.2, 118.8, 113.7, 111.3, 109-6, 104.7, 67.7, 67.2, 55-6, 51-1, 49-2, 38.5, 36.2, 36.1, 35-4, 31-8, 30.2, 24.7, 23.7, 22.5, 17.7; MS (ES+): m/z = 757 (M+H)

LCMS (Method A): t R = 5.80 min. HRMS (El, m/z): calculated for C 42 H 44 N 8 O 6 (M+i) + 757-3457, observed 757-3457-

(S)-/V-(4-Aminophenyl)-4-(4-(4-(4-((2-methoxy-i2-oxo-5,6, 6a,7,8,9,io,i2- octahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i- methyl-iii-pyrrole-2-carboxamido)phenyl)-i-methyl-iii-pyrrol e-2- carboxamide (16)

To a solution of (S)dV-(4-aminophenyl)-4-(4-(4-(4-((2-methoxy-i2-oxo-6a,7,8,9 ,io,i2- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i-methyl-iH- pyrrole-2-carboxamido)phenyl)-i-methyl-iff-pyrrole-2-carboxa mide (255 mg, 0.337 mmol) in tetrahydrofuran (7 mL) was sequentially added ammonium formate (170 mg, 2.70 mmol), water (700 pL) and Pd / C (10% w/w , 130 mg). The reaction mixture was heated at 35 °C for 20 h. On completion, the reaction mixture was filtered through Celite® and washed with ethyl acetate (100 mL). The filtrate was concentrated under reduced pressure to give the title compound 16 (75 mg, 29%) as a white solid. Ή NMR (DMSO-d 6 , 400 MHz) <5 9.90 (s, lH), 9.79 (s, lH), 9.48 (s, lH), 7.70 (d, J= 8.6 Hz, 2H), 7-49 (s, 2H), 7-47 (s, iH), 7.39 (d, J= 1.6 Hz, lH), 7.34 (s, lH), 7.32 (s, lH), 7.30 (d, J= 1.6 Hz, iH), 7.22 (d, J= 2.0 Hz, lH), 6.96 (d, J=i.6 Hz, lH), 6.54 (s, lH), 6.52 (s, lH), 6.37 (s, iH), 5-95 ((t, J= 3-9 Hz, lH), 4.87 (s, 2H), 4.17-4.08 (m, lH), 3.95 (t, J= 6.2 Hz, lH), 3.88 (s, 3H), 3-83 (s, 3H), 3-68 (s, 3H), 3·63-3·52 (m, lH), 3-26-3.21 (m, 2H), 3·ΐ5-3·q6 (m, lH), 2.44 (t, J=7.2 Hz, 2H), 2.04 (quin, J= 6.7 Hz, 2H), 1.79-1.68 (m, lH), 1.65-1.35 (m, lH); «C NMR (DMSO-d 6 , 100 MHz) d 169.3, 165.9, iόq.q, 159.7, 151.8, 145.9, 145-2, 141.6, 137.5, 130.1, 128.5, 127-1, 124-8, 123.1, 122.3, 122.2, 120.9, 119-8, 116.6, 114.2, 111.4,

110.1, 105.2, 101.8, 67.8, 59-2, 56.3, 51-9, 44-4, 36.8, 36.6, 32.3, 29.6, 25.1, 25.0, 23.0; MS (ES+): m/z = 759.2 (M+H) + ; LCMS (Method A): t R = 5.62 min. 4-(Benzyloxy)-3-methoxybenzaldehyde (17)

A mixture of 4-hydroxy-3-methoxybenzaldehyde (200 g, 1.31 mol), benzyl bromide (236 g, 1.38 mol) and potassium carbonate (545 g, 3.94 mol) in methanol (1.2 L) was refluxed for 5 h. The reaction mixture was filtered and the filtrate was evaporated under reduced pressure to give the title compound (271 g, 85%) as a light yellow solid. The product was carried through to the next step without any further purification. Ή NMR (400 MHz, CDCI3) d 9.83 (s, 1 H), 7.29-7.46 (m, 7 H), 6.98 (d, J = 8.1 Hz, 1 H), 5.25 (s, 2 H), 3.94 (s, 3 H); «C NMR (too MHz, CDCl 3 ) 6191.0, 153.6, 150.1, 136.0, 130.3, 128.7,

128.2, 127.2, 126.6, 112.3, 109.3, 70.9, 56.1; MS (ES+): m/z = 243 (M+H) + ; LCMS

(Method A): t R = 7.53 min

4-(Benzyloxy)-5-methoxy-2-nitrobenzaldehyde (18)

To a stirred solution of 4-(benzyloxy)-3-methoxybenzaldehyde (17) (130 g, 536.6 mmol) in trifluoroacetic acid (600 mL) was added potassium nitrate (65.1 g, 643.9 mmol, in 600 mL of trifluoroacetic acid) dropwise at o °C. The reaction mixture was stirred for another hour. The reaction mixture was diluted with water (2.4 L). The precipitate was hltered and washed with cold water (2 x 500 mL) to give the title compound (125 g, 81%) as a yellow solid. The product was carried through to the next step without any further purification. Ή NMR (400 MHz, CDCl 3 ) d 10.42 (s, 1 H), 7.66 (s, 1 H), 7.34-7.46 (m, 6 H), 5.26 (s, 2 H), 4.0 (s, 3 H); «C NMR (lOO MHz, CDCl 3 ) d 187-8, 153-7, 151-4, 134-85, 129-0, 128.9, 128.7, 127-6, 125.7, no.o, 108.9, 71-6, 56.73; MS (ES+): m/z = 286 (M-H) LCMS (Method A): t R = 7.87 min

4-Hydroxy-5-methoxy-2-nitrobenzaldehyde (19)

To a stirred solution of 4-(benzyloxy)-5-methoxy-2-nitrobenzaldehyde (18) (100 g, 348 mmol) in acetic acid (800 mL) was added hydrobromic acid (48% in acetic acid, 88.0 mL, 522 mmol). The reaction mixture was stirred at 85 °C for 1 h. The reaction mixture was diluted with water (1.6 L), the precipitate was hltered and washed with cold water (3 x 100 mL) to give the title compound (50.0 g, 73%) as a yellow solid. The product was carried through to the next step without any further purification. Ή NMR (400 MHz, , (CD 3 ) 2 SO ) d ii.ii (br s, 1 H), 10.15 (br s, 1 H), 7.50 (s, 1 H), 7.35 (s, 1 H), 3.94 (s, 3 H); MS (ES+): m/z = 196.1 (M-H>; LCMS (Method B): t R = 2.55 min. 5-Methoxy-2-nitro-4-((triisopropylsilyl)oxy)benzaldehyde (20)

A mixture of 4-hydroxy-5-methoxy-2-nitrobenzaldehyde (19) (50.0 g, 254 mmol), chlorotriisopropylsilane (59.7 mL, 279 mmol) and imidazole (51.8 g, 761 mmol) was heated at 100 °C for 30 min. The reaction mixture was poured into ice-cold water and extracted with ethyl acetate (3 x 500 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica), eluting with ethyl acetate/ hexane (isocratic 5%), to give the title compound (57.5 g, 64%) as a yellow solid. Ή NMR (400 MHz, CDCI 3 ) d 10.42 (s, 1 H), 7.59 (s, 1 H), 7.40 (s, 1 H), 3.95 (s, 3 H), 1.33- 1.24 (m, 3 H), 1.07 (s, 18 H).

5-Methoxy-2-nitro-4-((triisopropylsilyl)oxy)benzoic acid (21)

A solution of sodium chlorite (46.0 g, 407 mmol, 80% technical grade) and sodium dihydrogen phosphate (35.5 g, 228 mmol) in water (200 mL) was added to a solution of 5-methoxy-2-nitro-4-((triisopropylsilyl)oxy)benzaldehyde (20) (57.5 g, 163 mmol) in tetrahydrofuran (800 mL) at room temperature. Hydrogen peroxide (30% w/w, 235 mL, 2.28 mol) was immediately added to the vigorously stirred biphasic mixture. The starting material dissolved and the temperature of the reaction mixture rose to 45 °C. After 30 min, the reaction mixture was acidified to pH = 3-4 with citric acid (1 M, 100 mL) and extracted with ethyl acetate (3 x 500 mL). The combined extracts were washed with water (150 mL) and a saturated aqueous solution of sodium chloride (150 mL), dried over anhydrous sodium sulfate, filtered and concentrated under vacuum.

The resulting residue was purified by column chromatography (silica), eluting with ethyl acetate/ hexane (isocratic 10%) followed by methanol/dichloro methane (from 0% to 10%), to give the title compound (38.00 g, 63%) as a yellow oil. Ή NMR (400 MHz, CDCI 3 ) <59-81 (S, lH), 7.35 (s, lH), 7.25 (s, lH), 3.91 (s, 3H), 1.26 (q, J = 7.4 Hz, 3H), 1.09 (d, J = 7.4 Hz, 18H); MS (ES+): m/z = 368.1 (M-H)q LCMS (Method B): t R = 4.75 min. 0S)-(2-(Hydroxymethyl)piperidin-i-yl)(5-methoxy-2-nitro-4 ((triiso- propylsilyl)oxy)phenyl)methanone (22)

A solution of 5-methoxy-2-nitro-4-((triisopropylsilyl)oxy)benzoic acid (21) (28.0 g,

75.8 mmol), i-[bis(dirnethylamino)methylene]-i/f-i,2,3-triazolo[4,5-b]py ridinium 3- oxide hexafluorophosphate (31.7 g, 83.4 mmol) and dry triethylamine (44 mL) in anhydrous dichloromethane (300 mL) was stirred at room temperature for 30 min. (S)- Piperidin-2-ylmethanol (11.3 g, 98.5 mmol) was added and the reaction mixture was stirred at room temperature for 2 h. The reaction mixture was partitioned between dichloromethane (500 mL x 2) and water (too mL). Organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica), eluting with ethyl acetate/hexane (from 50% to 75%), to give the title compound (20.0 g, 57%) as a yellow solid. Ή NMR (400 MHz, CDCl 3 ) d 7.68 - 7.65 (m, lH), 7.03 - 6.65 (m, lH), 5.04 - 4.69 (m, lH), 4.12 - 4.05 (m, 0.41H), 4.01 - 3-95 (m, 0.46H), 3.92 - 3.89 (m, 2.57H), 3-83 - 3-74 (m, 1.47H), 3-64 - 3-59 (m, 0.35H), 3-45 - 3-40 (m, 0.28H), 3.21 - 3.01 (m, 1.39H), 2.87 - 2.79 (m, 0.38H), 1.97 - 1.94 (m, 0.55H), 1.88 - 1.77 (m, 0.58H), 1.73 - 1.62 (m, 3H), 1.56 - 1.44 (m, 2H), 1.29 - 1.24 (m, 3H), 1.09 (d, J = 7.3 Hz, 18H); MS (ES+): m/z = 467.3 (M+H) + ; LCMS (Method A): t R = 4.75 min.

0S)-(2-Amino-5-methoxy-4-((triisopropylsilyl)oxy)phenyl)( 2- (hydroxylmethyl)piperidin-i-yl)methanone (23)

A mixture of CS)-(2-(hydroxymethyl)piperidin-i-yl)(5-methoxy-2-nitro-4

((triisopropylsilyl)oxy)phenyl)methanone (22) (10.0 g, 21.4 mmol), palladium on activate charcoal (10% wt. basis) (1.00 g) in methanol (too mL) was stirred at room- temperature under an hydrogen atmosphere for 18 h. The reaction mixture was filtered through Celite® and the cake was washed with ethyl acetate (150 mL). The filtrate was concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica), eluting with ethyl acetate/hexane (from 50% to 67%), to give the title compound (8.00 g, 85%) as a yellow oil. Ή NMR (400 MHz, CDCl 3 ) d 6.67 (s, lH), 6.30 (s, lH), 4.00 - 3.81 (m, 4H), 3.72 (s, 3H), 3-57 (s, lH), 3.08 (s, lH), 1.68 - 1.64 (m, 4H), 1.57 - 1.43 (m, 2H), 1.28 - 1.17 (m, 3H), 1.08 (d, J = 7.4 Hz, 18H); MS (ES+): m/z = 437.3 (M+H) + ; LCMS (Method B): t R = 1.94 min. Allyl 0S)-(2-(2-(hydroxymethyl)piperidine-i-carbonyl)-4-methoxy-5 ((tri- isopropylsilyl)oxy)phenyl)carbamate (24)

To a stirred solution of (S)-(2-amino-5-methoxy-4-((triisopropylsilyl)oxy)phenyl)(2- (hydroxymethyl)piperidin-i-yl)methanone (23) (22.0 g, 50.4 mmol) and pyridine (7.97 g, 101 mmol) in dichloromethane (300 mL) was added allyl chloroformate (6.38 g, 52.9 mmol) dropwise at -10 °C. The reaction mixture was diluted with dichloromethane (500 mL) and washed with a saturated aqueous solution of copper (II) sulfate (150 mL), water (too mL) and a saturated aqueous solution of sodium hydrogen carbonate (too mL). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica), eluting with ethyl acetate/hexane (from 50% to 75%), to give the title compound (17.00 g, 65%) as a yellow oil. Ή NMR (400 MHz, CDCl 3 ) d 8.o8 (s, lH), 7.62 (s, lH), 6.75 (s, lH), 5.92 (ddt, J = 17.2, 10.7, 5-5 Hz, lH), 5.32 (dt, J = 17.3, 1 -7 Hz, lH), 5.20 (dt, J = 10.6, 1.4 Hz, lH), 4.61 (dt, J = 5.5, 1.5 Hz, 2H), 3.88 (t, J = 10.7 Hz, lH), 3.76 (s, 3H), 3.61 - 3.57 (m, lH), 3.20 - 3.02 (m, 2H), 2.03 (s, lH), 1.65 - 1.62 (m, 3H), 1.53 - 1.40 (m, 2H), 1.29 - 1.24 (m, 4H), 1.11 - 1.08 (m, 18H); MS (ES+): m/z = 522.3 (M+H) + ; LCMS (Method A): t R = 5.23 min.

Allyl (6a.S)-6-hydroxy-2-methoxy-i2-oxo-3-((triisopropyl-silyl)oxy )- 6,6a,7,8,9,io-hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine-5 (i2Ji)- carboxylate (25)

A mixture of allyl (S')-(2-(2-(hydroxymethyl)piperidine-i-carbonyl)-4-methoxy-5 ((triisopropylsilyl)oxy)phenyl)carbamate (24) (17.0 g, 32.7 mmol), (2, 2,6,6- Tetramethylpiperidin-i-yl)oxyl (510 mg, 3.3 mmol) and (diacetoxyiodo)benzene (12.6 g, 39.2 mmol) in dichloromethane (150 mL) was stirred at room temperature for 16 h. The reaction mixture was diluted with dichloromethane (350 mL), washed with a (100 mL), a saturated aqueous solution of sodium hydrogen carbonate (100 mL) and a saturated aqueous solution of sodium chloride (100 mL). The organic phase was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica), eluting with ethyl acetate/ hexane (from 50% to 75%), to give the title compound (13.0 g, 77%) as a yellow oil. Ή NMR (400 MHz, CDCl 3 ) d 7-13 (s, lH), 6.65 (s, lH), 5-90 (d, J = 10.3 Hz, lH), 5.78 (td, J = 10.6, 5.3 Hz, lH), 5.19 - 5.13 (m, 2H), 4-6O (dd, J = 13.1, 5-8 Hz, lH), 4.52 - 4.40 (m, lH), 4.35 (dt, J = 13.6, 4-5 Hz, lH), 3.84 (s, 3H), 3-57 - 3-39 (m, 2H), 3.14 -

2.99 (m, lH), 2.08 - 1.99 (m, lH), 1.76 - 1.61 (m, 5H), 1.25 - 1.18 (m, 3H), 1.09 - 1.05 (m, 18H); MS (ES+): m/z = 519.3 (M+H) + ; LCMS (Method A): t R = 2.41 min.

Allyl (6a-S)-2-methoxy-i2-oxo-6-((tetrahydro-2//-pyran-2-yl)oxy)-3 - ((triisopropylsilyl)oxy)-6,6a,7,8,9,io-hexahydrobenzo[e]pyri do-[i,2- «][i,4]diazepine-5(i2Ji)-carboxylate (26)

A mixture of allyl (6aS)-2-methoxy-i2-oxo-6-((tetrahydro-2//-pyran-2-yl)oxy)-3- ((triisopropylsilyl)oxy)-6, 6a, 7,8,9, io-hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine- 5(i2ff)-carboxylate (25) (14.0 g, 27.0 mmol), 3,4-dihydro-2ff-pyran (22.7 g, 270 mmol) and para-toluene sulfonic acid monohydrate (140 mg, 0.76 mmol) in

tetrahydrofuran (130 mL) was stirred at room temperature for 18 h. The reaction mixture was diluted with ethyl acetate (360 mL), washed with a saturated aqueous solution of sodium hydrogen carbonate (200 mL) and a saturated aqueous solution of sodium chloride (100 mL). The organic phase was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica), eluting with ethyl acetate/hexane

(isocratic 17%), to give the title compound (12.50 g, 77%) as a yellow oil. Ή NMR (400 MHz, CDCI 3 ) d 7-13 (s, 0.38H), 7.10 (s, 0.53H), 6.90 (s, 0.50H), 6.52 (s, 0.35H), 6.15 (d, J = 10.0 Hz, 0.37H), 5.98 (d, J = 10.0 Hz, 0.51H), 5.80 - 5.68 (m, 0.88H), 5.17 - 4.94 (m, 2.73H), 4.64 - 4-21 (m, 3H), 3·9ΐ - 3-85 (m, 0.85H), 3.83 (d, J = 1.8 Hz, 3H), 3.66 - 3-39 (m, 2H), 3.14 - 3.00 (m, lH), 2.08 - 1.87 (m, lH), 1.83 - 1.33 (m, 12H), 1.26 - 1.19 (m, 3H), 1.08 - 1.05 (m, 18H); MS (ES+): m/z = 603.4 (M+H) + ; LCMS (Method A): t R = 6.41 min.

Allyl (6aS')-3,6-dihydroxy-2-methoxy-i2-oxo-6, 6a, 7,8,9, 10-hexahydro- benzo[e]pyrido[i,2-a][i,4]diazepine-5(i2Ji)-carboxylate (27) A 1 M solution of tetrabutylammonium fluoride in tetrahydrofuran (0.3 mL) was added to a mixture of allyl (6aS)-2-methoxy-i2-oxo-6-((tetrahydro-2//-pyran-2-yl)oxy)-3- ((triisopropylsilyl)oxy)-6, 6a, 7,8,9, io-hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine- 5(i2if)-carboxylate (26) (50 mg, 0.10 mmol) in 1,4-dioxane (2 mL). the reaction mixture was stirred for 30 min and it was then washed with a saturated aqueous solution of sodium chloride (30 mL). The aqueous solution was washed with ethyl acetate (2 x 30 mL) and the organic solvent was concentrated under vacuum. The resulting residue was purified by column chromatography (silica), eluting with acetone /dichloromethane to give the title compound (36 mg, 99%) as a yellow oil. MS (ES+): m/z = 363 (M+H) + ; LCMS (Method B): £R = 2.60 min.

0S)-3-Hydroxy-2-methoxy-7,8,9,io-tetrahydrobenzo[e]-pyrid o[i,2- «][i,4]diazepin-i2(6aH)-one (28)

To a solution of allyl (6aS)-3,6-dihydroxy-2-methoxy-i2-oxo-6,6a,7,8,9,io- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine-5(i2H)-carboxyl ate (27) (36.0 mg, 0.10 mmol) in dichloromethane (3 mL) was sequentially added tetrakisftriphenyl- phosphine)palladium(o) (5-7mg, 5 mol%), and pyrrolidine (9.7 pL, 0.12 mmol). The reaction mixture was stirred at room temperature for 30 min. The reaction mixture concentrated in vacuo and subjected to high vacuum for 40 min until excess pyrrolidine was removed. The resulting residue was then purified by column chromatography (silica), eluting with methanol/ dichloromethane (from 0% to 10%) to give the title compound (20 mg, 76%) as a cream solid.

Methyl 4-(4-((tert-butoxycarbonyl)amino)phenyl)-i-methyl-iii-pyrrol e-2- carboxylate (29)

To a solution of methyl 4-bromo-i-methyl-iif-pyrrole-2-carboxylate (1.0 g, 4.60 mmol) in acetonitrile (40 mL) and water (36 mL) tert-butyl (4-(4,4,5,5-tetramethyl-i,3,2- dioxaborolan-2-yl)phenyl)carbamate (1.8 mg, 5.06 mmol), potassium carbonate (1.7 g, 13.36 mmol), and tetrakis(triphenylphosphine)palladium (280 mg, mol 5%) were l80 added. The reaction mixture was purged with nitrogen for 5 min and the reaction was irradiated with microwaves at 100 °C for 6 min. The mixture was filtered through a celite pad. The pad was washed with ethyl acetate (500 mL) and the resulting organic solution was concentrated in vacuo. The residue was purified by column

chromatography (silica), eluting with ethyl acetate /hexane (from 0% to 40%), to give the title compound (958 mg, 63%) as a white solid. Ή NMR (400 MHz, CDCl 3 ) d 7.37- 742 (m, 2H), 7.32-7.36 (m, 2H), 7.16 (d, J = 2.0 Hz, lH), 7.02 (d, J = 2.0 Hz, lH), 6.56 (s, lH), 3-94 (s, 3H), 3-83 (s, 3H), 1.52 (s, 9H); «C NMR (100 MHz, CDCl 3 ) 5 161.7, 136.5, 129.4, 127-1, 125-5, 123-6, 119.0, 115 6, 114 6, 60.4, 51.1, 36.9, 28.3; MS (ES+): m/z = 330.9 (M+H) + ; LCMS (Method B): £R = 4.22 min.

Methyl 4-(4-(4-((tert-butoxycarbonyl)amino)-i-methyl-iii-pyrrole-2- carboxamido)phenyl)-i-methyl-iii-pyrrole-2-carboxylate (30)

To a solution of methyl 4-(4-((tert-butoxycarbonyl)amino)phenyl)-i-methyl-iH- pyrrole-2-carboxylate (29) (950 mg, 2.88 mmol) in 1,4-dioxane (4 mL) and methanol (4 mL) hydrochloric acid (4 M in 1,4-dioxane) (8 mL) was added drop wise. The reaction mixture was stirred for 3 h and then concentrated in vacuo. The residue was added to a mixture of 4-((tert-butoxycarbonyl)amino)-i-methyl-ii/-pyrrole-2- carboxylic acid (830 mg, 3.45 mmol), ,iV-dimethylpyridin-4-amine (1.05 g, 8.64 mmol) and -(3-Dimethylaminopropyl)-/V'-ethylcarbodiimide hydrochloride (1.38 g, 7.20 mmol) in N, - d i m e t h y 1 f 0 r m a m i d e (15 mL) which was previously stirred for 30 min. The resulting solution was allowed to react at room temperature for 18 h. The reaction mixture was quenched with a saturated aqueous solution of sodium hydrogen carbonate (20 mL) and washed with a saturated aqueous solution of sodium chloride (150 mL). The aqueous phase was extracted with ethyl acetate (2 x 60 mL). The combined organic extracts were concentrated in vacuo. The resulting residue was purified by column chromatography (silica), eluting with acetone/dichloromethane (from 0% to 30%), to give the title compound (860 mg, 66%) as a cream solid. Ή NMR (400 MHz, CDCI 3 ) 88.01 (s, lH), 7.71 (s, iH), 7·49-7·54 (m, 2H), 7 40-7 44 (m, 2H),

7.17 (d, J = 2.0, lH), 7.03 (d, J = 1.8, lH), 6.85 (s, lH), 6.63 (s, lH), 3.94 (s, 3H), 3-88 (s, 3H), 3-83 (s, 3H) 1.50 (s, 9H); «C NMR (100 MHz, CDCl 3 ) d 161.7, 159.5, 136.0, 130.4, 126.0, 125.5, 123-5, 121.8, 120.3, 118.6, 114.6, 103.7, 5ΐ·ΐ, 36.9, 36.7, 28.4; MS (ES+): m/z = 453.I(M+H) + ; LCMS (Method B): t R = 4.07 min.

4-(4-(4-((tert-Butoxycarbonyl)amino)-i-methyl-iii-pyrrole -2- carboxamido)phenyl)-i-methyl-iii-pyrrole-2-carboxylic acid (31)

To a solution of methyl 4-(4-(4-((tert-butoxycarbonyl)amino)-i-methyl-iff-pyrrole-2- carboxamido)phenyl)-i-methyl-iff-pyrrole-2-carboxylate (30) (3.1 g, 6.85 mmol) in 1,4-dioxane (120 mL) was added an aqueous solution of sodium hydroxide (1 M, 120 mL, 120 mmol). The reaction mixture was stirred at room temperature for 18 h and was then concentrated in vacuo, after which water (80 mL) was added and the aqueous layer was acidified to pH = 4 with an aqueous solution of citric acid (1 M, 80 mL). The aqueous layer was then extracted with ethyl acetate (2 x 150 mL). The organic layer was washed with a saturated aqueous solution of sodium chloride (150 mL). The combined organic extracts were dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give the title compound (2.5 g, 83%) as a cream solid. The product was carried through to the next step without any further purification. MS (ES+): m/z = 438.8 (M+H) + ; LCMS (Method B): t R = 3.27 min. 2nd Method: 4-(4-(4-((tert-Butoxycarbonyl)amino)-i-methyl-iJT-pyrrole-2- carboxamido)phenyl)-i-methyl-iii-pyrrole-2-carboxylic acid (31)

To a solution of methyl 4-(4-(4-((tert-butoxycarbonyl)amino)-i-methyl-iif-pyrrole-2- carboxamido)phenyl)-i-methyl-iff-pyrrole-2-carboxylate (30) (500 mg, 1.1 mmol) in a mixture of tetrahydrofuran/ methanol/water (3:1:1) (15 mL) was added lithium hydroxide (132 mg, 5.5 mmol). The mixture was stirred at room temperature for 48 h then water (50 mL) was added and the solution was acidified to pH 3-4 with acetic acid and extracted with ethyl acetate. The organic layers were dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure to give the title compound (425 mg, 88%) as a beige solid. The product was carried through to the next step without any further purification. MS (ES+): m/z = 439 (M+H) + ; LCMS (Method A): t R = 7.03 min LCMS (Method B): t R = 3.67 min. 4-Amino-JV-(4-(5-((4-aminophenyl)carbamoyl)-i-methyl-iii-pyr rol-3- yl)phenyl)-i-methyl-iff-pyrrole-2-carboxamide (32)

To a solution of tert-butyl (5-((4-(5-((4-aminophenyl)carbamoyl)-i-methyl-iii-pyrrol- 3-yl)phenyl)carbamoyl)-i-methyl-iff-pyrrol-3-yl)carbamate (31) (100.0 mg, 0.19 mmol) in 1,4-dioxane (1 mL) and methanol (1 mL), hydrochloric acid (4 M in 1,4- dioxane) (2 mL) was added dropwise. The reaction mixture was stirred for 4 h and then quenched through the addition of an aqueous solution of sodium hydroxide (1 M, 10 mL, 10 mmol). The mixture was then diluted with a saturated aqueous solution of sodium chloride (30 mL) and the resulting aqueous phase was washed with

dichloromethane (3 x 30 mL). The organic layer was dried over anhydrous magnesium sulfate, filtered and concentrated in vacuo affording the titled compound (70 mg, 86%) as a brown oil. Ή NMR (400 MHz, CD 3 OD) d 7.58-7.54 (m, 3H), 7.53-7.49 (m, 2H), 7-35-7-30 (m, 2H), 7.22 (s, 2H), 6.75-6.71 (m, 2H), 6.61 (s, lH), 3.91 (s, 3H), 3 83 (s, 3H); «C NMR (100 MHz, CD 3 OD) d 126.5, 124-9, 124.6, 123.1, 122.7, 121.0, 115.3, 110.0,

35-6, 35.2; MS (ES+): m/z = 429 (M+H) + ; LCMS (Method B): t R = 2.35 min.

Methyl 4-(4-(4-amino-i -methyl- i//-pyrrole-2-carboxamido)-phenyl)-i- methyl-iJT-pyrrole-2-carboxylate hydrochloride (33)

Methyl 4-(4-(4-((tert-butoxycarbonyl)amino)-i-methyl-iff-pyrrole-2- carboxamido)- phenyl)-i-methyl-iif-pyrrole-2-carboxylate (30) (1.0 g, 2.21 mmol) was dissolved in hydrochloric acid (4 M in 1,4-dioxane) (4 mL, 16 mmol) and the reaction mixture was stirred at room temperature for 1.5 h. The reaction mixture was concentrated in vacuo to give the title compound (745 mg, 87%) as a salmon solid. The product was carried through to the next step without any further purification. Ή NMR (400MHz, DMSO- d 6 ) d 10.13 (br s, 2H), 9-97 (s, lH), 7.71-7.67 (m, 2H), 7-57 (d, J= 2.0 Hz, lH), 7-54-7-51 (m, 2H), 7.20 (d, J= 2.0 Hz, lH), 7.16 (d, J= 2.0 Hz, lH), 7.11 (d, J= 2.0 Hz, lH), 3.89 (d, « 7=2.3 Hz, 6H), 3.76 (s, 3H); «C NMR (100 MHz, DMSO-d 6 ): d I6I.2, 159-4, 137-4, 129.7, 127-5, 125-3, 125-1, 123-1, 122.7, 122.6, 121.1, 114.3, 113-5, no.o, 108.4, 5ΐ·5, 37·ΐ, 37-0; MS (ES+): m/z = 353.4 (M+H) + ; LCMS (Method B): t R = 2.57 min.

Methyl 5-(4-((tert-butoxycarbonyl)amino)-i-methyl-iii-pyrrole-2- carboxamido)benzo[b]thiophene-2-carboxylate (34)

A solution of 4-((tert-butoxycarbonyl)amino)-i-methyl-iff-pyrrole-2-carbox ylic acid (500 mg, 2.10 mmol) in L/V- d i m e t h y 1 f 0 r m a m i d e (10 mL) was charged with i-(3- dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (725 mg, 3.80 mmol) and 4- (dimethylamino)pyridine (577 mg, 4.70 mmol). The reaction mixture was stirred at room temperature for 2 h. Methyl 5-aminobenzo[b]thiophene-2-carboxylate (392 mg, 1.90 mmol) was then added and the resulting mixture was stirred at room temperature for 16 h. This was then poured into ice-water (20 mL) and extracted with ethyl acetate (3 x 50 mL). The combined organic extracts were sequentially washed with an aqueous solution of citric acid (1 M, 30 mL), a saturated aqueous solution of sodium hydrogen carbonate (35 mL), water (35 mL) and brine (35 mL). The organic layer was then dried over sodium sulfate, filtered and concentrated. The resulting residue was purified by column chromatography (silica), eluting with ethyl acetate/hexane (from 0% to 50%), to give the title compound (610 mg, 75%) as a beige solid. MS (ES+): m/z = 430 (M+H) + ; LCMS (Method A): t R = 7.90 min.

Methyl 5-(4-amino-i-melhyl-i//-pyrrole-2-carboxamido)- benzo[b]thiophene-2-carboxylate hydrochloride (35)

Methyl 5-(4-((tert-butoxycarbonyl)amino)-i-methyl-ii/-pyrrole-2-car boxamido)benzo- [b]thiophene-2-carboxylate (34) (610 mg, 1.40 mmol) was dissolved in hydrochloric acid (4 M in 1,4-dioxane) (3.6 mL) and the reaction mixture was stirred at room temperature for 2 h. The reaction mixture was concentrated in vacuo to give the title compound (600 mg, 99%) as a brown solid. The product was carried through to the next step without any further purification. MS (ES+): m/z = 330 (M+H) + ; LCMS

(Method A): t R = 5.52 min. Allyl (6aS , )-2-methoxy-3-(4-((5-((2-(methoxycarbonyl)benzo[b]thio phen-5- yl)carbamoyl)-i-methyl-iiT-pyrrol-3-yl)amino)-4-oxobutoxy)-i 2-oxo-6- ((tetrahydro-2JT-pyran-2-yl)oxy)-6, 6a, 7,8,9, io-hexahydrobenzo[e]pyrido- [i,2-a][i,4]diazepine-5(i2Ji)-carboxylate (36)

A solution of 4-(((6aS)-5-((allyloxy)carbonyl)-2-methoxy-i2-oxo-6-((tetrah ydro-2/7- pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydrobenzo[e]pyrido[i ,2-a][i,4]diazepin-3- yl)oxy)butanoic acid (9) (150 mg, 0.280 mmol) in /V,/V- dimethyl formamide (4 mL) was charged with i-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (too mg, 0.520 mmol) and 4-(dimethylamino)pyridine (80 mg, 0.65 mmol). The reaction mixture was stirred at room temperature for 30 min. Methyl 5-(4-amino-i-methyl-iff- pyrrole-2-carboxamido)benzo[b]thiophene-2-carboxylate hydrochloride (14) (95 mg, 0.26 mmol) was then added and the resulting mixture was stirred at room temperature for 16 h. This was then poured onto ice-water (20 mL) and extracted with ethyl acetate (3 x 50 mL). The combined organic extracts were sequentially washed with an aqueous solution of citric acid (1 M, 30 mL), a saturated aqueous solution of sodium hydrogen carbonate (35 mL), water (35 mL) and brine (35 mL). The organic layer was then dried over sodium sulfate, filtered and concentrated in vacuo to give the title compound (190 mg, 87%) as a yellow oil. The product was carried through to the next step without any further purification. MS (ES+): m/z = 844 (M+H) + ; LCMS (Method A): £ R = 8.10 min.

Methyl (S)-5-(4-(4-((2-methoxy-i2-oxo-6a,7,8,9,io,i2-hexahydrobenzo [e]- pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iJT -pyrrole-2- carboxamido)benzo[b]thiophene-2-carboxylate (37)

To a solution of allyl (6aS')-2-methoxy-3-(4-((5-((2-(methoxycarbonyl)benzo[¾]thio - phen-5-yl)carbamoyl)-i-methyl-iff-pyrrol-3-yl)amino)-4-oxobu toxy)-i2-oxo-6- ((tetrahydro-2ff-pyran-2-yl)oxy)-6,6a,7,8,9,io-hexahydrobenz o[e]pyrido[i,2- <3][i,4]diazepine-5(i2ff)-carboxylate (36) (190 mg, 0.220 mmol) in dichloromethane (10 mL) was added tetrakis(triphenylphosphine)palladium(o) (13 mg, 5 mol%) and pyrrolidine (22 pL, 0.27 mmol). The reaction mixture was stirred at room temperature for 30 min. The reaction mixture was subjected to high vacuum for 30 min until excess pyrrolidine was thoroughly removed. The resulting residue was then purified by column chromatography (silica), eluting with acetone/dichloromethane (from 0% to 70%), to give the title compound (60 mg, 40%) as a yellow solid. Ή NMR (400 MHz, CDCI 3 ) d 8.35 (s, lH), 8.28 (s, lH), 8.02 (s, lH), 7.94 (s, lH), 7.90 (d, J= 5.7 Hz, lH), 7.75 (d, J=8.8 Hz, lH), 7.58 (dd, J= 8.7, 2.1 Hz, lH), 7.42-7.41 (m, lH), 7.13 (d, J=i.6

Hz, lH), 6.78 (s, lH), 6.56 (d, J=i.6 Hz, lH), 4·25 ·ΐ8 (m, lH), 4.08 (t, J= 6.0 Hz, 2H), 3-93 (s, 3H), 3-88 (s, 3H), 3 83 (s, 3H), 3·79-3·75 (m, lH), 3·23-3·ΐ6 (m, lH), 2.52-2.47 (m, 2H), 2.21 (d, J= 6.4 Hz, lH), 2.18 (d, J= 2.1 Hz, lH), 1.96 (br s, 2H), 1.86-1.81 (m, 2H), 1.77-1.66 (m, 2H); «C NMR (100 MHz, CDCl 3 ) d 170.0, 167.6, 163.4, 163.2, iόq.q, 150.7, 148.0, 140.0, 139.2, 137.6, 135.8, 134.2, 130.6, 123.0, 122.9, 121.5, 121.0, ΐ20.i,

116.2, 111.7, IIO·3, 104-3, 68.1, 56.1, 53-5, 52-5, 49-7, 40.0, 36.8, 33-0, 24.9, 24-5, 22.9, I8·3; MS (ES+): m/z = 658 (M+H) + ; LCMS (Method 6.92 min.

Methyl CS)-5-(4-(4-((2-methoxy-i2-oxo-5,6,6a,7,8,9,io,i2-octahydrob enzo- [e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl- iJT-pyrrole- 2-carboxamido)benzo[b]thiophene-2-carboxylate (38)

To a solution of methyl 0S)-5-(4-(4-((2-methoxy- 12-0x0-63,7,8,9, 10,12- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i-methyl-iff- pyrrole-2-carboxamido)benzo[b]thiophene-2-carboxylate (37) (89 mg, 0.13 mmol) in tetrahydrofuran (5 mL) were sequentially added ammonium formate (171 mg, 2.71 mmol), water (500 pL) and Pd / C (10% w/w, 8.9 mg). The reaction mixture was heated at 70 °C for 4 h. On completion, the reaction mixture was diluted with ethyl acetate and filtered through a syringe driven filter (Millex®-HN 0.45 pm) the filter was washed with ethyl acetate (2 x 7 mL) and the filtrate was concentrated under reduced pressure. The resulting residue was then purified by column chromatography (silica), eluting with methanol/dichloro methane (from 0% to 15%), to give the title compound (46.3 mg, 54%) as a yellow solid. Ή NMR (400 MHz, DMSO-d 6 ) d IO.OI (s, lH), 9.92

(s, lH), 8.49 (d, J=i.9 Hz, lH), 8.18 (s, lH), 8.01-7.96 (m, lH), 7.79 (dd, J= 1.9, 9.0 Hz, lH), 7.49 (s, lH), 7.24 (d, J=i.6 Hz, lH), 7.03 (d, J= 1.9 Hz, lH), 6.37 (s, lH), 5.98-5.91

(m, lH), 3.95 (t, J=6.4 HZ, 2H), 3.89 (s, 3H), 3 * 85 (s, 3H), 3-68 (s, 3H), 3-62-3.51 (m,

2H), 3-24 (d, =3-5 Hz, 2H), 3-H (d, =3-5 Hz, lH), 2.44 (t, J=7- 2 Hz, 2H), 2.04 (t,

J= 6.6 Hz, 2H), 1.72-1.37 (m, 6H); «C NMR (100 MHz, DMSO-d 6 ) d 169.3, 165.9, 162.9,

160.3, 151-8, 145-9, 141-6, 139-3, 137-4, 136.4, 133-6, 131-5, 123-3, 122.9, 122.6, 119.5, 116.4, 111.4, 105-5, 101.8, 67.8, 59-2, 56.3, 56.1, 55-4, 53-1, 44-5, 41-2, 36.7, 36.2, 32.3, 29.6, 25.3, 25.0; MS (ES+): m/z = 660 (M+H) + ; LCMS (Method A): t R = 6.92 min; LCMS (Method B): t R = 3.60 min.

Allyl (6aS)-3-(4-((2-(ethoxycarbonyl)-i-methyl-iJi-imidazol-4-yl)a mino)-4- oxobutoxy)-2-methoxy-i2-oxo-6-((tetrahydro-2ff-pyran-2-yl)ox y)- 6,6a,7,8,9,io-hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine-5 (i2Ji)- carboxylate (39)

A solution of 4-(((6aS)-5-((allyloxy)carbonyl)-2-methoxy-i2-oxo-6-((tetrah ydro-2H- pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydrobenzo[e]pyrido[i ,2-a][i,4]diazepin-3- yl)oxy)butanoic acid (9) (340 mg, 0.640 mmol) in AyV-dimethylformamide (10 mL) was charged with i-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (222 mg, 1.20 mmol) and 4-(dimethylamino)pyridine (177 mg, 1.40 mmol). The reaction mixture was stirred at room temperature for 30 min. Ethyl 4-amino-i-methyl-iif- imidazole-2-carboxylate hydrochloride (120 mg, 0.580 mmol) was then added and the resulting mixture was stirred at room temperature for 16 h. This was then poured onto ice-water (40 mL) and extracted with ethyl acetate (3 x 100 mL). The combined organic extracts were sequentially washed with an aqueous solution of citric acid (1 M, 60 mL), a saturated aqueous solution of sodium hydrogen carbonate (70 mL), water (70 mL) and brine (70 mL). The organic layer was then dried over sodium sulfate, filtered and concentrated in vacuo to give the title compound (350 mg, 80%) as a yellow oil. The product was carried through to the next step without any further purification. MS (ES+): m/z = 684 (M+H) + ; LCMS (Method A): t R = 7.35 min.

4-(4-(((6aS)-5-((Allyloxy)carbonyl)-2-methoxy-i2-oxo-6-(( tetrahydro-2ii- pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydrobenzo[e]pyrido[i ,2-a][i,4]- diazepin-3-yl)oxy)butanamido)-i-methyl-iii-imidazole-2-carbo xylic acid

To a solution of allyl (6aS)-3-(4-((2-(ethoxycarbonyl)-i-methyl-i//-imidazol-4- yl)amino)-4-oxobutoxy)-2-methoxy-i2-oxo-6-((tetrahydro-2ff-p yran-2-yl)oxy)-

6,6a,7,8,9,io-hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepin e-5(i2ff)-carboxylate (39) (350 mg, 0.460 mmol) in 1,4-dioxane (10 mL) was added an aqueous solution of sodium hydroxide (0.5 M, 10 mL, 5.0 mmol). The reaction mixture was stirred at room temperature for 2 h and was then concentrated in vacuo, after which water (20 mL) was added and the aqueous layer was acidified to pH = 1 with an aqueous solution of citric acid (1 M, 10 mL). The aqueous layer was then extracted with ethyl acetate (2 x 50 mL). The combined organic extracts were then washed with brine (50 mL), dried over sodium sulfate, filtered and concentrated. The resulting residue was triturated in hexane, filtered and dried to give the title compound (220 mg, 74%) as a beige solid. The product was carried through to the next step without any further purification. MS (ES+): m/z = 656 (M+H) + ; LCMS (Method A): t R = 6.53 min.

Allyl (6aS')-2-methoxy-3-(4-((2-((2-(methoxycarbonyl)benzo[b]thiop hen-5- yl)carbamoyl)-i-methyl-iiT-imidazol-4-yl)amino)-4-oxobutoxy) -i2-oxo-6- ((tetrahydro-2JT-pyran-2-yl)oxy)-6, 6a, 7,8,9, io-hexahydrobenzo[e]pyrido-

[i,2-a][i,4]diazepine-5(i2Ji)-carboxylate (41)

40

A solution of 4-(4-(((6aS , )-5-((allyloxy)carbonyl)-2-methoxy-i2-oxo-6-((tetrahyd ro-2if- pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydrobenzo[e]pyrido[i ,2-a][i,4]diazepin-3- yl)oxy)butanamido)-i-methyl-iff-imidazole-2-carboxylic acid (40) (110 mg, 0.170 mmol) in AyV- d i m e t h y 1 f 0 r m a m i d e (4 mL) was charged with i-(3-dimethylamino- propyl)-3-ethylcarbodiimide hydrochloride (59 mg, 0.31 mmol) and 4-(dimethyl- amino)pyridine (47 mg, 0.38 mmol). The reaction mixture was stirred at room temperature for 30 min. Methyl 5-aminobenzo[b]thiophene-2-carboxylate (32 mg, 0.15 mmol) was then added and the resulting mixture was stirred at room temperature for 16 h. This was then poured onto ice-water (40 mL) and extracted with ethyl acetate (3 x 100 mL). The combined organic extracts were sequentially washed with an aqueous solution of citric acid (1 M, 60 mL), a saturated aqueous solution of sodium hydrogen carbonate (70 mL), water (70 mL) and brine (70 mL). The organic layer was then dried over sodium sulfate, filtered and concentrated. The resulting residue was then purified by column chromatography (silica), eluting with ethyl acetate/dichloromethane (0% to 100%), followed by methanol/ dichloromethane (from 0% to 10%), to give the title compound (50 mg, 39%) as a yellow oil. MS (ES+): m/z = 845 (M+H) + ; LCMS

(Method A): £ R = 8.22 min.

Methyl CS)-5-(4-(4-((2-methoxy-i2-oxo-6a,7,8,9,io,i2-hexahydrobenzo [e]- pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-i77 -imidazole-2- carboxamido)benzo[b]thiophene-2-carboxylate (42)

To a solution of allyl (6aS')-2-methoxy-3-(4-((2-((2-(methoxycarbonyl)benzo[¾]- thiophen-5-yl)carbamoyl)-i-methyl-ii7-imidazol-4-yl)amino)-4 -oxobutoxy)-i2-oxo-6- ((tetrahydro-277-pyran-2-yl)oxy)-6,6a,7,8,9,io-hexahydrobenz o[e]pyrido[i,2-a][i,4]- diazepine-5(i2Tf)-carboxylate (41) (50 mg, 0.06 mmol) in dichloromethane (3 mL) was added tetrakis(triphenylphosphine)palladium(o) (3.5 mg, 5 mol%) and pyrrolidine (5.8 uL, 0.07 mmol). The reaction mixture was stirred at room temperature for 30 min. The reaction mixture was subjected to high vacuum for 30 min until excess pyrrolidine was thoroughly removed. The resulting residue was then purified by column

chromatography (silica), eluting with acetone/di chloro methane (from 0% to 50%), to give the title compound (10 mg, 26%) as a yellow solid. Ή NMR (400 MHz, CDCl 3 ) d

9.07 (s, lH), 8.36 (d, J= 2.0 Hz, lH), 8.13 (s, lH), 8.03 (s, lH), 7.90 (d, J= 5.7 Hz, lH), 7.82 (d, J= 8.7 Hz, lH), 7.56 (dd, J= 8.7, 2.1 Hz, lH), 7.49-7.43 (m, 2H), 6.81 (s, lH), 4.26-4.17 (m, 2H), 4.10-4.06 (m, 3H), 3·98-3·93 (m, 6H), 3·93-3·85 (m, lH), 3.74 (td,7=5.8, 4.0 Hz, lH), 3.27-3.16 (m, lH), 2.68-2.60 (m, 2H), 2.29 (quin, J=6.4 Hz, 2H), 2.10-2.02 (m, lH), 1.97-1.89 (m, lH), 1.83-1.77 (m, 2H), 1.76 (s, 2H); ^C NMR (100

MHz, CDCI 3 ) d 169.7, 167-5, I63-3, 163.2, 160.3, 156.7, 150-4, 148-0, 140.0, 139.3, 135-8, 135-0, 130.6, 123.2, 120.1, 115.4, 114-9, 110.3, 98.0, 67.8, 65.2, 56.1, 52.6, 49-6, 39-8, 35-9, 32-9, 31-0, 29.3, 24.7, 24.6, 22.9, 18.4; MS (ES+): m/z = 659 (M+H) + ; LCMS

(Method A): £R = 7.00 min. Methyl CS)-5-(4-(4-((2-methoxy-i2-oxo-5,6,6a,7,8,9,io,i2-octahydrob enzo- [e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl- iii- imidazole-2-carboxamido)benzo[b]thiophene-2-carboxylate (43)

To a solution of methyl (S)-5-(4-(4-((2-methoxy- 12-0x0-60,7, 8, 9, 10,12- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i-methyl-iff- imidazole-2-carboxamido)benzo[b]thiophene-2-carboxylate (42) (120 mg, 0.18 mmol) in tetrahydrofuran (5 mL) were sequentially added ammonium formate (56.7 mg, 0.90 mmol), water (500 uL) and Pd / C (10% w/w, 12 mg). The reaction mixture was heated at 70 °C for 150 min. On completion, the reaction mixture was diluted with ethyl acetate and filtered through a syringe driven filter (Millex®-HN 0.45 pm) the filter was washed with ethyl acetate (2 x 7 mL), then the filtrate was concentrated under reduced pressure. The resulting residue was then purified by column chromatography (silica), eluting with methanol/dichloro methane (from 0% to 10%), to give the title compound (90 mg, 54%) as a yellow solid. Ή NMR (400 MHz, DMSO-d 6 ) d 10.41 (s, lH), 10.09 (s, lH), 8.55 (d, 7=1.6 Hz, lH), 8.20 (s, lH), 8.02 (d, J= 9.0 Hz, lH), 7.79 (dd, 7=2.1, 8.8 Hz, lH), 7.53 (s, lH), 7.49 (s, lH), 6.37 (s, lH), 5-94 (s, lH), 4-12 (d, « 7=13-3 Hz, lH), 3.98 (s, 3H), 3-94 (t, « 7=6.2 Hz, 2H), 3 89 (s, 3H), 3-67 (s, 3H), 3-61-3-53 (m, 2H), 3 23 (br s, 2H), 3.11 (t, « 7=10.0 Hz, 2H), 2.03 (t, 7= 6.8 Hz, 2H), 1.71-1.34 (m, 6H); «C NMR (100 MHz, DMSO-de) d 165.9, 161.3, 157-6, 156.4, 154-2, 151.8, 145-9, 143-7, 14ΐ·6,

139-3, 137-0, 136.4, 134-0, 13 , 126.0, 123.7, 121.6, 116.3, H5-2, 111.5, 105-7, 92.1, 73-8, 70.4, 67.7, 59-2, 56.4, 55-4, 51-9, 38.7, 35-5, 32.0, 25.0; MS (ES+): t h /z = 66i (M+H) + ; LCMS (Method A): f R = 6.97 min; LCMS (Method B): f R = 3.63 min.

Methyl (S)-4-(4-(4-(4-((2-methoxy-i2-oxo-6a,7,8,9,io,i2-hexahydrobe nzo- [e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl- iT7-pyrrole- 2-carboxamido)phenyl)-i-methyl-i77-pyrrole-2-carboxylate (44)

To a solution of allyl (6aS , )-2-methoxy-3-(4-((5-((4-(5-(methoxycarbonyl)-i-methyl -iif- pyrrol-3-yl)phenyl)carbamoyl)-i-methyl-iff-pyrrol-3-yl)amino )-4-oxobutoxy)-i2-oxo- 6-((tetrahydro-2ff-pyran-2-yl)oxy)-6,6a,7,8,9,io-hexahydrobe nzo[e]pyrido[i,2-a][i,4]- diazepine-5(i2ff)-carboxylate (13) (80 mg, 0.090 mmol) in dichloromethane (3 mL) was added tetrakis(triphenylphosphine)palladium(o) (5.3 mg, 5 mol%) and pyrrolidine (9.1 pL, 0.11 mmol). The reaction mixture was stirred at room temperature for 30 min. The reaction mixture was subjected to high vacuum for 30 min until excess pyrrolidine was thoroughly removed. The resulting residue was then purified by (silica)column chromatography, eluting with acetone/di chloro methane (from 0% to 50%), to give the title compound (23 mg, 37%) as a yellow solid. Ή NMR (400 MHz, CDCl 3 ) d 8.09 (s, lH), 8.04-8.01 (m, lH), 7.90 (d, J=5.8 HZ, IH), 7.58 (s, lH), 7.56 (s, lH), 7.44-7.40 (m, 3H), 7.18 (d, J= 2.0 Hz, lH), 7.12 (d, J=i.8 Hz, lH), 7.04 (d, J= 2.0 Hz, lH), 6.78 (s, lH), 6.50 (d, J=i.9 Hz, lH), 4.26-4.18 (m, lH), 4.07 (t, J=6.o Hz, 2H), 3.94 (s, 3H), 3.87 (s,

3H), 3-84 (d, J= 2.9 Hz, 6H), 3.76 (td, 7=5.7, 3-9 Hz, lH), 3-25-3-15 (m, lH), 2.49 (t, J=7.o Hz, 2H), 2.24-2.18 (m, 2H), 2.10-2.03 (m, lH), 2.01-1.93 (m, 2H), 1.86-1.80 (m, 2H), 1.73-1.66 (m, lH); «C NMR (lOO MHz, CDCl 3 ) d 169.9, 167.6, 163.5, 161.7, 159-7, 150.7, 147-9, 139-9, 136.4, 130.2, 126.1, 125.4, 123-3, 123-0, 120.6, 119.8, 114.6, 111.7, 110.2, 103-9, 68.1, 56.1, 53-8, 51-2, 49-7, 39-9, 37-0, 36.7, 33-0, 31-0, 29.3, 24.9, 24.5,

22.9, 18.4; MS (ES+): m/z = 681 (M+H) + ; LCMS (Method A): £R = 6.98 min.

Methyl (S)-4-(4-(4-(4-((2-methoxy-i2-oxo-5,6,6a,7,8,9,io,i2-octahyd ro- benzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-me thyl-iiT- pyrrole-2-carboxamido)phenyl)-i-methyl-iiT-pyrrole-2-carboxy late (45)

To a solution of methyl (S)-4-(4-(4-(4-((2-methoxy-i2-oxo-6a,7,8,9,io,i2-hexahydro- benzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-me thyl-iff-pyrrole-2- carboxamido)phenyl)-i-methyl-iff-pyrrole-2-carboxylate (44) (680 mg, 1.00 mmol) in tetrahydrofuran (10 mL) was sequentially added ammonium formate (505 mg, 8.01 mmol), water (1 mL) and Pd / C (10% w/w , 340 mg). The reaction mixture was heated at 35 °C for 16 h. On completion, the reaction mixture was filtered through Celite® and washed with ethyl acetate (100 mL). The filtrate was concentrated under reduced pressure to give the title compound (522 mg, 76%) as a white solid. Ή NMR (400 MHz, DMSO-de) <59-90 (s, lH), 9.79 (s, lH), 7.69 (d, J= 8.6 Hz, 2H), 7.56-7.51 (m, 2H), 7.50 (s, 2H), 7.21 (dd, J=i.8, 8.4 Hz, 2H), 6.98-6.94 (m, lH), 6.37 (s, lH), 5-97-5-91 (m, lH),

4.16-4.08 (m, lH), 3.95 (t, J= 6.1 Hz, 2H), 3.89 (s, 3H), 3-83 (s, 3H), 3-77 (s, 3H), 3-68 (s, 3H), 3-57 (d, J= 3-9 Hz, lH), 3.23 (br s, 2H), 3.15-3.07 (m, lH), 2.44 (t, J=7- 2 Hz, 2H), 2.04 (quin, J= 6.7 Hz, 2H), 1.77-1.67 (m, lH), 1.66-1.36 (m, 5H); NMR (100 MHz, DMSO-d 6 ) <5 169.3, 166.0, 161.3, 160.0, 151.8, 149.6, 145.9, 141-7, 137-8, 129.4,

127.5, 125-1, 123-2, 123.2, 122.7, 122.5, 120.8, 119.2, 116.6, 114.3, Hi-5, 110.0, 105.2

101.8, 67.8, 59-2, 56.4, 51-9, 51-4, 444, 37-0, 36.6, 32.3, 29.6, 25.3; MS (ES+): m/z

683.5 (M+H) + ; LCMS (Method B): t R = 3.20 min.

4-(4-(4-(4-(((6aS')-5-((Allyloxy)carbonyl)-2-methoxy-i2-o xo-6-((tetra- hydro-2ff-pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydrobenzo[ e]pyrido[i,2- a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iii-pyrrole-2- carboxamido)phenyl)-i-methyl-iii-pyrrole-2-carboxylic acid (46)

To a solution of allyl (6aS)-2-methoxy-3-(4-((5-((4-(5-(methoxycarbonyl)-i-methyl- iH-pyrrol-3-yl)phenyl)carbamoyl)-i-methyl-iH-pyrrol-3-yl)ami no)-4-oxobutoxy)-i2- oxo-6-((tetrahydro-2if-pyran-2-yl)oxy)-6,6a,7,8,9,io-hexahyd robenzo[e]pyrido[i,2- a][ i,4]diazepine-5(i2if)-carboxylate (13) (600 mg, 0.69 mmol) in 1,4-dioxane (10 mL) was added an aqueous solution of sodium hydroxide (1 M, 10 mL, 10 mmol). The reaction mixture was stirred at room temperature for 18 h and was then concentrated in vacuo, after which water (100 mL) was added and the aqueous layer was acidified to pH = 4 with an aqueous solution of acetic acid (5 M, 20 mL). The aqueous layer was then extracted with ethyl acetate (2 x 100 mL). The combined organic extracts were dried over sodium sulfate, filtered and concentrated to give the title compound (558 mg, 97%) as a cream solid. The product was carried through to the next step without any further purification (mixture of diastereomers). Ή NMR (400 MHz, CD 3 OD)

8 7-58-7-54 (m, 2H), 7.46 (d, J=8-3 Hz, 2H), 7 2 4 (s, lH), 7 1 8 (s, 2H), 7-13 (s, lH), 6.88

(br s, 2H), 6.17 (d, J=9.8 HZ, lH), 5-78-5-74 (m, lH), 4.66-4.38 (m, 3H), 4.26-4.12 (m, lH), 4.06 (m, 3H), 3-91 (s, 3H), 3-87 (s, 3H), 3-84 (br s, 4H), 3-67-3-49 (m, 2H), 3-44 (br s, lH), 3.11-2.96 (m, lH), 2.51 (t, J=j.30 Hz, 2H), 2.15-2.12 (m, 2H), 1.72-1.48 (m, 12H); «C NMR (100 MHz, CD 3 OD) d 175.6, 172.2, 171.4, 164.6, 102.2, 152.1, 150.9, 137.8, 133-5, I32.i, 129.2, 127.6, 126.1, 125.0, 124.7, 124 6, 123.4, 122.4, H7-6, 115-8,

115.6, 106.4, 85.5, 69.5, 67.7, 56.6, 40.2, 37.3, 37.0, 31-8, 26.5, 26.4, 24.0, 21.0, 20.6, 19.1; MS (ES+): m/z = 853 (M+H) + ; LCMS (Method B): t R = 3.83 min. iV-(4-((S')-2-((S')-2-(6-(2,5-Dioxo-2,5-dihydro-iii-pyrrol-i -yl)hexanamido)- 3-methylbutanamido)propanamido)phenyl)-4-(4-(4-(4-((CS)-2-me thoxy- i2-oxo-5,6,6a,7,8,9,io,i2-octahydrobenzo[e]pyrido[i,2-a][i,4 ]diazepin-3- yl)oxy)butanamido)-i-methyl-iii-pyrrole-2-carboxamido)phenyl )-i- methyl-iff-pyrrole-2-carboxamide (47)

To a solution of 0S)-iV-(4-aminophenyl)-4-(4-(4-(4-((2-methoxy-i2-oxo- 5,6,6a,7,8,9,io,i2-octahydrobenzo[e]pyrido[i,2-a][i,4]diazep in-3-yl)oxy)butanamido)- i-methyl-iff-pyrrole-2-carboxamido)phenyl)-i-methyl-iff-pyrr ole-2-carboxamide (16) (50 mg, 0.0659 mmol), (6-(2,5-dioxo-2,5-dihydro-iff-pyrrol-i-yl)hexanoyl)-L-valyl- L- alanine (26 mg, 0.0682 mmol) and anhydrous pyridine (134 pL, 1.66 mmol) in anhydrous Ay V- d i m e t h y 1 f 0 r m a m i d e (0.5 mL) at - 78 °C was added dropwise T3P (50% in DMF, 105 uL, 0.165 mmol). The reaction mixture was stirred from - 78 °C to room temperature for 3 h. On completion, the reaction mixture was directly loaded onto a C18 samplet and the impure residue was purified by reverse-phase flash column chromatography (Biotage, 30 g cartridge) eluting with 0.1% formic acid in water / 0.1% formic acid in acetonitrile (95/5 to 5/95, and then back to 95/5) to give the title compound (10.8 mg, 15%) as a cream solid. Ή NMR (400 MHz, DMSO-de) <5 9.93 (s, lH), 9.88 (s, lH), 9.81 (s, 2H), 8.43 (br s, lH), 8.18 (d, J=7-0 Hz, lH), 7.84 (d, J=8.6 Hz, lH), 7.71 (d, J= 8.6 Hz, 2H), 7.65 (d, J= 8.6 Hz, 2H), 7.55 (d, J= 9.0 Hz, 2H), 7.51 (s, lH), 7.50-7.47 (m, 2H), 7.44 (s, lH), 7.38 (s, lH), 7.22 (s, lH), 7.01-6.99 (m, 2H), 6.98- 6.96 (m, lH), 6.37 (s, lH), 5.96 (br s, lH), 4.39 (t, J=7· o Hz, lH), 4.21-4.08 (m, 2H), 3-95 (b d=6.2 Hz, lH), 3.91 (s, 3H), 3-84 (s, 3H), 3-68 (s, 3H), 3-60-3-55 (m, 2H), 3.37 (t, J= 6.8 Hz, lH), 3.24 (br s, 2H), 3.15-3.08 (m, lH), 2.44 (t, J=7- 2 Hz, 2H), 2.24-2.09 (m, 2H), 2.06-1.99 (m, 2H), 1.77-1.67 (m, lH), 1.65-1.56 (m, 2H), 1.51-145 (m, 4H), 1.31 (d, J= 6.6 Hz, 3H), 1.26-1.16 (m, 3H), 0.90-0.81 (m, 6H); «C NMR (lOO MHz, DMSO- d 6 ) d 196.7, 193-8, 193-0, 188.9, 172-7, 171-5, 171-4, i69·3, 169 2, 165.9, 160.0, 159-9, 155-5, 151-7, 145-8, 141-6, 141.1, 134-8, 129.9, 124-8, 123.1, 122.5, 120.9, 120.7, H9-7, 115.6, 113.7, 110.7, 105-2, 101.8, 75-6, 59-2, 56.3, 51-9, 49-4, 47-4, 44-4, 37-4, 36.6, 35-3, 34.5, 30.8, 29.6, 28.2, 26.1, 25.3, 23.0, 19.6, 18.6, 184; MS (ES+): m/z = 1123.2 (M+H) + ; LCMS (Method B): t R = 3.13 min.

Allyl (6aS)-3-(4-((5-((4-(5-((4-(CS)-2-(CS)-2-((((9if-fluoren-9-yl )methoxy)- carbonyl)amino)-3-methylbutanamido)propanamido)phenyl)carbam oyl)- i-methyl-iff-pyrrol-3-yl)phenyl)carbamoyl)-i-methyl-iJi-pyrr ol-3- yl)amino)-4-oxobutoxy)-2-methoxy-i2-oxo-6-((tetrahydro-2ii-p yran-2- yl)oxy)-6, 6a, 7,8,9, io-hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine- 5(i2/

To a stirred solution of allyl (6aS')-3-(4-((5-((4-(5-((4-aminophenyl)carbamoyl)-i- methyl-iff-pyrrol-3-yl)phenyl)carbamoyl)-i-methyl-iff-pyrrol -3-yl)amino)-4- oxobutoxy)-2-methoxy-i2-oxo-6-((tetrahydro-2 7-pyran-2-yl)oxy)-6,6a,7,8,9,io- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine-5(i2ff)-carboxy late (14) (870 mg, 0.923 mmol) and (((9H-fluoren-9-yl)methoxy)carbonyl)-L-valyl-L-alanine (450 mg, 1.10 mmol) in methanol / dichloromethane (10 mL, 1:10 v/v), was added N- ethoxycarbonyl-2-ethoxy-i,2-dihydroquinoline (340 mg, 1.37 mmol) at room temperature. The reaction mixture was stirred for 16 h. The reaction mixture was concentrated in vacuo and the resulting residue was purified by column chromatography (silica), eluting with methanol / dichloromethane (isocratic 5%), to give the title compound (860 mg, 70%) as a cream solid. MS (ES+): m/z = 1335.9 (M+H) + ; LCMS (Method B): t R = 4.20 min. Allyl (6aS)-3-(4-((5-((4-(5-((4-((S , )-2-((S , )-2-amino-3-methylbutanamido)- propanamido)phenyl)carbamoyl)-i-methyl-iii-pyrrol-3-yl)pheny l)- carbamoyl)-i-methyl-iii-pyrrol-3-yl)amino)-4-oxobutoxy)-2-me thoxy-i2- oxo-6-((tetrahydro-2ff-pyran-2-yl)oxy)-6, 6a, 7,8,9, io-hexahydrobenzo[e]- pyrido[i,2-a][i,4]diazepine-5(i2ii)-carboxylate (49)

To a stirred of allyl (6aS , )-3-(4-((5-((4-(5-((4-((S , )-2-((S')-2-((((9ff-fluoren-9- yl)methoxy)carbonyl)amino)-3-methylbutanamido)propanamido)ph enyl)carbamoyl)- i-methyl-iff-pyrrol-3-yl)phenyl)carbamoyl)-i-methyl-iff-pyrr ol-3-yl)amino)-4- oxobutoxy)-2-methoxy-i2-oxo-6-((tetrahydro-2ff-pyran-2-yl)ox y)-6,6a,7,8,9,io- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine-5(i2ff)-carboxy late (48) (1.0 g, 0.750 mmol) in /V,/V- dimethyl formamide (5 mL) was added piperidine (0.5 mL) at room temperature. The reaction mixture was stirred for 16 h. The reaction mixture was concentrated in vacuo and the resulting residue was triturated with diethyl ether / ethyl acetate (20 mL, 5:1 v/v), filtered and dried under high vacuum, to give the title compound (640 mg, 76%) as a cream solid. Ή NMR (400 MHz, DMSO-d 6 ) d g.g8 (s, lH), 9.91 (s, lH), 9.80 (d, J=y.4 Hz, 2H), 8.24-8.10 (m, lH), 7.67 (d, J=9.o Hz, 2H),

7.71 (d, J=8.6 Hz, 2H), 7.50 (d, J=8.2 HZ, 2H), 7.55 (d, J=9-0 Hz, 2H), 7.44 (s, lH), 7.39 (s, lH), 7.22 (s, lH), 7.06 (d, J= 2.7 Hz, lH), 6.96 (s, lH), 6.93-6.76 (m, lH), 6.09-5.89 (m, lH), 5.75 (d, J=o.8 Hz, lH), 5-U-4-95 (m, 2H), 4.68-4.53 (m, lH), 4.48 (br s, 2H),

4.09 (d, J=4-3 Hz, lH), 4.06-3.96 (m, 2H), 3-91 (s, 3H), 3.86-3.80 (m, 6H), 3·79-3·72 (m, lH), 3·58-3·44 (m, lH), 3.41-3.36 (m, lH), 3.02 (d, J=4-7 Hz, lH), 2.90 (d, J=9-4 Hz, lH), 2.52-2.48 (m, 2H), 2.44 (t, J= 6.8 Hz, 2H), 2.11-1.99 (m, 2H), 1.95-1.83 (m, 3H), 1.73-1-58 (m, 5H), 1.53-142 (m, 4H), 1.31 (d, J= 6.6 Hz, 3H), 1.12-1.05 (m, 2H), 0.90 (d, J=7.0 Hz, 3H), O.80 (d, J= 6.6 Hz, 3H); MS (ES+): m/z = 1114.2 (M+H) + ;

LCMS (Method A): t R = 6.37 min. iV-(4-((S')-2-((S')-2-Amino-3-methylbutanamido)propanamido)p henyl)-4- (4-(4-(4-(((S')-2-methoxy-i2-oxo-6a,7,8,9,io,i2-hexahydroben zo[e]pyrido- [i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iii-pyrro le-2- carboxamido)phenyl)-i-methyl-iii-pyrrole-2-carboxamide (50)

To a solution of allyl allyl (6aS)-3-(4-((5-((4-(5-((4-((S)-2-((S)-2-amino-3- methylbutanamido)propanamido)phenyl)carbamoyl)-i-methyl-iff- pyrrol-3- yl)phenyl)carbamoyl)-i-methyl-iff-pyrrol-3-yl)amino)-4-oxobu toxy)-2-methoxy-i2- oxo-6-((tetrahydro-2ff-pyran-2-yl)oxy)-6,6a,7,8,9,io-hexahyd robenzo[e]pyrido[i,2- <3][i,4]diazepine-5(i2ff)-carboxylate (49) (80 mg, 0.0719 mmol) in dichloro methane (2 mL) was added tetrakis(triphenylphosphine)palladium(o) (4.2 mg, 5 mol%) and pyrrolidine (7.1 pL, 0.0865 mmol). The reaction mixture was stirred at room temperature for 30 min. The reaction mixture was subjected to high vacuum for 30 min until excess pyrrolidine was thoroughly removed. The resulting residue was then purified by column chromatography (silica), eluting with methanol / dichloromethane (from 0% to 100%), to give the title compound (54 mg, 81%) as a cream solid.

Ή NMR (400 MHz, DMSO-de) d 9.98 (s, lH), 9-91 (s, lH), 9-8o (d, J=5-i Hz, 2H), 8.16 (d, J= 5.9 Hz, lH), 7.71 (d, J= 8.6 Hz, 2H), 7.68-7.64 (m, J= 9.0 Hz, 2H), 7.56-7.52 (m, J= 9.0 Hz, 2H), 7.49 (d, J= 8.6 Hz, 2H), 7.44 (s, lH), 7.38 (d, J=i.6 Hz, lH), 7.24-7.20 (m, lH), 7.08 (s, lH), 6.97 (d, J=i.6 Hz, lH), 6.54 (s, lH), 6.12 (s, lH), 4.56 (dd, J=i.8, 9.2 Hz, lH), 4.46 (d, J= 5.9 Hz, lH), 4.09 (d, J=5-5 Hz, lH), 4.01-3.94 (m, 2H), 3-91 (s, 3H), 3-83 (s, 3H), 3-73-3-67 (m, 3H), 3-46-3-39 (m, lH), 3.25 (s, lH), 3.17 (d, J=4-3 Hz, 2H), 3.02 (d, J=5.i Hz, lH), 2.48-2.42 (m, 2H), 2.04 (quin, J= 6.7 Hz, 3H), 1.98-1.88 (m, lH), 1.74 (d, J= 5.5 Hz, lH), 1.62 (dd, J= 3.9, 9.0 Hz, 2H), 1.54 (br s, lH), 1.31 (d,

J=7.o Hz, 3H), 0.90 (d, J= 6.6 Hz, 3H), 0.79 (d, J= 6.6 Hz, 3H); MS (ES+): m/z = 927.9 (M+H) + ; LCMS (Method B): t R = 2.73 min. iV-(4-((S')-2-((S')-2-amino-3-methylbutanamido)propanamido)p henyl)-4- (4-(4-(4-(((S')-2-methoxy-i2-oxo-5,6,6a,7,8,9,io,i2-octahydr obenzo[e]- pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iJT -pyrrole-2- carboxamido)phenyl)-i-methyl-iJT-pyrrole-2-carboxamide (51)

To a solution of A/-(4-((S , )-2-((S')-2-amino-3-methylbutanamido)propanamido)pheny l)- 4-(4-(4-(4-(((S)-2-methoxy-i2-oxo-6a,7,8,9,io,i2-hexahydrobe nzo[e]pyrido[i,2- a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iff-pyrrole-2- carboxamido)phenyl)-i- methyl-iff-pyrrole-2-carboxamide (50) (50 mg, 0.0539 mmol) in tetrahydrofuran (3.5 mL) was sequentially added ammonium formate (28 mg, 0.444 mmol), water (350 uL) and Pd / C (10% w/w, 25 mg). The reaction mixture was heated at 35 °C for 16 h. On completion, the reaction mixture was filtered through Celite® and washed with ethyl acetate (too mL). The filtrate was concentrated under reduced pressure to give the title compound (33 mg, 66%) as a white solid. Ή NMR (400 MHz, DMSO-d 6 ) d 9.98 (s, lH), 9.90 (s, lH), 9.80 (d, J=5-5 Hz, 2H), 8.17 (d, J= 6.2 Hz, lH), 7.71 (d, J= 8.6 Hz, 2H), 7.68-7.65 (m, J= 9.0 Hz, 2H), 7.56-7.52 (m, J= 9.0 Hz, 2H), 7-51-7-49 (m, 2H), 7.48 (s, lH), 7.44 (d, J=i.6 Hz, lH), 7.38 (s, lH), 7.22 (d, J=i.6 Hz, lH), 6.96 (d, J=i.6 Hz, lH), 6.37 (s, lH), 5.95 (t, J=3-7 Hz, lH), 4.46 (d, J= 5.9 Hz, 1 H), 4.16-4.07 (m, lH), 3.95 (t,

J= 6.2 Hz, 2H), 3-91 (s, 3H), 3-83 (s, 3H), 3-68 (s, 3H), 3.61-3.54 (m, lH), 3-44 (d, J= 6.6 Hz, lH), 3.24 (d, J=3.i Hz, lH), 3.17 (s, lH), 3.15-3.07 (m, lH), 3.03 (d, J=5-i Hz, lH), 2.44 (t, J=7.2 Hz, 2H), 2.07-2.00 (m, 2H), 2.00-1.89 (m, lH), 1.72 (dd, J= 4.9, 7.6 Hz, lH), 1.66-1.53 (m, 3H), 1.52-1.39 (m, 2H), 1.35 (s, lH), 1.31 (d, J=7- o Hz, 3H), 0.90 (d, J=7. o Hz, 3H), 0.80 (d, J= 6.6 Hz, 3H); MS (ES+): m/z = 930.0 (M+H) + ; LCMS

(Method A): t R = 5.75 min.

(S')-4-(4-(4-(4-((2-Methoxy-i2-oxo-6a,7,8,9,io,i2-hexahyd robenzo[e]- pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iJT -pyrrole-2- carboxamido)phenyl)-i-methyl-iJT-pyrrole-2-carboxylic acid (52)

To a solution of 4-(4-(4-(4-(((6aS')-5-((allyloxy)carbonyl)-2-methoxy-i2-oxo- 6- ((tetrahydro-2ff-pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydr obenzo[e]pyrido[i,2- <3][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iff-pyrrol e-2-carboxamido)phenyl)-i- methyl-iff-pyrrole-2-carboxylic acid (46) (1.0 g, 1.17 mmol) in dichloromethane (10 mL) was added tetrakis(triphenylphosphine)palladium(o) (68 mg, 5 mol%) and pyrrolidine (115 pL, 1.40 mmol). The reaction mixture was stirred at room temperature for 5 min. The reaction mixture was subjected to high vacuum for 30 min until excess pyrrolidine was thoroughly removed. The resulting residue was then purified by column chromatography (silica), eluting with methanol/di chloro methane (from 0% to 100%), to give the title compound (381 mg, 49%) as a bright yellow solid.

MS (ES+): m/z = 666.9 (M+H) + ; LCMS (Method B): t R = 2.82 min.

CS)-4-(4-(4-(4-((2-Methoxy-i2-oxo-5,6,6a,7,8,9,io,i2-octa hydrobenzo[e]- pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iJT -pyrrole-2- carboxamido)phenyl)-i-methyl-iJT-pyrrole-2-carboxylic acid (53)

To a solution of GS)-4-(4-(4-(4-((2-methoxy-i2-oxo-6a,7,8,9,io,i2-hexahydrobe nzo[e]- pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iff -pyrrole-2- carboxamido)phenyl)-i-methyl-iff-pyrrole-2-carboxylic acid (52) (380 mg, 0.567 mmol) in tetrahydrofuran (35 mL) was sequentially added ammonium formate (286 mg, 4.53 mmol), water (3.5 mL) and Pd / C (10% w/w, 190 mg). The reaction mixture was heated at 35 °C for 16 h. On completion, the reaction mixture was filtered through Celite® and washed with ethyl acetate (200 mL). The filtrate was concentrated under reduced pressure to give the title compound (345 mg, 91%) as an off-white solid.

MS (ES+): m/z = 669.0 (M+H) + ; LCMS (Method B): t R = 2.93 min.

CS)-4-(4-((2-Methoxy-i2-oxo-5,6,6a,7,8,9,io,i2-octahydrob enzo[e]pyrido- [i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-JV-(4-(i- methyl-5-(p- tolylcarbamoyl)-iiT-pyrrol-3-yl)phenyl)-iJT-pyrrole-2-carbox amide (54)

A solution of GS)-4-(4-(4-(4-((2-methoxy-i2-oxo-5,6,6a,7,8,9,io,i2-octahyd robenzo[e]- pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iff -pyrrole-2-carbox- amido)phenyl)-i-methyl-iff-pyrrole-2-carboxylic acid (53) (50 mg, 0.0748 mmol) in anhydrous dichloromethane (0.5 mL) was charged with A/-[(dimethylamino)-iff-i, 2,3- triazolo-[4,5-0]pyridin-i-ylmethylene]-V-methylmethanaminiii m hexafluorophosphate N-oxide (30 mg, 0.0789 mmol) and anhydrous triethylamine (44 uL, 0.317 mmol). The reaction mixture was stirred at room temperature for 15 min. p-Toluidine (8.0 mg, 0.0747 mmol) was then added and the resulting mixture was stirred at room

temperature for 16 h. The reaction mixture was quenched with a saturated aqueous solution of sodium hydrogen carbonate (20 mL) and extracted with dichloromethane (2 x 50 mL). The combined organic extracts were washed with water containing a few drops of acetic acid (30 mL). The organic layer was then dried over sodium sulfate, filtered and concentrated in vacuo. The resulting residue was then purified by column chromatography (silica), eluting with acetone/di chloro methane (from 0% to 100%), to give the title compound (30 mg, 50%) as a cream solid. Ή NMR (400 MHz, DMSO-d 6 ) d 9-99 (s, lH), 9.86 (s, lH), 9.82 (s, lH), 9.12-8.75 (m, lH), 7-79-7-73 (m, J=8.6 Hz,

2H), 7.70-7.64 (m, 2H), 7-58-7-52 (m, 2H), 7.49 (s, lH), 7.45 (br s, 2H), 7.27 (s, lH), 7-21-7-15 (m, =7- 8 Hz, 2H), 7.03 (s, lH), 6.74 (br s, lH), 4.05-4.01 (m, lH), 3.95 (s,

3H), 3.88 (s, 3H), 3.78 (s, 3H), 3.68 (br s, 2H), 3.33 (d, J=n.7 Hz, 2H), 3-i6-3-io (m, lH), 2.50 (d, J= 14.8 Hz, 2H), 2.32 (s, 3H), 2.09 (d, J= 6.2 Hz, 2H), 1.91-1.84 (m, lH), 1.71 (br s, 2H), 1.58 (br s, 4H); MS (ES+): m/z = 758.8 (M+H) + ; LCMS (Method B): t R = 3.47 min.

Allyl (6aS')-2-methoxy-3-(4-((i-methyl-5-((4-(2,2,2-trifluoroaceta mido)- phenyl)carbamoyl)-iii-pyrrol-3-yl)amino)-4-oxobutoxy)-i2-oxo -6- ((tetrahydro-2JT-pyran-2-yl)oxy)-6, 6a, 7,8,9, io-hexahydrobenzo[e]pyrido- [i,2-a][i,4]diazepine-5(i2Ji)-carboxylate (55)

A solution of 4-(4-(((6aS')-5-((allyloxy)carbonyl)-2-methoxy-i2-oxo-6-((te trahydro-2ff- pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydrobenzo[e]pyrido[i ,2-a][i,4]diazepin-3- yl)oxy)butanamido)-i-methyl-iff-pyrrole-2-carboxylic acid (12) (600 mg, 0.916 mmol) in /V,/V- dimethyl formamide (5 mL) was charged with i-(3-dimethylaminopropyl)-3- ethylcarbodiimide hydrochloride (352 mg, 1.84 mmol) and 4-(dimethylamino)pyridine (280 mg, 2.29 mmol). The reaction mixture was stirred at room temperature for 30 min. A/-(4-Aminophenyl)-2,2,2-trifluoroacetamide (188 mg, 0.921 mmol) was then added and the resulting mixture was stirred at room temperature for 16 h. This was then poured into ice-cold water (30 mL) and extracted with ethyl acetate (2 x 50 mL). The organic layer was then dried over sodium sulfate, filtered and concentrated in vacuo to give the title compound (770 mg, 99%) as a brown oil. The product was carried through to the next step without any further purification. MS (ES+): m/z = 841.6 (M+H) + ; LCMS (Method B): t R = 3.50 min.

Allyl (6aS)-3-(4-((5-((4-aminophenyl)carbamoyl)-i-methyl-iii-pyrro l-3- yl)amino)-4-oxobutoxy)-2-methoxy-i2-oxo-6-((tetrahydro-2JT-p yran-2- yl)oxy)-6, 6a, 7,8,9, io-hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine- 5(i2Ji)-carboxylate (56)

A solution of allyl (6aS)-2-methoxy-3-(4-((i-methyl-5-((4-(2,2,2-trifluoroacetam ido)- phenyl)carbamoyl)-iff-pyrrol-3-yl)amino)-4-oxobutoxy)-i2-oxo -6-((tetrahydro-2if- pyran-2-yl)oxy)-6, 6a, 7,8,9, io-hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine-5(i2if)- carboxylate (55) (770 mg, 0.916 mmol) in 1,4-dioxane (18 mL) was stirred at room temperature for 2 min. A 1 M aqueous sodium hydroxide solution (18 mL, 18.0 mmol) was then added and the resulting mixture was stirred at room temperature for 16 h. The reaction mixture was concentrated under reduced pressure. The resulting residue was then partitioned between ice-cold water (30 mL) and extracted with ethyl acetate (2 x 50 mL). The organic layer was then dried over sodium sulfate, filtered and

concentrated in vacuo to give the title compound (588 mg, 86%) as a beige solid. The product was carried through to the next step without any further purification. MS (ES+): m/z = 745.6 (M+H) + ; LCMS (Method B): t R = 2.83 min.

( < S)-iV-(4-Aminophenyl)-4-(4-((2-methoxy-i2-oxo-6a,7,8,9 ,io,i2- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i- methyl-iJT-pyrrole-2-carboxamide (57)

To a solution of allyl (6aS)-3-(4-((5-((4-ami nophenyl )carbamoyl)-i-methyl-i//-pyrrol- 3-yl)amino)-4-oxobutoxy)-2-methoxy-i2-oxo-6-((tetrahydro-2ff -pyran-2-yl)oxy)- 6,6a,7,8,9,io-hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine-5 (i2ff)-carboxylate (56) (588 mg, 0.789 mmol) in dichloromethane (5 mL) was added tetrakis(triphenyl- phosphine)palladium(o) (46 mg, 5 mol%) and pyrrolidine (78 pL, 0.950 mmol). The reaction mixture was stirred at room temperature for 1 h. The reaction mixture was subjected to high vacuum for 30 min until excess pyrrolidine was thoroughly removed. The resulting residue was then purified by column chromatography (silica), eluting with methanol / dichloromethane (from 0% to 100%), to give the title compound (228 mg, 52%) as a cream solid. MS (ES+): m/z = 559.4 (M+H) + ; LCMS (Method B): t R = 2.35 min. ( < S)-iV-(4-Aminophenyl)-4-(4-((2-methoxy-i2-oxo-5,6,6a,7 ,8,9,io,i2- octahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i- methyl-iJT-pyrrole-2-carboxamide (58)

To a solution of GS)-iV-(4-aminophenyl)-4-(4-((2-methoxy-i2-oxo-6a,7,8,9,io,i 2- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i-methyl-iif- pyrrole-2-carboxamide (57) (20 mg, 0.0358 mmol) in tetrahydrofuran (2 mL) was sequentially added ammonium formate (18 mg, 0.285 mmol), water (0.2 mL) and Pd / C (10% w/w , 10 mg). The reaction mixture was heated at 35 °C for 16 h. On

completion, the reaction mixture was filtered through Celite® and washed with ethyl acetate (too mL). The filtrate was concentrated under reduced pressure to give the title compound (10.9 mg, 54%) as a salmon solid. Ή NMR (400 MHz, DMSO-d 6 ) d 9.86 (s, iH), 9-43 (s, lH), 7.49 (s, lH), 7.31-7.25 (m, 7=7. o Hz, 2H), 7-17 (s, lH), 6.83 (s, lH), 6.54-6.47 (m, J=7. o Hz, 2H), 6.37 (s, lH), 5.95 (br s, lH), 4.86 (br s, 2H), 4-12 (d,7=13-3 Hz, lH), 3.94 (br s, 2H), 3 80 (s, 3H), 3·69-3·65 (m, 3H), 3-57 (br s, lH), 3.23 (br s, 2 H), 3.11 (t, 7=11.3 Hz, lH), 2.42 (t, « 7=6.4 Hz, 2H), 2.07-1.98 (m, 2H), 1.77-1.67 (m, lH), 1.66-1.52 (m, 4H), 1.06 (dt, J= 2.0, 7.0 Hz, lH); MS (ES+): m/z = 561.5 (M+H) + ; LCMS (Method A): t R = 4.87 min.

Methyl 5-(4-(4-(((6aS')-5-((allyloxy)carbonyl)-2-methoxy-i2-oxo-6-( (tetra- hydro-2JT-pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydrobenzo[ e]pyrido[i,2- «][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iJT-imidazole -2- carboxamido)benzo[d]oxazole-2-carboxylate (59)

A solution of 4-(4-(((6a.S)-5-((allyloxy)carbonyl)-2-methoxy-i2-oxo-6-((te trahydro-277- pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydrobenzo[e]pyrido[i ,2-a][i,4]diazepin-3- yl)oxy)butanamido)-i-methyl-iii-imidazole-2-carboxylic acid (40) (200 mg, 0.305 mmol) in anhydrous /V,/V- dimethyl formamide (1.5 mL) was charged with V-[(dimethyl- amino)-i/7-i,2,3-triazolo-[4,5-0]pyridin-i-ylmethylene]-V-me thyl-methanaminium hexafluorophosphate A-oxide (122 mg, 0.321 mmol) and anhydrous triethylamine (180 nL, 1.29 mmol). The reaction mixture was stirred at room temperature for 15 min.

Methyl 5-aminobenzo[d]oxazole-2-carboxylate (59 mg, 0.307 mmol) was then added and the resulting mixture was stirred at room temperature for 16 h. The reaction mixture was poured into ice-cold water (30 mL) and extracted with ethyl acetate (2 x 50 mL). The organic layer was dried over sodium sulfate, filtered and concentrated in vacuo. The resulting residue was purified by column chromatography (silica), eluting with methanol / dichloromethane (from 0% to 100%), to give the title compound (212 mg, 84%) as a brown oil. MS (ES+): m/z = 830.6 (M+H) + ; LCMS (Method B): t R = 3.50 min. Methyl CS)-5-(4-(4-((2-methoxy-i2-oxo-6a,7,8,9,io,i2-hexahydrobenzo [e]- pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iii -imidazole-2- carboxamido)benzo[d]oxazole-2-carboxylate (60)

To a solution of allyl methyl 5-(4-(4-(((6aS)-5-((allyloxy)carbonyl)-2-methoxy-i2-oxo- 6-((tetrahydro-2ff-pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahy drobenzo[e]pyrido[i,2- <3][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iff-imidaz ole-2-carboxamido)benzo- [d]oxazole-2-carboxylate (59) (60 mg, 0.0723 mmol) in dichloromethane (2 mL) was added tetrakis(triphenylphosphine)palladium(o) (4.2 mg, 5 mol%) and pyrrolidine (7.2 uL, 0.0877 mmol). The reaction mixture was stirred at room temperature for 15 min. The reaction mixture was subjected to high vacuum for 30 min until excess pyrrolidine was thoroughly removed. The resulting residue was then purified by column

chromatography (silica), eluting with methanol / dichloromethane (from 0% to 100%), to give the title compound (19 mg, 41%) as a cream solid. Ή NMR (400 MHz, DMSO- d 6 ) d 10.40 (s, lH), 10.23 (s, lH), 8.40 (d, J= 2.0 Hz, lH), 7-93-7-84 (m, 2H), 7.53 (s, lH), 7-13-7-04 (m, lH), 6.53 (s, lH), 6.11 (s, lH), 4-59-4-51 (m, lH), 3.98 (s, 6H), 3.95- 3-93 (m, lH), 3.82 (s, lH), 3-74-3-66 (m, 3H), 3-48-3-39 (m, lH), 3.24 (s, lH), 3.17 (d, J= 5.5 Hz, lH), 2.54 (br s, lH), 2.08-2.00 (m, 2H), 1.92-1.71 (m, 2H), 1.68-1.47 (m, 4H); MS (ES+): m/z = 644.5 (M+H) + ; LCMS (Method B): t R = 2.92 min.

Methyl CS)-5-(4-(4-((2-methoxy-i2-oxo-5,6,6a,7,8,9,io,i2- octahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i- methyl-iJT-imidazole-2-carboxamido)benzo[d]oxazole-2-carboxy late (61)

To a solution of methyl GS)-5-(4-(4-((2-methoxy-i2-oxo-6a,7,8,9,io,i2-hexahydro- benzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-me thyl-iif-imidazole-2- carboxamido)benzo[d]oxazole-2-carboxylate (60) (16.6 mg, 0.0258 mmol) in tetra- hydrofuran (3.5 mL) was sequentially added ammonium formate (13 mg, 0.206 mmol), water (350 pL) and Pd / C (10% w/w, 9 mg). The reaction mixture was heated at 35 °C for 16 h. On completion, the reaction mixture was filtered through Celite® and washed with ethyl acetate (too mL). The filtrate was concentrated under reduced pressure. The resulting residue was loaded onto a C18 samplet and purified by reverse-phase flash column chromatography (Biotage, 30 g cartridge) eluting with 0.1% formic acid in water / 0.1% formic acid in acetonitrile (95/5 to 5/95, and then back to 95/5), to give the title compound (4.83 mg, 29%) as a cream solid. Ή NMR (400 MHz, DMSO-d 6 ) d 10.38 (s, lH), 10.21 (s, lH), 8.38 (d, J= 2.0 Hz, lH), 7.92-7.81 (m, 2H), 7.47 (s, lH), 7.51 (s, lH), 6.35 (s, lH), 5.94 (br s, lH), 4.13-4.07 (m, lH), 3.96 (s, 6H), 3.92 (t, J= 6.2 Hz,

2H), 3.65 (s, 3H), 3-57-3-53 (m, 2H), 3.21 (br s, 2H), 3.14-3.02 (m, 2H), 2.02 (t, J= 6.6 Hz, 2H), 1.72-1.68 (m, lH), 1.61-1.52 (m, 3H), 1.47-1.40 (m, 2H); MS (ES+): m/z = 646.7 (M+H) + ; LCMS (Method A): t R = 6.27 min. Allyl (6aS)-2-methoxy-3-(4-((2-((2-(methoxycarbonyl)-iii- benzo[d]imidazol-5-yl)carbamoyl)-i-methyl-iJT-imidazol-4-yl) amino)-4- oxobutoxy)-i2-oxo-6-((tetrahydro-2JT-pyran-2-yl)oxy)-6,6a,7, 8,9,io- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine-5(i2H)-carboxyl ate (62)

A solution of 4-(4-(((6aS')-5-((allyloxy)carbonyl)-2-methoxy-i2-oxo-6-((te trahydro-2H- pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydrobenzo[e]pyrido[i ,2-a][i,4]diazepin-3- yl)oxy)butanamido)-i-methyl-iff-imidazole-2-carboxylic acid (40) (200 mg, 0.305 mmol) in anhydrous N,iV-dimethylformamide (1.5 mL) was charged with iV- [(dimethyl - amino)-iff-i,2,3-triazolo-[4,5-h]pyridin-i-ylmethylene]-A/-m ethylmethanaminium hexafluorophosphate iV-oxide (122 mg, 0.321 mmol) and anhydrous triethylamine (180 uL, 1.29 mmol). The reaction mixture was stirred at room temperature for 15 min. Methyl 5-amino-iif-benzo[d]imidazole-2-carboxylate (59 mg, 0.309 mmol) was then added and the resulting mixture was stirred at room temperature for 5 h. The reaction mixture was poured into ice-cold water (30 mL) and extracted with ethyl acetate (2 x 50 mL). The organic layer was dried over sodium sulfate, filtered and concentrated in vacuo. The resulting residue was purified by column chromatography (silica), eluting with methanol / dichloromethane (from 0% to 100%), to give the title compound (84 mg, 33%) as a cream solid. Ή NMR (400 MHz, CDCl 3 ) d 9.45 (br s, lH), 8.91 (br s, lH), 8.07 (br s, lH), 7.76 (br s, lH), 7.66 (br s, lH), 7.45 (br s, lH), 7.23 (d, J= 4.3 Hz, lH), 6.92 (br s, lH), 6.64 (br s, lH), 6.32-5.97 (m, lH), 5.71 (br s, lH), 5.13 (br s, lH), 5.07- 4.95 (m, 2H), 4.61 (br s, lH), 4.33 (br s, lH), 4.07 (br s, 3H), 4.05 (br s, 3H), 3.92 (br s, 3H), 3.86 (d, J= 3.9 Hz, 2H), 3.11 (br s, 2H), 2.68 (d, J=7.8 Hz, 3H), 2.26 (br s, 2H), 2.07 (br s, lH), 1.97 (d, J=i3-7 Hz, lH), 1.74 (d, J=8.6 Hz, 4H), 1.67 (br s, 3H), 1.55 (br s, 3H), 1.35-1-23 (m, 2H); MS (ES+): m/z = 829.7 (M+H) + ; LCMS (Method A): t R = 6.85 min.

Methyl CS)-5-(4-(4-((2-methoxy-i2-oxo-6a,7,8,9,io,i2-hexahydrobenzo [e]- pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iJi -imidazole-2- carboxamido)-iiT-benzo[d]imidazole-2-carboxylate (63)

To a solution of allyl (6aS)-2-methoxy-3-(4-((2-((2-(methoxycarbonyl)-iH-benzo[d]- imidazol-5-yl)carbamoyl)-i-methyl-iff-imidazol-4-yl)amino)-4 -oxobutoxy)-i2-oxo-6- ((tetrahydro-2ff-pyran-2-yl)oxy)-6,6a,7,8,9,io-hexahydrobenz o[e]pyrido[i,2-a][i,4]- diazepine-5(i2ff)-carboxylate (62) (30 mg, 0.0362 mmol) in dichloromethane (2 mL) was added tetrakis(triphenylphosphine)palladium(o) (2.1 mg, 5 mol%) and pyrrolidine (3.6 pL, 0.0438 mmol). The reaction mixture was stirred at room temperature for 15 min. The reaction mixture was subjected to high vacuum for 30 min until excess pyrrolidine was thoroughly removed. The resulting residue was then purified by column chromatography (silica), eluting with methanol / dichloromethane (from 0% to 100%), to give the title compound (17.7 mg, 76%) as an off-white solid. Ή NMR (400 MHz, DMSO-d 6 ) <5 13.44 (br s, lH), 10.43 (s, lH), 10.04 (br s, lH), 8.27 (br s, lH), 8.00 (d, J= 5.5 Hz, lH), 7.69 (br s, lH), 7.60-7.39 (m, 2H), 7.16-7.05 (m, lH), 6.60-6.43 (m, lH), 4-61-4-50 (m, lH), 4.14-4.02 (m, lH), 3.98 (s, 3H), 3-94 (s, 3H), 3-84-3-79 (m, lH), 3-73-3-65 (m, 3H), 3-48-3-37 (m, lH), 3.24 (s, lH), 3-19-3-14 (m, lH), 3-14-3-02 (m, lH), 2.08-2.01 (m, 2H), 1.92-1.80 (m, lH), 1.80-1.71 (m, lH), 1.70-1.44 (m, 4H); MS (ES+): m/z = 643.5 (M+H) + ; LCMS (Method A): t R = 5.60 min.

Methyl CS)-5-(4-(4-((2-methoxy-i2-oxo-5,6,6a,7,8,9,io,i2-octahydrob enzo- [e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl- iJT- imidazole-2-carboxamido)-iiT-benzo[d]imidazole-2-carboxylate (64)

To a solution of methyl GS)-5-(4-(4-((2-methoxy-i2-oxo-6a,7,8,9,io,i2-hexahydro- benzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-me thyl-iff-imidazole-2- carboxamido)-iff-benzo[d]imidazole-2-carboxylate (63) (16.1 mg, 0.0250 mmol) in tetrahydrofuran (3.5 mL) was sequentially added ammonium formate (13 mg, 0.206 mmol), water (350 uL) and Pd / C (10% w/w, 9 mg). The reaction mixture was heated at 35 °C for 16 h. On completion, the reaction mixture was filtered through Celite® and washed with ethyl acetate (too mL). The filtrate was concentrated under reduced pressure. The resulting residue was loaded onto a C18 samplet and purified by reverse- phase flash column chromatography (Biotage, 30 g cartridge) eluting with 0.1% formic acid in water / 0.1% formic acid in acetonitrile (95/5 to 5/95, and then back to 95/5), to give the title compound (4.0 mg, 25%) as a cream solid. MS (ES+): m/z = 645.5 (M+H) + ; LCMS (Method A): t R = 5.65 min.

Allyl (6aS , )-2-methoxy-3-(4-((i-methyl-5-((4-(i-methyl-5-(phenyl- carbamoyl)-iiT-pyrrol-3-yl)phenyl)carbamoyl)-iiT-pyrrol-3-yl )amino)-4- oxobutoxy)-i2-oxo-6-((tetrahydro-2JT-pyran-2-yl)oxy)-6,6a,7, 8,9,io- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine-5(i2H)-carboxyl ate (65)

A solution of 4-(4-(4-(4-(((6aS')-5-((allyloxy)carbonyl)-2-methoxy-i2-oxo- 6- ((tetrahydro-2ff-pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydr obenzo[e]pyrido[i,2- <3][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iff-pyrrol e-2-carboxamido)phenyl)-i- methyl-iff-pyrrole-2-carboxylic acid (46) (too mg, 0.117 mmol) in anhydrous dichloromethane (0.5 mL) was charged with /V-[(dimethylamino)-iif-i, 2, 3-triazolo- [4,5-h]pyridin-i-ylmethylene]-V-methylmethanaminiiim hexafluorophosphate V-oxide (47 mg, 0.124 mmol) and anhydrous triethylamine (69 pL, 0.496 mmol). The reaction mixture was stirred at room temperature for 15 min. Aniline (11 uL, 0.120 mmol) was then added and the resulting mixture was stirred at room temperature for 1.5 h. The reaction mixture was directly loaded onto a cartridge (25 g) and purified by column chromatography (silica), eluting with methanol / dichloromethane (from 0% to 100%), to give the title compound (105 mg, 97%) as a yellow oil. MS (ES+): m/z = 928.8 (M+H) + ; LCMS (Method B): t R = 3.73 min.

CS)-4-(4-((2-Methoxy-i2-oxo-6a,7,8,9,ic>,i2-hexahydrob enzo[e]pyrido[i,2- a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iV-(4-(i-methy l-5- (phenylcarbamoyl)-iiT-pyrrol-3-yl)phenyl)-iJT-pyrrole-2-carb oxamide (66)

To a solution of allyl (6aS')-2-methoxy-3-(4-((i-methyl-5-((4-(i-methyl-5-(phenyl- carbamoyl)-iff-pyrrol-3-yl)phenyl)carbamoyl)-iff-pyrrol-3-yl )amino)-4-oxobutoxy)- i2-oxo-6-((tetrahydro-2ff-pyran-2-yl)oxy)-6,6a,7,8,9,io-hexa hydrobenzo[e]pyrido[i,2- a][i,4]diazepine-5(i2ff)-carboxylate (65) (102 mg, 0.110 mmol) in dichloromethane (2 mL) was added tetrakis(triphenylphosphine) palladium(o) (6.4 mg, 5 mol%) and pyrrolidine (11 pL, 0.134 mmol). The reaction mixture was stirred at room temperature for 30 min. The reaction mixture was subjected to high vacuum for 30 min until excess pyrrolidine was thoroughly removed. The resulting residue was then purified by column chromatography (silica), eluting with methanol / dichloromethane (from 0% to 100%), to give the title compound (29 mg, 36%) as a cream solid. MS (ES+): m/z = 742.7 (M+H) + ; LCMS (Method A): t R = 7.05 min.

CS)-4-(4-((2-Methoxy-i2-oxo-5,6,6a,7,8,9,io,i2-octahydrob enzo[e]pyrido- [i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-JV-(4-(i- methyl-5-

(phenylcarbamoyl)-iiT-pyrrol-3-yl)phenyl)-iJT-pyrrole-2-c arboxamide (67)

To a solution of 0S)-4-(4-((2-methoxy- 12-0x0-63,7,8, 9,10, i2-hexahydrobenzo[e]- pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iV- (4-(i-methyl-5-(phenyl- carbamoyl)-iff-pyrrol-3-yl)phenyl)-iff-pyrrole-2-carboxamide (66) (29 mg, 0.0391 mmol) in tetrahydrofuran (3.5 mL) was sequentially added ammonium formate (20 mg, 0.317 mmol), water (350 uL) and Pd / C (10% w/w, 15 mg). The reaction mixture was heated at 35 °C for 16 h. The reaction mixture was filtered through Celite® and washed with ethyl acetate (100 mL). The filtrate was concentrated in vacuo to give the title compound (18 mg, 62%) as a cream solid. Ή NMR (400 MHz, DMSO-d 6 ) d 9.91 (s, lH), 9.83 (s, lH), 9.80 (s, lH), 7.73 (t, J= 9.0 Hz, 4H), 7.51 (s, lH), 7.49 (s, 2H), 7.46 (s, lH), 7.41 (s, lH), 7.33 (t, J=7- 6 Hz, 2H), 7.22 (s, lH), 7.09-7.03 (m, lH), 6.97 (s, lH), 6.37 (s, lH), 5.95 (br s, lH), 4.18-4.07 (m, lH), 3.95 (t, J= 6.2 Hz, 2H), 3.91 (s, 3H), 3 84 (s, 3H), 3-68 (s, 3H), 3-58 (d, J=3-i Hz, lH), 3.24 (br s, 3H), 3·ΐ8-3·07 (m, 2H), 2.44 (t, J=7-4 Hz, 2H), 2.08-1.99 (m, 2H), 1.77-1.68 (m, lH), 1.65-1.53 (m, 3H); MS (ES+): m/z = 744.7 (M+H) + ; LCMS (Method A): t R = 7.27 min.

Allyl (6aS)-3-(4-((5-((4-(5-((4-acetamidophenyl)carbamoyl)-i-methy l-ifi- pyrrol-3-yl)phenyl)carbamoyl)-i-methyl-iiT-pyrrol-3-yl)amino )-4-oxobut- oxy)-2-methoxy-i2-oxo-6-((tetrahydro-2JT-pyran-2-yl)oxy)-6,6 a,7,8,9,io- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine-5(i2H)-carboxyl ate (68)

A solution of 4-(4-(4-(4-(((6aS')-5-((allyloxy)carbonyl)-2-methoxy-i2-oxo- 6- ((tetrahydro-2ff-pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydr obenzo[e]pyrido[i,2- a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iff-pyrrole-2- carboxamido)phenyl)-i- methyl-iff-pyrrole-2-carboxylic acid (46) (100 mg, 0.117 mmol) in anhydrous dichloromethane (0.5 mL) was charged with /V-[(dimethylamino)-iif-i, 2, 3-triazolo- [4,5-b]pyridin-i-ylmethylene]-V-methylmethanaminiiim hexafluorophosphate V-oxide (47 mg, 0.124 mmol) and anhydrous triethylamine (69 pL, 0.496 mmol). The reaction mixture was stirred at room temperature for 15 min. L G - (4- A m i n 0 p h e nyl )aceta m i d e (18 mg, 0.120 mmol) was then added and the resulting mixture was stirred at room temperature for 1.5 h. The reaction mixture was directly loaded onto a cartridge (25 g) and purified by column chromatography (silica), eluting with methanol /

dichloromethane (from 0% to 100%), to give the title compound (113 mg, 98%) as a cream solid. MS (ES+): m/z = 985.9 (M+H) + ; LCMS (Method B): t R = 3.43 min.

(S)-/V-(4-Acetamidophenyl)-4-(4-(4-(4-((2-methoxy-i2-oxo- 6a,7,8,9,io,i2- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i- methyl-iJT-pyrrole-2-carboxamido)phenyl)-i-methyl-iJT-pyrrol e-2- carboxamide (69)

To a solution of allyl (6aS)-3-(4-((5-((4-(5-((4-acetamidophenyl)carbamoyl)-i-methy l- iff-pyrrol-3-yl)phenyl)carbamoyl)-i-methyl-iff-pyrrol-3-yl)a mino)-4-oxobutoxy)-2- methoxy-i2-oxo-6-((tetrahydro-2/7-pyran-2-yl)oxy)-6,6a,7,8,9 ,io-hexahydrobenzo[e]- pyrido[i,2-a][i,4]diazepine-5(i2ff)-carboxylate (68) (110 mg, 0.112 mmol) in dichloromethane (2 mL) was added tetrakis(triphenylphosphine)palladium(o) (6.5 mg, 5 mol%) and pyrrolidine (11 pL, 0.134 mmol). The reaction mixture was stirred at room temperature for 30 min. The reaction mixture was subjected to high vacuum for 30 min until excess pyrrolidine was thoroughly removed. The resulting residue was then purified by column chromatography (silica), eluting with methanol / dichloromethane (from 0% to 100%), to give the title compound (68 mg, 75%) as a cream solid. Ή NMR (400 MHz, DMSO-d 6 ) <59.91 (s, lH), 9.87 (s, lH), 9.79 (d, J= 5.5 Hz, 2H), 8.00 (d, J= 5.5 Hz, lH), 7.71 (d, J= 8.6 Hz, 2H), 7.64 (d, J= 9.4 Hz, 2H), 7.51 (t, J= 9.2 Hz, 4H), 7.46- 7.42 (m, lH), 7.37 (d, J= 2.0 Hz, lH), 7.27 (s, lH), 7.24-7.20 (m, lH), 6.97 (d, J=i.6 Hz, lH), 6.80 (s, lH), 4.09 (q, =5-i Hz, lH), 4-05-3-93 (m, 2H), 3.90 (s, 3H), 3-85-3-71 (m, 6H), 3.68 (s, lH), 3.25 (s, lH), 3.14-3.02 (m, lH), 2.45 (t, J=7- 2 Hz, 2H), 2.05 (d, J=7- o Hz, 2H), 2.03 (s, 3H), 1.92-1.81 (m, lH), 1.79-1.68 (m, 2H), 1.57 (dd, J= 5.5, 16.0 Hz, 2H); MS (ES+): m/z = 799.8 (M+H) + ; LCMS (Method A): t R = 6.37 min.

(S)-/V-(4-Acetamidophenyl)-4-(4-(4-(4-((2-methoxy-i2-oxo- 5,6,6a,7,8,9,io, i2-octahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)buta namido)-i- methyl-iii-pyrrole-2-carboxamido)phenyl)-i-methyl-iii-pyrrol e-2- carboxamide (70)

To a solution of GS)-iV-(4-acetamidophenyl)-4-(4-(4-(4-((2-methoxy-i2-oxo-6a, 7,8,9, io,i2-hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)b utanamido)-i-methyl- iff-pyrrole-2-carboxamido)phenyl)-i-methyl-iff-pyrrole-2-car boxamide (69) (63.5 mg, 0.0795 mmol) in tetrahydrofuran (3.5 mL) was sequentially added ammonium formate (41 mg, 0.650 mmol), water (350 pL) and Pd / C (10% w/w, 32 mg). The reaction mixture was heated at 35 °C for 4 h. The reaction mixture was filtered through Celite® and washed with ethyl acetate (too mL). The filtrate was concentrated in vacuo to give the title compound (40 mg, 63%) as a cream solid. Ή NMR (400 MHz, DMSO-d 6 ) d 9-90 (s, lH), 9.87 (s, lH), 9.79 (d, J= 4.7 Hz, 2H), 7.71 (d, J= 8.6 Hz, 2H), 7.64 (d, J= 9.0 Hz, 2H), 7.53 (s, lH), 7.51 (s, 2H), 7.50-7.48 (m, 2H), 7.44 (s, lH), 7.37 (s, lH), 7.22 (s, lH), 6.97 (s, lH), 6.37 (s, lH), 5.95 (br s, lH), 4.16-4.06 (m, 2H), 3.95 (t,

J= 6.2 Hz, 2H), 3.90 (s, 3H), 3-83 (s, 3H), 3-68 (s, 3H), 3-57 (d, =3-i Hz, lH), 3-26-3.21 (m, 2H), 3-15-3-05 (m, lH), 2.44 (t, J=7- 2 Hz, 2H), 2.05 (br s, lH), 2.03 (s, 3H), 1.78- 1.67 (m, lH), 1.66-1.53 (m, 3H), 1.51-1.38 (m, 2H); MS (ES+): m/z = 801.7 (M+H) + ; LCMS (Method A): t R = 6.53 min.

Allyl (6aS')-2-methoxy-3-(4-((i-methyl-5-((4-(i-methyl-5-(pyridin- 4- ylcarbamoyl)-iiT-pyrrol-3-yl)phenyl)carbamoyl)-iiT-pyrrol-3- yl)amino)-4- oxobutoxy)-i2-oxo-6-((tetrahydro-2JT-pyran-2-yl)oxy)-6,6a,7, 8,9,io- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine-5(i2H)-carboxyl ate (71)

A solution of 4-(4-(4-(4-(((6aS , )-5-((allyloxy)carbonyl)-2-methoxy-i2-oxo-6- ((tetrahydro-2/7-pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydr obenzo[e]pyrido[i,2- <3][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iii-pyrrol e-2-carboxamido)phenyl)-i- methyl-iif-pyrrole-2-carboxylic acid (46) (100 mg, 0.117 mmol) in anhydrous dichloromethane (0.5 mL) was charged with /V-[(dimethylamino)-iif-i, 2, 3-triazolo- [4,5-b]pyridin-i-ylmethylene]-iV-methylmethanaminium hexafluorophosphate V-oxide (47 mg, 0.124 mmol) and anhydrous triethylamine (69 pL, 0.496 mmol). The reaction mixture was stirred at room temperature for 15 min. 4-Aminopyridine (11 mg, 0.117 mmol) was then added and the resulting mixture was stirred at room temperature for 1.5 h. The reaction mixture was directly loaded onto a cartridge (25 g) and purified by column chromatography (silica), eluting with methanol / dichloromethane (from 0% to 100%), to give the title compound (105 mg, 97%) as a yellow oil. MS (ES+): m/z =

930.0 (M+H) + ; LCMS (Method B): t R = 3.00 min.

CS)-4-(4-((2-Methoxy-i2-oxo-6a,7,8,9,io,i2-hexahydrobenzo [e]pyrido[i,2- a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iV-(4-(i-methy l-5-(pyridin- 4-ylcarbamoyl)-iiT-pyrrol-3-yl)phenyl)-iJT-pyrrole-2-carboxa mide (72)

To a solution of allyl (6aS)-2-methoxy-3-(4-((i-methyl-5-((4-(i-methyl-5-(pyridin-4 - ylcarbamoyl)-iff-pyrrol-3-yl)phenyl)carbamoyl)-iii-pyrrol-3- yl)amino)-4-oxobutoxy)- i2-oxo-6-((tetrahydro-2ff-pyran-2-yl)oxy)-6,6a,7,8,9,io-hexa hydrobenzo[e]pyrido[i,2- <3][i,4]diazepine-5(i2ff)-carboxylate (71) (102 mg, 0.110 mmol) in dichloromethane (2 mL) was added tetrakis(triphenylphosphine)palladium(o) (6.4 mg, 5 mol%) and pyrrolidine (11 pL, 0.134 mmol). The reaction mixture was stirred at room temperature for 30 min. The reaction mixture was subjected to high vacuum for 30 min until excess pyrrolidine was thoroughly removed. The resulting residue was then purified by column chromatography (silica), eluting with methanol / dichloromethane (from 0% to 100%), to give the title compound (61 mg, 75%) as a cream solid. Ή NMR (400 MHz, DMSO-d 6 ) <5 10.15 (s, lH), 9.91 (s, lH), 9.81 (s, lH), 8.43 (d, J= 5.1 Hz, 2H), 8.00 (d,

J= 5-9 Hz, lH), 7-78-7-69 (m, 4H), 7-56-7-45 (m, 4H), 7-22 (s, lH), 7.14-7.06 (m, lH), 6.97 (s, lH), 6.54 (s, lH), 4.60-4.50 (m, lH), 4.09 (d, J=5-i Hz, lH), 4-03-3-94 (m, 2H),

3.92 (s, 3H), 3-83 (s, 3H), 3-73-3-65 (m, 3H), 3-47-3-39 (m, lH), 3.25 (s, lH), 2.48-2.41 (m, 2H), 2.04 (t, J= 6.8 Hz, 2H), 1.81-1.45 (m, 5H); MS (ES+): m/z = 743.7 (M+H) + ; LCMS (Method A): t R = 5.50 min. (S)-4-(4-((2-Methoxy-i2-oxo-5,6,6a,7,8,9,io,i2-octahydrobenz o[e]pyrido- [i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-JV-(4-(i- methyl-5- (pyridin-4-ylcarbamoyl)-iii-pyrrol-3-yl)phenyl)-iii-pyrrole- 2-carboxamide (73)

To a solution of 0S)-4-(4-((2-methoxy- 12-0x0-63,7,8,9, 10,12-hexahyd robenzo[e]pyrido- [i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-A/-(4-(i- methyl-5-(pyridin-4- ylcarbamoyl)-iff-pyrrol-3-yl)phenyl)-iff-pyrrole-2-carboxami de (57) (56.5 mg, 0.0761 mmol) in tetrahydrofuran (3.5 mL) was sequentially added ammonium formate (40 mg, 0.634 mmol), water (350 uL) and Pd / C (10% w/w, 30 mg). The reaction mixture was heated at 35 °C for 16 h. The reaction mixture was filtered through Celite® and washed with ethyl acetate (100 mL). The filtrate was concentrated in vacuo to give the title compound (33 mg, 58%) as a cream solid. Ή NMR (400 MHz, DMSO-d 6 ) d io.2i (s, lH), 9.90 (s, lH), 9.81 (s, lH), 8.45 (d, J= 5-5 Hz, 2H), 7 78 (d, J= 5-9 Hz, 2H), 7-72 (d, J=8.6 Hz, 2H), 7.56-7.47 (m, 5H), 7.22 (s, lH), 6.97 (s, lH), 6.37 (s, lH), 5.95 (br s, lH), 4.16-4.07 (m, lH), 3.97-3.94 (m, lH), 3.92 (s, 3H), 3 84 (s, 3H), 3-68 (s, 3H), 3-57 (d, J=3-9 Hz, lH), 3.23 (br s, 2H), 3-17 (br s, lH), 3.15-3.07 (m, lH), 2.44 (t, J=7- 2 Hz, 2H), 2.04 (quin, J= 6.6 Hz, 2H), 1.72 (d, J=7 -4 Hz, lH), 1.66-1.53 (m, 3H), 1.52-1.37 (m,

2H); MS (ES+): m/z = 745.7 (M+H) + ; LCMS (Method A): t R = 5.73 min.

Allyl (6aS)-2-methoxy-3-(4-((5-((4-(5-((5-(methoxycarbonyl)-i-meth yl-iJT- pyrrol-3-yl)carbamoyl)-i-methyl-iJT-pyrrol-3-yl)phenyl)carba moyl)-i- methyl-iJT-pyrrol-3-yl)amino)-4-oxobutoxy)-i2-oxo-6-((tetrah ydro-2JT- pyran-2-yl)oxy)-6,6a,7,8,9,io-hexahydrobenzo[e]pyrido[i,2- «][i,4]diazepine-5(i2Ji)-carboxylate (74)

A solution of 4-(4-(4-(4-(((6aS')-5-((allyloxy)carbonyl)-2-methoxy-i2-oxo- 6- ((tetrahydro-2if-pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydr obenzo[e]pyrido[i,2- <3][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iff-pyrrol e-2-carboxamido)phenyl)-i- methyl-iff-pyrrole-2-carboxylic acid (46) (100 mg, 0.117 mmol) in anhydrous dichloromethane (0.5 mL) was charged with /V-[(dimethylamino)-iif-i, 2, 3-triazolo- [4,5-b]pyridin-i-ylmethylene]-V-methylmethanaminiiim hexafluorophosphate V-oxide (47 mg, 0.124 mmol) and anhydrous triethylamine (69 pL, 0.496 mmol). The reaction mixture was stirred at room temperature for 15 min. Methyl 4-amino-i-methyl-iff- pyrrole-2-carboxylate hydrochloride (23 mg, 0.121 mmol) was then added and the resulting mixture was stirred at room temperature for 16 h. The reaction mixture was directly loaded onto a cartridge (25 g) and purified by column chromatography (silica), eluting with methanol / dichloromethane (from 0% to 100%), to give the title compound (102 mg, 88%) as a cream solid. MS (ES+): m/z = 990.0 (M+H) + ; LCMS (Method B): t R = 3.67 min.

Methyl (S)-4-(4-(4-(4-(4-((2-methoxy-i2-oxo-6a,7,8,9,io,i2- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i- methyl-iJT-pyrrole-2-carboxamido)phenyl)-i-methyl-iJT-pyrrol e-2- carboxamido)-i-methyl-iJT-pyrrole-2-carboxylate (75)

To a solution of allyl (6aS')-2-methoxy-3-(4-((5-((4-(5-((5-(methoxycarbonyl)-i-met hyl- iff-pyrrol-3-yl)carbamoyl)-i-methyl-iff-pyrrol-3-yl)phenyl)c arbamoyl)-i-methyl-iii- pyrrol-3-yl)amino)-4-oxobutoxy)-i2-oxo-6-((tetrahydro-2ff-py ran-2-yl)oxy)-6,6a,7,8, 9,io-hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine-5(i2ff)-ca rboxylate (74) (102 mg, 0.103 mmol) in dichloromethane (2 mL) was added tetrakis(triphenylphosphine)- palladium(o) (6.0 mg, 5 mol%) and pyrrolidine (11 pL, 0.134 mmol). The reaction mixture was stirred at room temperature for 15 min. The reaction mixture was subjected to high vacuum for 30 min until excess pyrrolidine was thoroughly removed. The resulting residue was then purified by column chromatography (silica), eluting with methanol / dichloromethane (from o% to 100%), to give the title compound (61 mg, 74%) as a cream solid. Ή NMR (400 MHz, DMSO-d 6 ) d 9.95 (s, lH), 9.91 (s, lH), 9.79 (s, lH), 8.00 (d, J= 5.9 Hz, lH), 7.71 (d, « 7=9.0 Hz, 2H), 7.48 (dd, J= 3.3, 5.3 Hz, 3H), 7-41 (d, J= 1.6 Hz, lH), 7.29-7.24 (m, lH), 7.22 (s, lH), 7.14-7.06 (m, lH), 6.97 (d, « 7=1.6 Hz, lH), 6.90 (d, « 7=1.6 Hz, lH), 4.62-4.45 (m, lH), 4.11-3.96 (m, 3H), 3.90 (s,

3H), 3-85 (s, 3H), 3-83 (s, 3H), 3-77-3-67 (m, 6H), 3-47-3-39 (m, lH), 3.17 (d, « 7=4-7 Hz, lH), 2.69 (s, lH), 2.48-2.41 (m, 2H), 2.05 (t, J= 6.8 Hz, 2H), 1.87 (t, « 7=6.4 Hz, lH), 1.80-1.69 (m, 2H), 1.64-1.50 (m, 2H); MS (ES+): m/z = 803.7 (M+H) + ; LCMS

(Method A): t R = 6.85 min.

Methyl (S)-4-(4-(4-(4-(4-((2-methoxy-i2-oxo-5,6,6a,7,8,9,io,i2- octahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i- methyl-iff-pyrrole-2-carboxamido)phenyl)-i-methyl-iJi-pyrrol e-2- carboxamido)-i-methyl-iii-pyrrole-2-carboxylate (76)

To a solution of methyl (S)-4-(4-(4-(4-(4-((2-methoxy-i2-oxo-6a,7,8,9,io,i2-hexa- hydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanamido) -i-methyl-iii-pyrrole- 2-carboxamido)phenyl)-i-methyl-i77-pyrrole-2-carboxamido)-i- methyl-ii7-pyrrole-2- carboxylate (75) (56 mg, 0.0697 mmol) in tetrahydrofuran (3.5 mL) was sequentially added ammonium formate (36 mg, 0.571 mmol), water (350 pL) and Pd / C (10% w/w, 30 mg). The reaction mixture was heated at 35 °C for 16 h. The reaction mixture was filtered through Celite® and washed with ethyl acetate (100 mL). The filtrate was concentrated in vacuo to give the title compound (45 mg, 80%) as a grey solid. Ή NMR (400 MHz, DMSO-d 6 ) d 9.95 (s, lH), 9.91 (s, lH), 9.80 (s, lH), 7.71 (d, J= 8.6 Hz, 2H),

7.51-7.45 (m, 4H), 7.41 (d, 7=1.6 Hz, lH), 7.26 (d, J= 2.0 Hz, lH), 7.22 (d, J= 2.0 Hz, lH), 6.96 (d, 7=1.6 Hz, lH), 6.90 (d, 7= 2.0 Hz, lH), 6.37 (s, lH), 5.95 (t, 7= 3.7 Hz, lH), 4.16-4.06 (m, lH), 3.95 (t, « 7=6.4 Hz, 2H), 3-90 (s, 3H), 3 85 (s, 3H), 3-83 (s, 3H), 3-75 (s, 3H), 3-68 (s, 3H), 3-57 (d, « 7=3-9 Hz, lH), 3.24 (d, « 7=4-3 Hz, 2H), 3-17 (d, « 7=5-1 Hz, lH), 3.14-3.07 (m, lH), 2.46-2.42 (m, 2H), 2.07-2.00 (m, 2H), 1.78-1.67 (m, lH), 1.65- 1-53 (m, 3H), 1.47-1.40 (m, lH); MS (ES+): m/z = 805.6 (M+H) + ; LCMS (Method A): t R = 7.02 min. Allyl (6aS)-2-methoxy-3-(4-((i-methyl-5-((4-(i-methyl-5-(pyridin-3 - ylcarbamoyl)-iff-pyrrol-3-yl)phenyl)carbamoyl)-iii-pyrrol-3- yl)amino)-4- oxobutoxy)-i2-oxo-6-((tetrahydro-2ff-pyran-2-yl)oxy)-6,6a,7, 8,9,io- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine-5(i2H)-carboxyl ate (77)

A solution of 4-(4-(4-(4-(((6aS')-5-((allyloxy)carbonyl)-2-methoxy-i2-oxo- 6-

((tetrahydro-2ff-pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octah ydrobenzo[e]pyrido[i,2-

<3][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iff-pyr role-2-carboxamido)phenyl)-i- methyl-iff-pyrrole-2-carboxylic acid (46) (100 mg, 0.117 mmol) in anhydrous dichloromethane (0.5 mL) was charged with /V-[(dimethylamino)-iif-i, 2, 3-triazolo- [4,5-b]pyridin-i-ylmethylene]-V-methylmethanaminium hexafluorophosphate V-oxide (47 mg, 0.124 mmol) and anhydrous triethylamine (69 pL, 0.496 mmol). The reaction mixture was stirred at room temperature for 15 min. 3-Aminopyridine (11 mg, 0.117 mmol) was then added and the resulting mixture was stirred at room temperature for 16 h. The reaction mixture was directly loaded onto a cartridge (25 g) and purified by column chromatography (silica), eluting with methanol / dichloromethane (from 0% to 100%), to give the title compound (110 mg, 99%) as a yellow oil. MS (ES+): m/z = 930.1 (M+H) + ; LCMS (Method B): t R = 3.15 min.

CS)-4-(4-((2-Methoxy-i2-oxo-6a,7,8,9,io,i2-hexahydrobenzo [e]pyrido[i,2- a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iV-(4-(i-methy l-5-(pyridin- 3-ylcarbamoyl)-iiT-pyrrol-3-yl)phenyl)-iJT-pyrrole-2-carboxa mide (78)

To a solution of allyl (6aS)-2-methoxy-3-(4-((i-methyl-5-((4-(i-methyl-5-(pyridin-3 - ylcarbamoyl)-iff-pyrrol-3-yl)phenyl)carbamoyl)-ifi-pyrrol-3- yl)amino)-4-oxobutoxy)- i2-oxo-6-((tetrahydro-2/7-pyran-2-yl)oxy)-6,6a,7,8,9,io-hexa hydrobenzo[e]pyrido[i,2- <2][i,4]diazepine-5(i2f/>carboxylate (77) (110 mg, 0.118 mmol) in dichloromethane (2 mL) was added tetrakis(triphenylphosphine)palladium(o) (7.0 mg, 5 mol%) and pyrrolidine (12 nL, 0.146 mmol). The reaction mixture was stirred at room temperature for 15 min. The reaction mixture was subjected to high vacuum for 30 min until excess pyrrolidine was thoroughly removed. The resulting residue was then purified by column chromatography (silica), eluting with methanol / dichloromethane (from 0% to 100%), to give the title compound (51 mg, 58%) as a cream solid. Ή NMR (400 MHz, DMSO-d 6 ) <5 10.04 (s, lH), 9.91 (s, lH), 9.81 (s, lH), 8.90 (d, J= 2.7 Hz, lH), 8.27 (dd, « 7=1.2, 4.7 Hz, lH), 8.15 (dd, J= 2.1, 8.8 Hz, lH), 8.00 (d, J= 5.9 Hz, lH), 7.72 (d, J= 8.6 Hz, 2H), 7.54-7.48 (m, 3H), 7.44 (d, « 7=2.0 Hz, lH), 7.37 (dd, « 7=4-7, 8.2 Hz, lH), 7.27 (s, lH), 7.25-7.18 (m, lH), 6.97 (d, 7=1.6 Hz, lH), 6.80 (s, lH), 4.15-4.04 (m, 2H), 3.92 (s, 3H), 3-83 (d, « 7=4-3 Hz, 6H), 3.73-3.66 (m, 2H), 3-17 (d, « 7=5-1 Hz, 2H), 2.45 (t, « 7=7-4 Hz, 2H), 2.11-1.96 (m, 3H), 1.91-1.82 (m, lH), 1.79-1.68 (m, 2H), 1.61-1.54 (m, lH); MS (ES+): m/z = 743.7 (M+H) + ; LCMS (Method A): t R = 5.72 min.

CS)-4-(4-((2-Methoxy-i2-oxo-5,6,6a,7,8,9,io,i2-octahydrob enzo[e]pyrido-

[i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-JV-(4- (i-methyl-5-

(pyridin-3-ylcarbamoyl)-iT7-pyrrol-3-yl)phenyl)-iT7-pyrro le-2-carboxamide

(79)

To a solution of 0S)-4-(4-((2-methoxy- 12-0x0-63,7, 8, 9, 10, i2-hexahydrobenzo[e]pyrido- [i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iV-(4-(i- methyl-5-(pyridin-3- ylcarbamoyl)-ii7-pyrrol-3-yl)phenyl)-i77-pyrrole-2-carboxami de (78) (46 mg, 0.0619 mmol) in tetrahydrofuran (3.5 mL) was sequentially added ammonium formate (32 mg, 0.507 mmol), water (350 pL) and Pd / C (10% w/w, 25 mg). The reaction mixture was heated at 35 °C for 5 h. The reaction mixture was filtered through Celite® and washed with ethyl acetate (100 mL). The filtrate was concentrated in vacuo to give the title compound (26 mg, 56%) as a cream solid. Ή NMR (400 MHz, DMSO-d 6 ) d 10.04 (s, lH), 9.91 (s, lH), 9.81 (s, lH), 8.90 (d, « 7= 2.3 Hz, lH), 8.27 (dd, « 7=1.6, 4.7 Hz, lH),

8.18-8.12 (m, lH), 7.72 (d, « 7=8.6 Hz, 2H), 7.52 (s, lH), 7-51-7-50 (m, lH), 7.49 (d, « 7=3.1 Hz, 2H), 7.44 (d, « 7=2.0 Hz, lH), 7.37 (dd, « 7= 4.7, 8.2 Hz, lH), 7.22 (d, « 7=1.6 Hz, lH), 6.97 (d, « 7=1.6 Hz, lH), 6.37 (s, lH), 5.95 (br s, lH), 4.16-4.08 (m, lH), 3.95 (t, « 7=6.4 Hz, 2H), 3-92 (s, 3H), 3-84 (s, 3H), 3-68 (s, 3H), 3-57 (d, =3-9 Hz, 2H), 3 2 3 (br s, 2H),

3.15-3.06 (m, 2H), 2.44 (t, J=7.2 Hz, 2H), 2.04 (quin, J= 6.7 Hz, 2H), 1.76-1.67 (m, lH), 1.62-1.54 (m, 2H), 1.47-1.41 (m, lH); MS (ES+): m/z = 745.7 (M+H) + ; LCMS (Method A): £ R = 5.83 min.

Allyl (6aS)-2-methoxy-3-(4-((5-((4-(5-((4-(methoxycarbonyl)phenyl) - carbamoyl)-i-methyl-iii-pyrrol-3-yl)phenyl)carbamoyl)-i-meth yl-iJi- pyrrol-3-yl)amino)-4-oxobutoxy)-i2-oxo-6-((tetrahydro-2Ji-py ran-2- yl)oxy)-6, 6a, 7,8,9, io-hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine- 5(i2f/)-carboxylate (80)

A solution of 4-(4-(4-(4-(((6aS')-5-((allyloxy)carbonyl)-2-methoxy-i2-oxo- 6- ((tetrahydro-2ff-pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydr obenzo[e]pyrido[i,2- a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iff-pyrrole-2- carboxamido)phenyl)-i- methyl-iff-pyrrole-2-carboxylic acid (46) (100 mg, 0.117 mmol) in anhydrous dichloromethane (0.5 mL) was charged with /V-[(dimethylamino)-iif-i, 2, 3-triazolo- [4,5-b]pyridin-i-ylmethylene]-V-methylmethanaminium hexafluorophosphate V-oxide (47 mg, 0.124 mmol) and anhydrous triethylamine (69 pL, 0.496 mmol). The reaction mixture was stirred at room temperature for 15 min. Methyl 4-aminobenzoate (18 mg, 0.119 mmol) was then added and the resulting mixture was stirred at room temperature for 16 h. The reaction mixture was directly loaded onto a cartridge (25 g) and purified by column chromatography (silica), eluting with methanol / dichloromethane (from 0% to 100%), to give the title compound (103 mg, 89%) as a yellow solid. MS (ES+): m/z = 986.9 (M+H) + ; LCMS (Method B): t R = 3.77 min.

Methyl (S)-4-(4-(4-(4-(4-((2-methoxy-i2-oxo-6a,7,8,9,io,i2- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i- methyl-iJT-pyrrole-2-carboxamido)phenyl)-i-methyl-iJT-pyrrol e-2- carboxamido)benzoate (81)

To a solution of allyl (6aS')-2-methoxy-3-(4-((5-((4-(5-((4-(methoxycarbonyl)phenyl )- carbamoyl)-i-methyl-iff-pyrrol-3-yl)phenyl)carbamoyl)-i-meth yl-iff-pyrrol-3- yl)amino)-4-oxobutoxy)-i2-oxo-6-((tetrahydro-2ff-pyran-2-yl) oxy)-6,6a,7,8,9,io- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine-5(i2ff)-carboxy late (80) (103 mg, 0.104 mmol) in dichloromethane (2 mL) was added tetrakis(triphenylphosphine) palladium(o) (6.0 mg, 5 mol%) and pyrrolidine (10 pL, 0.122 mmol). The reaction mixture was stirred at room temperature for 20 min. The reaction mixture was subjected to high vacuum for 30 min until excess pyrrolidine was thoroughly removed. The resulting residue was then purified by column chromatography (silica), eluting with methanol / dichloromethane (from 0% to 100%), to give the title compound (28 mg, 34%) as a bright yellow solid. MS (ES+): m/z = 800.7 (M+H) + ; LCMS (Method B): t R = 3-37 min.

Methyl (S)-4-(4-(4-(4-(4-((2-methoxy-i2-oxo-5,6,6a,7,8,9,io,i2- octahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i- methyl-iJT-pyrrole-2-carboxamido)phenyl)-i-methyl-iJT-pyrrol e-2- carboxamido)benzoate (82)

To a solution of methyl (S)-4-(4-(4-(4-(4-((2-methoxy-i2-oxo-6a,7,8,9,io,i2- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i-methyl-iff- pyrrole-2-carboxamido)phenyl)-i-methyl-iff-pyrrole-2-carboxa mido)benzoate (81) (28 mg, 0.0350 mmol) in tetrahydrofuran (3.5 mL) was sequentially added ammonium formate (18 mg, 0.285 mmol), water (350 pL) and Pd / C (10% w/w, 15 mg). The reaction mixture was heated at 35 °C for 3 h. The reaction mixture was filtered through

Celite® and washed with ethyl acetate (100 mL). The filtrate was concentrated in vacuo. The resulting residue was loaded onto a C18 samplet and purified by reverse- phase flash column chromatography (Biotage, 30 g cartridge) eluting with 0.1% formic acid in water / 0.1% formic acid in acetonitrile (95/5 to 5/95, and then back to 95/5), to give the title compound (8.7 mg, 31%) as an off-white solid. Ή NMR (400 MHz, DMSO-d 6 ) d 10.15 (s, lH), 9.91 (s, lH), 9.81 (s, lH), 7.97-7.89 (m, 4H), 7.72 (d, J= 8.6 Hz, 2H), 7·54-7·45 (m, 5H) , 6.97 (d, J= 2.0 Hz, lH), 6.37 (s, lH), 5-95 (L J=3-7 Hz, lH), 4.12 7-3·93 (m, 2H), 3.92 (s, 3H), 3 84 (s, 6H), 3.68 (s, 3H), 3-57 (d, J=3-5 Hz, lH), 3·25-3·22 (m, 2H), 3.15-3.08 (m, lH), 2.44 (t, J=7-4 Hz, 2H), 2.04 (t, J= 6.8 Hz, 2H), 1.77-1.66 (m, lH), 1.66-1.53 (m, 3H), 1.51 (br s, 2H); MS (ES+): m/z = 802.7 (M+H) + ; LCMS (Method A): t R = 7.28 min.

(S) -Methyl 4-(4-(2-(hydroxymethyl)pyrrolidine-i-carbonyl)-2-methoxy-5- nitrophenoxy)butanoate (83)

A catalytic amount of N,/V- dimethyl formamide (2 drops) was added to a solution of oxalyl chloride (16.8 mL, 195 mmol) and methyl (S)-4-(4-(2-(hydroxymethyl)- piperidine-i-carbonyl)-2-methoxy-5-nitrophenoxy)butanoate (4) (20.5 g, 65.4 mmol) in anhydrous dichloromethane (350 mL) in a round bottom flask. The reaction mixture was stirred at room temperature for 16 h. The resulting acid chloride solution was added dropwise to a solution of anhydrous triethylamine (20.0 mL, 144 mmol) and (S)- pyrrolidin-2-ylmethanol (7.3 mL, 72.2 mmol) in anhydrous dichloromethane (150 mL) at - 30 °C. The reaction mixture was stirred for 4 h. The reaction mixture was washed sequentially with 1 N HC1 (2 x 150 mL), water (2 x 120 mL) and brine (too mL). The combined organic extracts were dried over magnesium sulfate, filtered and

concentrated in vacuo to give the title compound (15 g, 58%) as a cream solid. The product was carried through to the next step without any further purification. Ή NMR (400 MHz, DMSO-de) d 7-67 (s, lH), 6.76 (s, lH), 4.13 (t, J= 4.4 Hz, 2H), 3.93 (s, 3H), 3.84 (m, lH), 3.69 (m, lH), 3.65 (s, 3H), 3-14 (L J= 6-8 Hz, 2H), 2.53 (t, J= 4.8 Hz, 2H), 2.17 (m, 3H), 1.86 (m, 3H), 1.56 (m, 2H); (too MHz, CDCl 3 ): d 173-2, 154-8, 148.4,

109-2, IO8.4, 68.4, 66.1, 61.5, 56.7, 51-7, 49-5, 30-3, 28.4, 24.4, 24.2; MS (ES+): m/z = 397.1 (M+H) + .

(N) -Methyl 4-(5-amino-4-(2-(hydroxymethyl)pyrrolidine-i-carbonyl)-2- methoxyphenoxy)butanoate (84)

A slurry of Pd/C (10% w/w) in ethyl acetate was added to a solution of (S)- methyl 4-(4- (2-(hydroxymethyl)pyrrolidine-i-carbonyl)-2-methoxy-5-nitrop henoxy)butanoate (83) (15 g , 37-8 mmol) in ethanol (140 mL). The reaction mixture was hydrogenated in a Parr hydrogenator at 45 psi for 3 h. The reaction mixture was filtered under vacuum through a pad of Celite® and was washed with ethyl acetate (300 mL). The filtrate was concentrated in vacuo to give the title compound (12.6 g, 91%) as a red foam. The product was carried through to the next step without any further purification. Ή NMR (400 MHz, CDCI 3 ) d 6.71 (s, lH), 6.15 (s, lH), 5.67 (s, lH), 4.51 (br s, lH), 4.11 (t, J= 4.4 Hz, 2H), 3-82 (s, 3H), 3.78 (m, lH), 3.56 (m, lH), 2.65 (t, J=4-8 Hz, 2H), 2.17 (m, 3H), 1.84 (m, 3H), 1.53 (m, 2H); (100 MHz, CDCl 3 ): d 172.5, 170-7, 150-3, 140.5, 140.1, 135-0, 112.3, HO.5, 101.2, 66.5, 59-9, 56.3, 52.4, 50.6, 29.4, 27.5, 23.9, 23.4; MS (ES+): m/z = 367.3 (M+H) + . (S)-Methyl 4-(5-(allyloxycarbonylamino)-4-(2-(hydroxymethyl)- pyrrolidine-i-carbonyl)-2-methoxyphenoxy)butanoate (85)

A solution of allyl chloroformate (2.6 mL, 24.4 mmol) in anhydrous dichloromethane (175 mL) was added dropwise to a solution of (S)-methyl 4-(5-amino-4-(2-(hydroxy- methyl)pyrrolidine-i-carbonyl)-2-methoxyphenoxy)butanoate (84) (8.5 g, 23.3 mmol) and anhydrous pyridine (4.3 mL, 53.2 mmol) in anhydrous dichloromethane (250 mL) at - io°C. The reaction mixture was stirred from - 10 °C to room temperature over 2 h. The reaction mixture was washed with a saturated aqueous solution of copper sulfate (II) (200 mL), water (200 mL), a saturated aqueous solution of sodium hydrogen carbonate (200 mL), and brine (200 mL). The combined organic extracts were dried over magnesium sulfate, filtered and concentrated in vacuo. The resulting residue was then purified by column chromatography (silica), eluting with ethyl acetate/n-hexane (0% to 100%), to give the title compound (9.8 g, 94%) as a cream solid. Ή NMR (400 MHz, CDCI 3 ) d 8.72 (s, lH), 7.75 (s, lH), 6.83 (s, lH), 5.95 (m, lH), 5.34 (dd, J=i7-2, 1.2 Hz, lH), 5.23 (dd, J=io.o, 0.8 Hz, lH), 4.62 (dd, J= 5.6, 1.2 Hz, 2H), 4.40 (br s, lH), 4,23 (br s, lH), 4.08 (t, J= 4.4 Hz, 2H), 3.81 (s, 3H), 3.67 (s, 3H), 3.56 (m, lH), 3.50 (m, lH), 2.54 (t, J= 4.8 Hz, 2H), 2.16 (m, 4H), 1.88 (m, lH), 1.69 (m, 3H); (100 MHz, CDCl 3 ): d 173-4, 170-9, 153-6, 150.5, 144-0, 132-3, 131-9, n8-2, 115.7, m-6, 105.6, 67.7, 66.6, 65.7, 61.6, 60.4, 56.6, 51.7, 30.7, 28.3, 25.1, 24.3; MS (ES+): m/z = 451.2 (M+H) + .

Allyl ii-hydroxy-7-methoxy-8-(4-methoxy-4-oxobutoxy)-5-oxo-2,3,ii, ua- hexahydro-iii-pyrrolo[2,i-c][i,4]benzodiazepine-io(5ii)-carb oxylate (86)

TEMPO (357 mg, 2.17 mmol) was added to a solution of (S)- methyl 4-(5-(allyloxy- carbonylamino)-4-(2-(hydroxymethyl)pyrrolidine-i-carbonyl)-2 -methoxyphenoxy)- butanoate (85) (9.8 g, 21.8 mmol) and diacetoxyiodobenzene (8.4 g, 26.1 mmol) in dichloromethane (500 mL). The reaction mixture was stirred at room temperature for 6 h. The reaction mixture was sequentially washed with a saturated aqueous solution of sodium metabisulphite (200 mL), a saturated aqueous solution of sodium hydrogen carbonate (2 x 200 mL), water (200 mL) and brine (200 mL). The combined organic extracts were dried over magnesium sulfate, filtered and concentrated in vacuo. The resulting residue was then purified by column chromatography (silica), eluting with ethyl acetate/n-hexane (0% to 100%), to give the title compound (6.7 g, 94%) as a cream solid. Ή NMR (400 MHz, CDCl 3 ) d 7.19 (s, lH), 6.66 (s, lH), 5.74 (m, lH), 5.59 (d, J=4.o Hz, lH), 5.07 (d, J=i2.o Hz, 2H), 4.61 (dd, =i3-2, 5.6 Hz, lH), 4.41 (d,

J=i2.o Hz, 2H), 3.98 (m, 2H), 3.84 (s, 3H), 3-62 (s, 3H), 3-49 (t, J= 8.0 Hz, lH), 3.43 (m, lH), 2.475 (L J=7- 2 Hz, 2H), 2.07 (m, 4H), 1.93 (m, 2H); (100 MHz, CDCl 3 ): d

173-4, 167-0, 155-9, 149-9, 148.7, 131-8, 128.3, 126.0, 117.9, H4-2, 110.8, 85.9, 67.9, 66.7, 60.3, 60.1, 56.1, 51.6, 46.3, 30.3, 28.7, 24.2, 23.0, 20.9; MS (ES+): m/z = 449.2 (M+H) + . Allyl (uaS)-7-methoxy-8-(4-methoxy-4-oxobutoxy)-5-oxo-ii-((tetrahy dro- 2//-pyran-2-yl)oxy)-2,3,ii,iia-lelrahydro-i//-benzo| e |pyrrolo| 1,2- «][i,4]diazepine-io(5ii)-carboxylate (87)

A mixture of allyl ii-hydroxy-7-methoxy-8-(4-methoxy-4-oxobutoxy)-5-oxo-2,3,n,i ia- hexahydro-iff-pyrrolo[2,i-c][i,4]benzodiazepine-io(5if)-carb oxylate (86) (323 mg, 0.720 mmol), 3,4-dihydro-2if-pyran (660 pL, 7.22 mmol) and -toluenesulfonic acid monohydrate (3.2 mg, 1% w/w) in ethyl acetate (5 mL) was stirred at room temperature for 2 h. The reaction mixture was then diluted with ethyl acetate (too mL) and washed with a saturated aqueous solution of sodium hydrogen carbonate (30 mL) and brine (50 mL). The organic layer was dried over sodium sulfate, filtered and concentrated in vacuo. The resulting residue was purified by column chromatography (silica), eluting with methanol/dichloro methane (from 0% to 10%), to give the title compound (380 mg, 99%) as a yellow gum. MS (ES+): m/z = 533.4 (M+H) + ; LCMS (Method B): t R =

3.32 min.

4-(((naS)-io-((Allyloxy)carbonyl)-7-methoxy-5-oxo-ii-((te trahydro-2JT- pyran-2-yl)oxy)-2,3,5,io,n,iia-hexahydro-iiT-benzo[e]pyrrolo [i,2- a][i,4]di

To a solution of allyl (iiaS)-7-methoxy-8-(4-methoxy-4-oxobutoxy)-5-oxo-n- ((tetrahydro-2if-pyran-2-yl)oxy)-2,3,ii,na-tetrahydro-iif-be nzo[e]pyrrolo[i,2- <2][i,4]diazepine-io(5if)-carboxylate (87) (380 mg, 0.713 mmol) in 1,4-dioxane (10 mL) was added a 1 M aqueous solution of sodium hydroxide (10 mL, 10.0 mmol). The reaction mixture was stirred at room temperature for 1 h and was then concentrated in vacuo, after which water (20 mL) was added and the aqueous layer was acidified to pH = 1 with an aqueous solution of citric acid (1 M, 5 mL). The aqueous layer was then extracted with ethyl acetate (2 x 50 mL). The combined organic extracts were then washed with brine (50 mL), dried over sodium sulfate, filtered and concentrated to give the title compound (250 mg, 56%) as a cream solid. The product was carried through to the next step without any further purification. MS (ES+): m/z = 519.4 (M+H) + ; LCMS (Method 6.38 min.

Allyl (iiaS , )-7-methoxy-8-(4-((5-((4-(5-(methoxycarbonyl)-i-methyl -iJT- pyrrol-3-yl)phenyl)carbamoyl)-i-methyl-iiT-pyrrol-3-yl)amino )-4-oxo- butoxy)-5-oxo-ii-((tetrahydro-2JT-pyran-2-yl)oxy)-2,3,ii,na- tetrahydro- iiT-benzo[e]pyrrolo[i,2-a][i,4]diazepine-io(5ii)-carboxylate (89)

A solution of 4-(((iiaS)-io-((allyloxy)carbonyl)-7-methoxy-5-oxo-n-((tetra hydro-2ff- pyran-2-yl)oxy)-2,3,5,io,ii,na-hexahydro-iff-benzo[e]pyrrolo [i,2-a][i,4]diazepin-8- yl)oxy)butanoic acid (88) (too mg, 0.193 mmol) in anhydrous dichloromethane (0.5 mL) was charged with A/-[(dimethylamino)-iff-i,2,3-triazolo-[4,5-b]pyridin-i- yl met hyl e n e] -L G - m et hyl m et ha n a m i n i 11 m hexafluorophosphate V-oxide (77 mg, 0.203 mmol) and anhydrous triethylamine (113 uL, 0.203 mmol). The reaction mixture was stirred at room temperature for 15 min. Methyl 4-(4-(4-amino-i-methyl-iif-pyrrole-2- carboxamido)phenyl)-i-methyl-iff-pyrrole-2-carboxylate hydrochloride (33) (75 mg, 0.193 mmol) was then added and the resulting mixture was stirred at room

temperature for 4 h. The reaction mixture was quenched with a saturated aqueous solution of sodium hydrogen carbonate (20 mL) and extracted with dichloromethane (2 x 50 mL). The combined organic extracts were washed with water containing a few drops of acetic acid (30 mL). The organic layer was then dried over sodium sulfate, filtered and concentrated in vacuo. The resulting residue was then purified by column chromatography (silica), eluting with methanol/ dichloro methane (from 0% to 100%), to give the title compound (138 mg, 84%) as a brown oil. MS (ES+): m/z = 853.7 (M+H) + ; LCMS (Method B): t R = 3.53 min. Methyl (S)-4-(4-(4-(4-((7-methoxy-5-oxo-2,3,5,na-tetrahydro-iiT- benzo[e]pyrrolo[i,2-a][i,4]diazepin-8-yl)oxy)butanamido)-i-m ethyl-iiT- pyrrole-2-carboxamido)phenyl)-i-methyl-iiT-pyrrole-2-carboxy late (90)

To a solution of allyl (naS)-7-methoxy-8-(4-((5-((4-(5-(methoxycarbonyl)-i-methyl- iff-pyrrol-3-yl)phenyl)carbamoyl)-i-methyl-iff-pyrrol-3-yl)a mino)-4-oxobutoxy)-5- oxo-ii-((tetrahydro-2ff-pyran-2-yl)oxy)-2,3,ii,na-tetrahydro -iii-benzo[e]pyrrolo[i,2- <3][i,4]diazepine-io(5ff)-carboxylate (89) (48 mg, 0.0563 mmol) in dichloro methane (2 mL) was added tetrakis(triphenylphosphine)palladium(o) (3.3 mg, 5 mol%) and pyrrolidine (5.6 LIL, 0.0682 mmol). The reaction mixture was stirred at room temperature for 15 min. The reaction mixture was subjected to high vacuum for 30 min until excess pyrrolidine was thoroughly removed. The resulting residue was then purified by column chromatography (silica), eluting with methanol/dichloromethane (from 0% to 10%), to give the title compound (27 mg, 72%) as a cream solid. Ή NMR (400 MHz, DMSO-d 6 ) <59.91 (s, lH), 9.79 (s, lH), 7.68 (d, J= 8.6 Hz, 2H), 7.55 (d, J= 2.0 Hz, lH), 7.51 (d, J= 8.6 Hz, 2H), 7.26-7.18 (m, 3H), 6.96 (d, J=i.6 Hz, lH), 3.96 (d,

J= 6.6 Hz, lH), 3.89 (s, 3H), 3-83 (s, 3H), 3-77 (s, 3¾, 3-72 (s, lH), 3.69-3-62 (m, 3H), 3.61-3.54 (m, lH), 3-53-3-38 (m, 2H), 3-24-3-21 (m, 2H), 3.17 (d, J=5-i Hz, lH), 2.44 (t, J=7.2 Hz, 2H), 2.05 (t, J= 6.4 Hz, 2H), 2.00-1.79 (m, 3H); MS (ES+): m/z = 667.6

(M+H) + ; LCMS (Method A): t R = 6.57 min.

Methyl (S)-4-(4-(4-(4-((7-methoxy-5-oxo-2,3,5,io,ii,na-hexahydro-ii T- benzo[e]pyrrolo[i,2-a][i,4]diazepin-8-yl)oxy)butanamido)-i-m ethyl-iiT- pyrrole-2-carboxamido)phenyl)-i-methyl-iiT-pyrrole-2-carboxy late (91)

To a solution of methyl ( < S)-4-(4-(4-(4-((7-methoxy-5-oxo-2,3,5,iia-tetrahydro-i //- benzo[e]pyrrolo[i,2-a][i,4]diazepin-8-yl)oxy)butanamido)-i-m ethyl-iii-pyrrole-2 carboxamido)phenyl)-i-methyl-iff-pyrrole-2-carboxylate (90) (25 mg, 0.0375 mmol) in tetrahydrofuran (3.5 mL) was sequentially added ammonium formate (19 mg, 0.301 mmol), water (350 pL) and Pd / C (10% w/w , 13 mg). The reaction mixture was heated at 35 °C for 16 h. On completion, the reaction mixture was filtered through Celite® and washed with ethyl acetate (100 mL). The filtrate was concentrated under reduced pressure to give the title compound (11.6 mg, 46%) as an off-white solid. Ή NMR (400 MHz, DMSO-de) d 9-90 (s, lH), 9.79 (s, lH), 7.68 (d, J=8.6 Hz, 2H), 7-56 (d, J= 2.0 Hz, lH), 7.51 (d, J=9.o Hz, 2H), 7.34 (s, lH), 7.21 (dd, J= 2.0, 7.4 Hz, 2H), 6.95 (d, J= 2.0 Hz, lH), 6.27-6.23 (m, 2H), 3-93 (t, J= 6.2 Hz, 2H), 3 89 (s, 3H), 3-83 (s, 3H), 3-77 (s, 3H), 3-65 (s, 3H), 3-64 (s, lH), 3.59 (dt, 7=3.7, 7-9 Hz, lH), 3-49-3-41 (m, 2H), 3-23-3-15 (m, lH), 3.04-2.95 (m, lH), 2.46-2.40 (m, 4H), 2.18 (s, lH), 2.07-2.00 (m, 2H); MS

(ES+): m/z = 669.5 (M+H) + ; LCMS (Method A): t R = 6.83 min. 0S)-(2-(Hydroxymethyl) indolin-i-yl) (5-methoxy-2-nitro-4- ((triisopropylsilyl)oxy)phenyl)methanone (92)

To a solution of 5-methoxy-2-nitro-4-((triisopropylsilyl)oxy)benzoic acid (21) (1.00 g, 2.71 mmol) in dichloromethane (25 mL) were added CS)-indolin-2-yl methanol (404 mg,

2.71 mmol), /V-[(dimethylamino)-ii/-i,2,3-triazolo-[4,5-¾]pyridin-i-ylm ethylene]-iV- methylmethanaminium hexafluorophosphate -oxide (1.54 g, 4.06 mmol) and triethyl- amine (685 mg, 6.77 mmol). The reaction mixture was stirred at room temperature for 3 h. Then diluted with water (too mL) and extracted with dichloromethane (2 x too mL). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was then purified by column chromatography (silica), eluting with ethyl acetate/petroleum ether (from 0% to 100%), to give the title compound (800 mg, 58%) as a yellow oil. Ή NMR (400 MHz, CDCI 3 ) d 7.22-7.14 (m, lH), 7.08-7.00 (m, lH), 6.95-6.90 (m, lH), 6.80-6.70 (m, lH), 5-69-5-65 (m, lH), 5.23 - 5.06 (m, lH), 4.00-3.82 (m, 3H), 2.80 (s, 5H), 2.04 (s, lH),

1.34-1.25 (m, 3H), 1.15-1.10 (m, 18H); MS (ES+): m/z = 501.0 (M+H) + ; LCMS

(Method B): t R = 4.44 min.

0S)-(2-Amino-5-methoxy-4-((triisopropylsilyl)oxy)phenyl)( 2- (hydroxymethyl)indolin-i-yl)methanone (93)

To a solution of (S)-(2-(hydroxymethyl)indolin-i-yl)(5-methoxy-2-nitro-4- ((triisopropylsilyl)oxy) phenyl)methanone (92) (800 mg, 1.60 mmol) in methanol (10 mL) was added Pd/C (80 mg). The mixture was stirred at room temperature under hydrogen atmosphere for 16 h. The reaction mixture was filtered through Celite® and the cake was washed with ethyl acetate (50 mL). The filtrate was concentrated to dryness under reduced pressure. The resulting residue was then purified by column chromatography (silica), eluting with ethyl acetate/petroleum ether (from 20% to 50%), to give the title compound (500 mg, 66%) as a yellow oil. Ή NMR (400 MHz, DMSO- d6) <57.2.2. (d, J-= 6.8 Hz, lH), 7.08 (s, lH), 7.00-6.93 (m, 2H), 6.75 (s, lH), 6.37 (d,

J= 2-8 Hz, lH), 4.98-4.88(01, 3H), 4-61-4-57 (m, lH), 3.58 (s, 3H), 3.47-3.44 (m, lH), 3.32-3.26 (m, lH), 3.01-2.97 (m, lH), 2.69 (s, lH), 1.27-1.21 (m, 3H), 1.08 (d , 7=7.2 Hz, 18H); MS (ES+): m/z = 471.3 (M+H) + ; LCMS (Method B): t R = 2.98 min.

Allyl 0S)-(2-(2-(hydroxymethyl)indoline-i-carbonyl)-4-methoxy-5- ((triisopropylsilyl) oxy)phenyl)carbamate (94)

To a solution of (S)-(2-amino-5-methoxy-4-((triisopropylsilyl)oxy)phenyl)(2-( hydroxyl- methyl)indolin-i-yl)methanone (93) (470 mg, 1.00 mmol) in dichloromethane (10 mL) at - 10 °C were added anhydrous pyridine (158 uL, 2.0 mmol) and allyl chloroformate (127 uL, 1.05 mmol). After 30 min, the reaction mixture was diluted with

dichloromethane (100 mL) and washed with a saturated aqueous solution of copper (II) sulfate (100 mL), water (100 mL) and a saturated aqueous solution of sodium bicarbonate (10 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was then purified by column chromatography (silica), eluting with ethyl acetate/petroleum ether (5% isocratic), to give the title compound (400 mg, 72%) as a yellow oil. Ή NMR (400 MHz, CDCI 3 ) d 8.36 (s, lH), 7.74 (s, lH), 7.19 (d, J=j.2 Hz, lH), 6.97-6.87 (m,

2H), 6.72 (s, lH), 6.40 (s, lH), 5-98-5-87 (m, lH), 5.31 (d, J=i6.8 Hz, lH), 5.22 (d,7=10.4 Hz, lH), 4.94-4.91 (m, lH), 4.60 (d, J= 5.6 Hz, 2H), 3.76 (d, J= 6.0 Hz, 2H), 3.54

(s, 3H), 3.45-3.38 (m, lH), 2.81-2.76 (m, lH), 1.35- 1.28 (m, 3H), 1.12 (d, J=7-6 Hz, 18H); MS (ES+): m/z 555.4 (M+H) + ; LCMS (Method B): t R = 2.79 min.

Allyl (i2a-S)-i2-hydroxy-8-methoxy-6-oxo-9-((triisopropylsilyl)oxy )-i2a,i3- dihydro-6ff-benzo[5,6][i,4]diazepino[i,2-a]indole-ii(i2Ji)-c arboxylate (95)

To a solution of allyl (S')-(2-(2-(hydroxymethyl)indoline-i-carbonyl)-4-methoxy-5- ((triisopropylsilyl)oxy)phenyl)carbamate (94) (391 mg, 0.71 mmol) in dichloromethane (13 mL) were added TEMPO (11 mg, 0.07 mmol) and (diacetoxyiodo)benzene (274 mg, 0.85 mmol). The reaction mixture was stirred at room temperature for 18 h, then diluted with dichloromethane (40 mL) and washed with a saturated aqueous solution of sodium sulfite (10 mL), saturated aqueous solution of sodium bicarbonate (10 mL) and brine (10 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was then purified by column chromatography (silica), eluting with ethyl acetate/petroleum ether (from 0% to 10%), to give the title compound (290 mg, 74%) as a yellow oil. Ή NMR (500 MHz, CDCI 3 ) d 8.19 (d, J=8.o Hz, lH), 7.21 (d, J=7- 5 Hz, lH), 7.08 (t, J=7- 5 Hz, lH), 6.71 (s, lH), 5.78 (s, lH), 5.73 (d, J=io.o Hz, lH), 5.20-5.14 (m, 2H), 4.62-4.58 (m, lH), 4.46 (s, lH), 4.15-4.06 (m, 2H), 3.86-3.84 (m, 3H), 3·49-3·43 (m, lH), 3.21 (d, J=i7.o Hz, lH), 2.04 (d, J =2.0 Hz, lH), 1.28-1.21 (m, 3H), 1.09-1.08 (m, 18H); MS (ES+): m/z 553.3 (M+H) + ; LCMS (Method B): t R = 2.68 min.

Allyl (i2aS')-8-methoxy-6-oxo-i2-((tetrahydro-2JT-pyran-2-yl)oxy)- 9- ((triisopropylsilyl)oxy)-i2a,i3-dihydro-6iT-benzo[5,6][i,4]d iazepino[i,2- a]indole-ii(i2ii)-carboxylate (96)

To a solution of allyl (i2aS)-i2-hydroxy-8-methoxy-6-oxo-9-((triisopropylsilyl)oxy) - i2a,i3-dihydro-6H-benzo[5,6][i,4]diazepino[i,2-a]indole-n(i2 H)-carboxylate (95) (282 mg, 0.51 mmol) in tetrahydrofuran (5 mL) were added dihydropyran (429 mg, 5.1 mmol) and -toluenesulfonic acid (3 mg, 1% w/w). The reaction mixture was stirred at room temperature for 18 h, then diluted with ethyl acetate (30 mL) and washed with a saturated aqueous solution of sodium bicarbonate (10 mL) and brine (10 mL). After the extraction, the combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was then purified by column chromatography (silica), eluting with ethyl acetate/petroleum ether (from 0% to 20%), to give the title compound (300 mg, 92%) as a colourless oil. Ή NMR (500 MHz, CDCI 3 ) d 8.20-8.13 (m, lH), 7.25-7.21 (m, 2H), 7.08-6.61 (m, 2H), 5-94 (dd, J= 64.5, 9-5 Hz, lH), 5.78-5-66 (m, lH), 5-13-5-04 (m, 2H), 4.96-4.94(1^1, 2H), 4.89 (d, J=6.o Hz, lH), 3.86 (d, J= 2.0 Hz, 3H), 3.65-3.61 (m, lH), 3.47-3.42 (m, lH), 2.07-2.01 (m, lH), 1.98-1.95 (m, lH), 1.79-1.73 (m, 6H), 1.33-1.26 (m, 3H), 1.11-1.08 (m,

18H); MS (ES+): m/z 637.0 (M+H) + ; LCMS (Method B): t R = 4.43 min. Allyl (i2a < S)-9-hydroxy-8-methoxy-6-oxo-i2-((tetrahydro-2H-pyran- 2- yl)oxy)-i2a,i3-dihydro-6ii-benzo[5,6][i,4]diazepino[i,2-a]in dole-ii(i2Ji)- carboxylate

To a solution of allyl (i2aS')-8-methoxy-6-oxo-i2-((tetrahydro-2ff-pyran-2-yl)oxy)- 9- ((triisopropylsilyl)oxy)-i2a,i3-dihydro-6ff-benzo[5,6][i,4]d iazepino[i,2-a]indole- n(i2ff)-carboxylate (96) (292 mg, 0.46 mmol) in tetrahydrofuran (5 mL) under inert atmosphere were added tetra-n-butylammonium fluoride solution lM in tetrahydro- furan (0.65 mL, 0.65 mmol). The mixture was stirred at room temperature for 1 h and quenched with water (10 mL), extracted with ethyl acetate (30 mL) and washed with brine (10 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was then purified by column chromatography (silica), eluting with ethyl acetate/petroleum ether (from 0% to 20%), to give the title compound (too mg, 45%) as a white solid. Ή NMR (500 MHz, CDCI 3 ) d 8.i6 (dd, J=2i.o, 8.0 Hz, lH), 7.28 (d, J= 5.0 Hz, lH), 7.25-7.20 (m, lH), 7.10-7.05 (m, lH), 6.77 (d, =37-5 Hz, lH), 6.02 (s, lH), 5·8i-5·72 (m, lH), 5.20-5.14 (m, lH), 5.13-4.84 (m, lH), 4.66-4.48 (m, 2H), 4-15-4-07 (m, lH), 3.95 (s, 3H), 3.60-3.42 (m, 2H), 3.30-3.18 (m, lH), 1.88-1.54 (m, 7H), 1.29-1.24 (m, lH); MS (ES+): m/z 481.3 (M+H) + ; LCMS (Method B): £R = 2.78 min.

Allyl (i2o.S)-8-methoxy-9-(4-methoxy-4-oxobutoxy)-6-oxo-i2-((tetra hydro- 2JT-pyran-2-yl)oxy)-i2a,i3-dihydro-6iT-benzo[5,6][i,4]diazep ino[i,2- a]indole-ii(i2ii)-carboxylate (98)

To a solution of allyl (i2aS)-9-hydroxy-8-methoxy-6-oxo-i2-((tetrahydro-2//-pyran-2 - yl)oxy)-i2a,i3-dihydro-6H-benzo[5,6][i,4]diazepino[i,2-a]ind ole-n(i2H)-carboxylate (97) (500 mg, 1.04 mmol) in /V,/V- dimethyl formamide (5 mL) were added potassium carbonate (200 mg, 1.56 mmol) and methyl 4-bromobutanoate (198 mg, 1.09 mmol) . The mixture was stirred at room temperature overnight then washed with brine (50 mL) and extracted with ethyl acetate (3 x 60 mL). The combined organic layers were dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was then purified by column chromatography (silica), eluting with ethyl acetate/petroleum ether (from 0% to 100%), to give the title compound (600 mg, 99%) as a colourless oil. Ή NMR (400 MHz, CDCl 3 ) d 8.14 (d, J=7.8 Hz, lH), 7.30-7.14 (m, 4H), 7.10-7.02 (m, lH), 6.65 (s, lH), 6.03 (d, J= 8.6 Hz, lH), 5.16-5.00 (m, 2H), 4.57 (br s, lH), 3.95 (d, J= 5.9 Hz, lH), 3.90 (s, 3H), 3.68 (s, 3H), 3-59 (br s, lH), 3-52-3-38 (m, 2H), 3.31 (br s, 2H), 2.55 (t, J=7- o Hz, 2H), 2.16

(quin, J= 6.5 Hz, 2H), 1.78 (d, J= 8.2 Hz, 2H), 1.69 (s, 2H), 1.57 (br s, 5H); NMR (100 MHz, CDCI 3 ) d 1734, i66.o, 165.9, 149-3, 141-9, 131-9, 130.0, 127.6, 125.0, 124.9, 124.4, 117-1, 114-7, 114-3, no-9, 100.2, 68.0, 67.8, 64.1, 61.1, 56.1, 56.1, 51.6, 32.5, 32.0, 31.1, 30.2, 25.2, 24.1, 20.3, 20.0; MS (ES+): m/z = 581 (M+H) + ; LCMS (Method B): t R = 4.28 min.

4-(((i2o-S)-n-((Allyloxy)carbonyl)-8-methoxy-6-oxo-i2-((t etrahydro-2ii- pyran-2-yl)oxy)-ii,i2,i2a, i3-tetrahydro-6if-benzo[5,6][i,4]diazepino[i,2- a]indol-9-yl)oxy)butanoic acid (99)

To a solution of allyl (i2aS)-8-methoxy-9-(4-methoxy-4-oxobutoxy)-6-oxo-i2- ((tetrahydro-2ff-pyran-2-yl)oxy)-i2a,i3-dihydro-6ff-benzo[5, 6][i,4]diazepino[i,2- a]indole-n(i2ff)-carboxylate (98) (600 mg, 1.03 mmol) in a mixture of tetrahydro- furan/methanol/water (3:1:1) (10 mL) was added lithium hydroxide (123 mg, 5.15 mmol). The mixture was stirred at room temperature overnight then water (50 mL) was added and the solution was acidihed to pH 3-4 with acetic acid and extracted with ethyl acetate. The organic layers were dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was then purihed by column chromatography (silica), eluting with methanol/di chloro methane (from 0% to 10%), followed by trituration in petroleum ether, to give the title compound (395 mg,

68%) as a white solid. MS (ES+): m/z = 567 (M+H) + ; LCMS (Method B): t R = 3.92 min.

Allyl tert- butyl 1,4-phenylenedicarbamate (100)

To a solution of tert-butyl (4-aminophenyl) carbamate (1.0 g, 4.8 mmol) in

dichloromethane (15 mL) at - 10 °C under inert atmosphere, were added anhydrous pyridine (873 uL, 11.04 mmol) and allyl chloroformate (607 uL, 5.04 mmol). After 1 h, the reaction mixture was diluted with dichloromethane (100 mL) and washed with a saturated aqueous solution of copper sulfate (II) (50 mL), and saturated aqueous solution of sodium bicarbonate (50 mL) and brine (50 mL). After the extraction, the combined organic layers were dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was triturated in dichloromethane and filtered to give the title compound (1.04 g, 74%) as a salmon solid. MS (ES+): m/z = 237 (M+H- t-butyl) + ; LCMS (Method B): t R = 3.83 min.

Allyl (4-aminophenyl) carbamate (101)

To a suspension of allyl tert-butyl 1,4-phenylenedicarbamate (100) (500 mg, 1.72 mmol) in methanol (0.5 mL), was added hydrochloric acid solution 4 M in 1,4 dioxane (5 mL, 20.0 mmol). After 2 h, the solvent was removed under reduced pressure to give the title compound (390 mg, 99%) as a grey solid. The product was carried through to the next step without any further purification. MS (ES+): m/z = 193 (M+H) + ; LCMS (Method B): t R = 1.95 min tert- Butyl (5-((4-(5-((4-(((allyloxy)carbonyl)amino)phenyl)carbamoyl)-i - methyl-iJT-pyrrol-3-yl)phenyl)carbamoyl)-i-methyl-iJT-pyrrol -3- yl)carbamate (102)

To a solution of allyl (4-aminophenyl)carbamate hydrochloride (101) (260 mg, 1.14 mmol) in N, L G - d i m e t h y 1 f 0 r m a m i d e (5 mL) were added 4-(4-(4-((tert-butoxycarbonyl)- amino)-i-methyl-i/7-pyrrole-2-carboxamido)phenyl)-i-methyl-i /7-pyrrole-2- carboxylic acid (31) (500 mg, 1.14 mmol), A/-[(dimethylamino)-iff-i,2,3-triazolo-[4,5- h ] py ri d i n - l-yl met hyl e n e] -L G - met hyl met h a n a m i n i 11 m hexafluorophosphate V-oxide (650 mg, 1.71 mmol) and triethylamine (633 pL, 4.56 mmol). The reaction mixture was stirred at room temperature for 24 h, then diluted with water (too mL), washed with brine (2 x too mL) and extracted with ethyl acetate (2 x too mL). The organic layer was dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was triturated in dichloromethane/methanol (2:1) and filtered to give the title compound (614 mg, 62%) as a white solid. MS (ES+): m/z = 613 (M+H) + ; LCMS (Method B): t R = 4.15 min. Allyl (4-(4-(4-(4-amino-i-methyl-iJT-pyrrole-2-carboxamido) phenyl)-i- methyl-iJT-pyrrole-2-carboxamido) phenyl) carbamate (103)

Alloc Alloc

102 103 To a suspension of tert-butyl (5-((4-(5-((4-(((allyloxy)carbonyl) amino) phenyl) carbamoyl)-i-methyl-iff-pyrrol-3-yl) phenyl) carbamoyl)-i-methyl-iff-pyrrol-3-yl) carbamate (102) (68 mg, 0.11 mmol) in methanol (0.1 mL), was added hydrochloric acid solution 4 M in 1,4 dioxane (1.0 mL, 4 mmol). After 4 h, the solvent was removed under reduced pressure to give the title compound (60 mg, 99%) as a brown solid. The product was carried through to the next step without any further purification. MS

(ES+): m/z = 513 (M+H) + ; LCMS (Method B): t R = 3.07 min

Allyl (i2aS)-9-(4-((5-((4-(5-((4-(((allyloxy)carbonyl)amino)phenyl )- carbamoyl)-i-methyl-iiT-pyrrol-3-yl)phenyl)carbamoyl)-i-meth yl-iJT- pyrrol-3-yl)amino)-4-oxobutoxy)-8-methoxy-6-oxo-i2-((tetrahy dro-2JT- pyran-2-yl)oxy)-i2a,i3-dihydro-6ii-benzo[5,6][i,4]diazepino[ i,2-a]indole- n(i2H)-carboxylate (104)

To a solution of 4-(((i2aS)-ii-((allyloxy)carbonyl)-8-methoxy-6-oxo-i2-((tetr ahydro- 2if-pyran-2-yl)oxy)-n,i2,i2a,i3-tetrahydro-6if-benzo[5,6][i, 4]diazepino[i,2-a]indol- 9-yl)oxy)butanoic acid (99) (61.76 mg, 0.109 mmol) in V,V-dimethylformamide (1 mL) were added allyl (4-(4-(4-(4-amino-i-methyl-iif-pyrrole-2-carboxamido)phenyl) -i- methyl-iff-pyrrole-2-carboxamido)phenyl)carbamate hydrochloride (103) (60 mg, 0.109 mmol), A/-[(dimethylamino)-iif-i,2,3-triazolo-[4,5-h] pyridin-i-ylmethylene]-A/- methylmethanaminium hexafluorophosphate V-oxide (45.6 mg, 0.12 mmol) and triethylamine (60.6 uL, 0.436 mmol). The reaction mixture was stirred at room temperature for 18 h. Then diluted with water (10 mL), washed with brine (2 x 10 mL) and extracted with ethyl acetate (3 x 20 mL). The organic layer was dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was then purified by column chromatography (silica), eluting with methanol/dichloro methane (from 0% to 10%), to give the title compound (108 mg, 93%) as an amber solid. Ή NMR (400 MHz, DMSO-d 6 ) d 8.19-8.11 (m, lH), 8.10-8.00 (m, lH), 7.55 (d, J=8.6 Hz, 3H), 7.36 (br s, 4H), 7-25-7-15 (m, 3H), 7 0 9 (br s, lH), 6.97 (s, lH), 7.00 (s, lH), 6.86 (s, lH), 6.08-5.81 (m, 2H), 5-74 (br s, 2H), 5-41-5-21 (m, 2H), 5.19 (br s, 2H), 5-07 (br s, 2H), 4-66 (d, J= 5.5 Hz, 2H), 4-56 (br s, 2H), 4-13 (d, =i4-4

Hz, 3H), 3-97 (s, 4H), 3-89 (br s, 3H), 3 69 (d, J=i2.5 Hz, 2H), 3-58 (br s, 2H), 3-54- 3.24 (m, 3H), 2.81 (s, 2H), 2.53 (br s, 2H), 1.77 (d, =9-8 Hz, 4H), 1.55 (br s, 5H); «C NMR (100 MHz, CDCI 3 ) d 170.0, 169.9, i66.o, 160.1, 155.4, 153-6, 140.0, 134.1, 132.5, 130.3, 130.0, 128.4, 127-6, 126.5, 125-1, 123-1, 121.2, 119.5, 118.0, 117.1, 109.8, 65.7, 64.1, 56.0, 49.8, 38.6, 36.8, 31.4, 31.1, 30.9, 25.2, 20.2; MS (ES+): m/z = 1062 (M+H) + ;

LCMS (Method B): t R = 4.32 min.

(S)-/V-(4-Aminophenyl)-4-(4-(4-(4-((8-methoxy-6-oxo-i2a,i 3-dihydro-6ii- benzo[5,6][i,4]diazepino[i,2-a]indol-9-yl)oxy)butanamido)-i- methyl-iii- pyrrole-2-carboxamido)phenyl)-i-methyl-iii-pyrrole-2-carboxa mide (105)

104 105

To a solution of allyl (i2aS')-9-(4-((5-((4-(5-((4-(((allyloxy)carbonyl)amino)pheny l)- carbamoyl)-i-methyl-iff-pyrrol-3-yl)phenyl)carbamoyl)-i-meth yl-iff-pyrrol-3-yl)- amino)-4-oxobutoxy)-8-methoxy-6-oxo-i2-((tetrahydro-2ff-pyra n-2-yl)oxy)-i2a,i3- dihydro-6ff-benzo[5,6][i,4]diazepino[i,2-a]indole-n(i2ff)-ca rboxylate (95) (100 mg, 0.094 mmol) in dichloromethane (1 mL) were added tefrafcis(triphenylphosphine)- palladium (o) (5.43 mg, 0.0047 mmol) and pyrrolidine (18.5 pL, 0.225 mmol). After 1 h, the reaction mixture was concentrated under reduced pressure. The resulting residue was then purified by column chromatography (silica), eluting with methanol/di chloro- methane (from 0% to 15%), to give the title compound (56 mg, 75%) as a yellow solid.

Ή NMR (400 MHz, DMSO-de) d 9-91 (s, lH), 9-79 (s, lH), 8.12 (d, J=7-8 Hz, lH), 8.04 (d, J=5.i Hz, lH), 7.70 (d, J=8.2 Hz, 2H), 748 (d, J=9-0 Hz, 3H), 7.41 (br s, 2H), 7-37- 7.29 (m, 2H), 7.22 (br s, 2H), 6.97 (br s, lH), 6.88 (s, lH), 6.69-6.60 (m, lH), 6.53 (d, J=8.6 Hz, lH), 4.87 (br s, lH), 4.55 (br s, lH), 4.12-4.05 (m, lH), 4-05-3-99 (m, lH), 3.89 (br s, 2H), 3-86 (s, 3H), 3 84 (s, 3H), 3-77-3-74 (m, 2H), 3.71 (s, lH), 3-65-3-57 (m, 2H), 3.17 (d, J=5.i Hz, 2H), 2.45 (d, J=7.8 Hz, 2H), 2.14-2.02 (m, 2H); MS (ES+): m/z = 791 (M+H) + ; LCMS (Method B): f R = 3.07 min.

(S)-/V-(4-Aminophenyl)-4-(4-(4-(4-((8-methoxy-6-oxo-ii,i2 ,i2a,i3- tetrahydro-6ii-benzo[5,6][i,4]diazepino[i,2-a]indol-9- yl)oxy)butanamido)-i-methyl-iJi-pyrrole-2-carboxamido)phenyl )-i- methyl-iff-pyrrole-2-carboxamide (106)

To a solution of GS)-iV-(4-aminophenyl)-4-(4-(4-(4-((8-methoxy-6-oxo-i2a, 13-dihydro- 6H-benzo[5,6][i,4]diazepino[i,2-a]indol-9-yl)oxy)butanamido) -i-methyl-iff-pyrrole- 2-carboxamido)phenyl)-i-methyl-iff-pyrrole-2-carboxamide (105) (50 mg, 0.063 mmol) in tetrahydrofuran (2.5 mL) were sequentially added ammonium formate (31.8 mg, 0.504 mmol), water (250 pL) and Pd / C (10% w/w, 25 mg). The reaction mixture was heated at 70 °C for 20 h. On completion, the reaction mixture was filtered through Celite® and washed with ethyl acetate (100 mL). The filtrate was concentrated under reduced pressure. The resulting residue was then purified by column chromatography (silica), eluting with methanol/dichloromethane (from 0% to 15%), to give the title compound (31.8 mg, 64%) as a yellow solid. Ή NMR (400 MHz, DMSO-d 6 ) d 9.91 (s, lH), 9.79 (s, lH), 9.48 (s, lH), 8.21 (d, J= 8.6 Hz, lH), 7.75-7.66 (m, J= 8.6 Hz, 2H), 7.52-7.44 (m, J= 8.2 Hz, 2H), 7.39 (s, lH), 7.36-7.15 (m, 6H), 7.05-6.99 (m, lH), 6.97 (s, lH), 6.53 (d, J=8.2 HZ, 2H), 6.35 (s, lH), 4.86 (br s, lH), 4.36 (d, J=5-i Hz, lH), 3.97 (t, J= 6.2 Hz, 2H), 3-89 (s, 3H), 3-84 (s, 3H), 3-70 (s, 3H), 3.61-3.38 (m, 2H), 3.29-3.21 (m, lH), 3.17 (d, J= 4.7 Hz, 2H), 2.89 (d, J=i6.8 Hz, 2H), 2.45 (t, J=7.o Hz, 2H), 2.06 (t, J=7.0 Hz, 2H); 13 C NMR (lOO MHz, DMSO-d 6 ) d 169.3, 166.2, 160.0, 159.7, 152.8, 145.2, 143.5, 142.8, 142.7, 141-2, 137-5, 135-6, 130.8, 130.1, 128.7, 127-5, 127.1, 125-8,

125.2, 124.8, 123.8, 123.2, 122.5, 122.3, 122.2, 120.9, 119-2, 116.7, 116.4, 115-3, 114-2, 110.3, 110.1, 109.8, 106.6, 102.1, 101.6, 67.8, 57.9, 56.4, 53-7,36.8, 36.6, 35.8, 33.3; MS (ES+): m/z = 793 (M+H) + ; LCMS (Method A): f R = 6.20 min; LCMS (Method B): f R = 3.28 min.

Methyl (S)-2-(4-(benzyloxy)-5-methoxy-2-nitrobenzoyl)-i, 2,3,4- tetrahydroisoquinoline-3-carboxylate (107) 107

A mixture of 4-(benzyloxy)-5-methoxy-2-nitrobenzoic acid (2.0 g, 6.6 mmol), oxalyl chloride (1.70 mL, 19.8 mmol) and anhydrous /V,/V- dimethyl formamide (2 drops) in anhydrous dichloromethane (40 mL) was stirred at room temperature for 3 h.

Anhydrous toluene (8 mL) was added to the reaction mixture which was then concentrated in vacuo. A solution of the resulting residue in anhydrous

dichloromethane (10 mL) was added dropwise to a solution of methyl (S)- 1, 2,3,4- tetrahydroisoquinoline-3-carboxylate (1.65 g, 7.26 mmol) and triethylamine (2.0 mL, 14.5 mmol) in anhydrous dichloromethane (30 mL) at - 10 °C. The reaction mixture was stirred at room temperature for 2 h and then washed with a saturated aqueous solution of hydrochloric acid (1 M, 20 mL) and brine (20 mL), dried over sodium sulfate, filtered and concentrated. The resulting residue was purified by flash column chromatography (silica), eluting with acetone/di chloro methane (from 0% to 30%), to give the title compound (2.5 g, 79%) as a yellow oil. Ή NMR (400 MHz, CDCl 3 ) d 7.49- 7.42 (m, 6H), 7-24-7-19 (m, 5H), 5-25 (s, 2H), 4-64-4-60 (m, lH), 4.38-4.26 (m, 2H),

3-93 (s, 3H), 3-58 (s, 3H), 3-33-3-23 (m, 2H); «C NMR (100 MHz, CDCl 3 ) d 170.8, 170.3, 154-6, 148.4, 135-3, 133-5, 130-5, 130-1, 128.9, 128.8, 128.6, 128.4, 127-7, 127-4, 126.7, 109-3, 109-1, 71-4, 56.8, 52.6, 31.8, 31.0, 30.5; MS (ES+): m/z = 477 (M+H) + ;

LCMS (Method B): £R = 4.10 min.

0S)-(4-(Benzyloxy)-5-methoxy-2-nitrophenyl)(3-(hydroxymet hyl)-3,4- dihydroisoquinolin-2(iii)-yl)methanone (108)

A solution of methyl (S')-2-(4-(benzyloxy)-5-methoxy-2-nitrobenzoyl)-i, 2,3,4- tetrahydroisoquinoline-3-carboxylate (107) (2.4 g, 5.0 mmol) in anhydrous tetrahydrofuran (48 mL) was charged with a solution of lithium boro hydride (2 M in tetrahydrofuran, 3.8 mL, 7.6 mmol) at o °C. The reaction mixture was stirred at room temperature for 3 h. Water (150 mL) was added dropwise at o °C and the reaction mixture was then extracted with ethyl acetate (2 x 100 mL). The combined organic extracts were then concentrated in vacuo. The resulting residue was purified by flash column chromatography (silica), eluting with acetone/di chloro methane (from 0% to 30%), to give the title compound (2.2 g, 97%) as a creamy oil. Ή NMR (400 MHz, CDCI 3 ) d 7·42-7·39 (m, 4H), 7·36-7·34 (m, 5H), 7-30 (s, lH), 7 2 9 (s, lH), 5-17 (s, 2H), 4.62 (s, lH), 4.36-4.25 (m, lH), 4.23-4.16 (m, 2H), 3 87 (s, 3H), 3-70-3-63 (m, lH), 3-58-3-50 (m, lH), 3.05-2.97 (m, 2H); «C NMR (too MHz, CDCl 3 ) d I68.2, 150.2, 148.3, 133.7, 128.9, 128.9, 128.8, 128.6, 127.7, 127-6, 127.5, 127-0, 126.5, 114-4, 110.6,

108.9, 103-9, 91-6, 71-4, 65.4, 54-4, 33-3; MS (ES+): m/z = 449 (M+H) + ; LCMS

(Method B): t R = 3.78 min.

0S)-(2-Amino-4-(benzyloxy)-5-methoxyphenyl)(3-(hydroxymet hyl)-3,4- dihydroisoquinolin-2(iii)-yl)methanone (109)

A solution of (S)-(4-(benzyloxy)-5-methoxy-2-nitrophenyl)(3-(hydroxymethyl )-3,4- dihydroisoquinolin-2(iff)-yl)methanone (108) (2.20 g, 4.90 mmol) in tetrahydrofuran (50 mL) and methanol (50 mL) was charged with iron (III) chloride hexahydrate (0.80 g, 2.90 mmol), activated charcoal (2.60 g, 221 mmol) and hydrazine (2.90 mL, 58.9 mmol). The reaction mixture was then stirred at reflux (85 °C) for 16 h. The mixture was subsequently allowed to cool to room temperature and filtered through a plug of celite®. The filter cake was washed with ethyl acetate and methanol and then concentrated in vacuo to give the title compound (1.7 g, 83%) as brown solid. The product was carried through to the next step without any further purification. Ή NMR

(400 MHz, MeOD) d 7.48 (s, lH), 7.46 (s, lH), 7-41-7-33 (m, 4H), 7.20-7.18 (m, 3H), 6.84 (s, lH), 6.56 (s, lH), 5.11 (s, 2H), 4.61 (s, lH), 4.54-4.40 (m, lH), 3.77 (s, 3H), 3.62-3.54 (m, 2H), 3.19 (dd, J=I6.2, 5.9 Hz, 2H), 2.92-2.80 (m, 2H); «C NMR (100 MHz, MeOD) d 169.1, 149.8, 141.0, 135.5, 130.7, 129.0, 128.7, 128.6, 128.5, 128.4, 128.2, 127 4, 127-0, 126-7, o.i, 109.1, 71.0, 68.7, 64.8, 56.4, 50.3, 27.9; MS (ES+): m/z = 419

(M+H) + ; LCMS (Method B): t R = 3.50 min.

Allyl 0S)-(5-(benzyloxy)-2-(3-(hydroxymethyl)-i,2,3,4-tetrahydro- isoquinoline-2-carbonyl)-4-methoxyphenyl)carbamate (110)

A solution of (S')-(2-amino-4-(benzyloxy)-5-methoxyphenyl)(3-(hydroxymethy l)-3,4- dihydroisoquinolin-2(iff)-yl)methanone (109) (1.50 g, 3.6 mmol) and anhydrous pyridine (696 uL, 8.97 mmol) in anhydrous dichloromethane (50 mL) at - 10 °C was slowly charged with a solution of allyl chloroformate (343 uL, 3.23 mmol) in anhydrous dichloromethane (30 mL). The reaction mixture was stirred at room temperature for 30 min and then sequentially washed with a saturated aqueous solution of copper (II) sulfate (50 mL), water (50 mL) and a saturated aqueous solution of sodium hydrogen carbonate (50 mL). The organic layer was dried over sodium sulfate, filtered and concentrated in vacuo. The resulting residue was purified by flash column

chromatography (silica), eluting with acetone/di chloro methane (from 0% to 20%), to give the title compound (1.47 g, 81%) as an off-white solid. Ή NMR (400 MHz, MeOD) d 8.14 (s, lH), 7.81 (s, lH), 7.51 (s, lH), 7.49 (s, iH), 7·42-7·32 (m, 4H), 7.23-7.17 (m, 3H), 6.82 (s, lH), 5·97-5·87 (m, lH), 5.33 (dq, J=i7-2, 1.5 Hz, lH), 5.22 (dq, J=io.6, 1.3 Hz, lH), 5.19 (s, 2H), 4-68-4.64 (m, lH), 4.61 (dd, 7=5.5, 1-3 Hz, 2H), 4-44 (br s, 2H), 3.82 (s, 3H), 3.70-3.64 (m, lH), 3.21-3.15 (m, lH), 2.74 (br s, lH); «C NMR (100 MHz,

CDCI 3 ) 8 169.4, 152-9, 148.7, 144-1, 140.1, 135-3, 131-4, 130-5, 129.1, 128.1, 127.5, 127-0, 126.7, 125-9, 125-5, 117-9, 116.8, 109.6, 105.7, 69.7, 67.4, 66.0, 64.7, 55-3, 53-8, 26.8; MS (ES+): m/z = 503 (M+H) + ; LCMS (Method B): t R = 3.95 min. Allyl (6aS')-3-(benzyloxy)-6-hydroxy-2-methoxy-i4-oxo-6,6a,7,i2-te tra- hydrobenzo[5,6][i,4]diazepino[i,2-b]isoquinoline-5(i4H)-carb oxylate

(m)

A solution of allyl (S)-(5-(benzyloxy)-2-(3-(hydroxymethyl)-i,2,3,4-tetrahydrois o- quinoline-2-carbonyl)-4-methoxyphenyl)carbamate (110) (1.4 g, 2.78 mmol) in dichloromethane (80 mL) was charged with 2,2,6,6-tetramethyl-i-piperidinyloxy (44 mg, 0.28 mmol) and (diacetoxyiodo)benzene (1.0 g, 3.33 mmol). The reaction mixture was stirred at room temperature for 16 h and was then sequentially washed with a saturated aqueous solution of sodium metabisulfite (40 mL), a saturated aqueous solution of sodium hydrogen carbonate (40 mL), water (30 mL) and brine (30 mL). The organic layer was then dried over sodium sulfate, filtered and concentrated. The resulting residue was purified by column chromatography (silica), eluting with acetone/dichloromethane (from 0% to 20%), to give the title compound (1.2 g, 86%) as an off-white solid. Ή NMR (400 MHz, CDCl 3 ) d 7.44-7.31 (m, 6H), 7.28-7.26 (m, 5H), 6.72 (s, lH), 5.70-5.61 (m, lH), 5.31 (d, J=9-8 Hz, lH), 5.20-5.17 (m, lH), 5-11-5-07 (m, 3H), 4.83 (d, J=i5-6 HZ, lH), 4.58 (d, J=i5-6 Hz, lH), 4.48-4.34 (m, 2H), 3,94 (s, 3H), 3.74-3.69 (m, lH), 3.17-3.05 (m, 2H); «C NMR (lOO MHz, CDCl 3 ) d 169.0, 149-0, 136.2, 134-3, 133-7, 131-8, 126.7, 128.2, 127.9, 127 8, 127.3, 126.7, 118.1, 114.0, 111.2, 84.8, 71.0,

66.7, 56.2, 53.5, 50.8, 44.3, 30.2; MS (ES+): m/z = 501 (M+H) + ; LCMS (Method B): t R = 3.80 min.

Allyl (6aS)-3-hydroxy-2,6-dimethoxy-i4-oxo-6,6a,7,i2-tetrahydro- benzo[5,6][i,4]diazepino[i,2-b]isoquinoline-5(i4H)-carboxyla te (112)

112

A solution of allyl (6aS)-3-(benzyloxy)-6-hydroxy-2-methoxy- 14-0x0-6, 6a, 7,12- tetrahydrobenzo[5,6][i,4]diazepino[i,2-b]isoquinoline-5(i4ff )-carboxylate (111) (100 mg, 0.199 mmol) in dichloromethane (1 mL) was charged with boron trichloride (1 M solution in dichloromethane, 600 qL, 0.600 mmol) and the resulting suspension was stirred at room temperature for 10 min, then methanol (2 mL) was added to the reaction mixture which was irradiated with microwaves 60 min at 55 °C. The reaction mixture was subsequently filtered through a cotton pad that was washed with dichloromethane and concentrated in vacuo. The resulting residue was purified by column chromatography (silica), eluting with ethyl acetate/petroleum ether (from 0% to 100%), to give the title compound (40 mg, 48%) as a cream solid. Ή NMR (400 MHz, DMSO-de) d 9·8i (s, lH), 7.39-7.20 (m, 4H), 7.09 (s, lH), 6.65 (s, lH), 5.75 (s, lH), 5.13-4.91 (m, 3H), 4.64-4-38 (m, 4H), 3.82 (s, 3H), 3-48 (br s, lH), 3.35 (s, 3H), 3.09-2.98 (m, 2H); MS (ES+): m/z = 424 (M+H) + ; LCMS (Method B): t R = 3.53 min.

Allyl (6aS')-2,6-dimethoxy-3-(4-methoxy-4-oxobutoxy)-i4-oxo-6,6a,7 ,i2- tetrahydrobenzo[5,6][i,4]diazepino[i,2-b]isoquinoline-5(i4H) -carboxylate

(113)

To a solution of allyl (6aS)-3-hydroxy-2,6-dimethoxy- 14-0x0-6, 6a, 7, 12-tetrahydro- benzo[5,6][i,4]diazepino[i,2-b]isoquinoline-5(i4if)-carboxyl ate (103) (200 mg, 0.47 mmol) in /V,/V- dimethyl formamide (2 mL) were added potassium carbonate (90 mg, 0.70 mmol) and methyl 4-bromobutanoate (90.52 mg; 0.50 mmol). The reaction mixture was stirred at room temperature overnight then washed with brine (50 mL) and extracted with ethyl acetate (2x30 mL). The combined organic layers were dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica), eluting with ethyl acetate/petroleum ether (from 0% to 70%), to give the title compound (180 mg, 73%) as a white glassy solid. Ή NMR (400 MHz, DMSO-d 6 ) d 6.55-6.40 (m, 4H), 6.29 (s, lH), 6.06 (br s, lH), 4.93 (br s, lH), 4.28-4.13 (m, 2H), 3.81 (d, J=i5-2 Hz, lH), 3.74- 3.51 (m, 3H), 3-19 (br s, 2H), 2.99 (s, 3H), 2.78 (s, 3H), 2.58 (s, 3H), 2.34-2.23 (m, 2H),

2.23-2.13 (m, 2H), 1.73-1.59 (m, 2H), 1.16 (quin, J= 6.7 Hz, 2H); NMR (100 MHz,

DMSO-de) d 173-4, 168.5, 155-3, 148.9, 141-5, 139-7, 135-3, 135-0, 133-7, 128.1, 128.0, 127.2, 126.8, 117.0, 114.4, in-4, 92-0, 68.1, 66.1, 56.4, 56.2, 55.9, 51.8, 45-4, 43-8, 30.2, 30.0, 24.4; MS (ES+): m/z = 525 (M+H) + ; LCMS (Method A): t R = 7.50 min; LCMS (Method B): t R = 3.88 min.

4-(((6aS')-5-((allyloxy)carbonyl)-2,6-dimethoxy-i4-oxo-5, 6,6a,7,i2,i4- hexahydrobenzo[5,6][i,4]diazepino[i,2-b]isoquinolin-3-yl)oxy )butanoic acid

To a solution of allyl (6aS)-2,6-dimethoxy-3-(4-methoxy-4-oxobutoxy)-i4-oxo-6,6a,7, i2-tetrahydrobenzo[5,6][i,4]diazepino[i,2-b]isoquinoline-5(i 4if)-carboxylate (113) (170 mg, 0.32 mmol) in a mixture of tetrahydrofuran/methanol/water (3:1:1) (3 mL) was added lithium hydroxide (38.3 mg, 1.6 mmol). The mixture was stirred at room temperature overnight then water (50 mL) was added then the solution was acidified to pH 1 with 1 M hydrochloric acid aqueous solution and extracted with ethyl acetate. The organic layers were dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure to give the title compound (162 mg, 99%) as a yellow oil. The product was carried through to the next step without any further purification. MS (ES+): m/z = 511 (M+H) + ; LCMS (Method B): t R = 3.55 min.

Allyl (6aS')-2,6-dimethoxy-3-(4-((5-((4-(5-(methoxycarbonyl)-i-met hyl-iJT- pyrrol-3-yl)phenyl)carbamoyl)-i-methyl-iiT-pyrrol-3-yl)amino )-4- oxobutoxy)-i4-oxo-6,6a,7,i2-tetrahydrobenzo[5,6][i,4]diazepi no[i,2- b]isoquinoline-5(i4H)-carboxylate (115)

To a solution of 4-(((6aS')-5-((allyloxy)carbonyl)-2,6-dimethoxy- 14-0x0-5, 6, 6a, 7, 12,14- hexahydrobenzo[5,6][i,4]diazepino[i,2-b]isoquinolin-3-yl)oxy )butanoic acid (114)

(170 mg, 0.33 mmol) in N, L G - d i m e t h y 1 f 0 r m a m i d e (2 mL) were added methyl 4-(4-(4- amino-i-methyl-iff-pyrrole-2-carboxamido)phenyl)-i-methyl-if f-pyrrole-2- carboxylate hydrochloride (12) (130 mg, 0.33 mmol), /V-[(di methyl ami no)-i//-i, 2,3- triazolo-[4,5-b] py ri d i n - l-yl m et hyl e n e] -/V- met hyl met h a n a m i n i u m hexafluoro- phosphate N-oxide (132 mg, 0.34 mmol) and triethylamine (133 mg, 1.32 mmol). The reaction mixture was stirred at room temperature for 24 h. Then diluted with water (20 mL), washed with brine (2 x 10 mL) and extracted with ethyl acetate (3 x 20 mL). The organic layer was dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica), eluting with methanol/dichloromethane (from 0% to 10%), to give the title compound (216 mg, 77%) as an amber glassy solid. Ή NMR (400 MHz, DMSO-d 6 ) d 9-90 (s, lH), 9.78 (s, lH), 7.74-7.61 (m, J= 8.2 Hz, 2H), 7.55 (s, lH), 7.54-7.48 (m, J= 8.2 Hz, 2H), 7.38-7.24 (m, 5H), 7.20 (d, J= 6.6 Hz, 2H), 7.12 (s, lH), 6.94 (s, 2H), 5.03 (d, J=io.i Hz, 2H), 4.49 (br s, 2H), 4.40 (br s, 2H), 4.03 (d, J=7-0 Hz, 2H), 3.89 (s, 3H), 3.82 (s, 6H), 3.77 (s, 3H), 2.47-2.41 (m, 5H), 2.07-2.01 (m, 2H), 1.99 (s, 2H); MS (ES+): m/z = 845 (M+H) + ; LCMS (Method B): £R = 4.12 min.

4-(4-(4-(4-(((6aS')-5-((Allyloxy)carbonyl)-2,6-dimethoxy- i4-oxo- 5,6,6a,7,i2,i4-hexahydrobenzo[5,6][i,4]diazepino[i,2-b]isoqu inolin-3- yl)oxy)butanamido)-i-methyl-iJT-pyrrole-2-carboxamido)phenyl )-i- methyl-iJT-pyrrole-2-carboxylic acid (116)

115 / ll6

To a solution of allyl (6aS')-2,6-dimethoxy-3-(4-((5-((4-(5-(methoxycarbonyl)-i- methyl-iff-pyrrol-3-yl)phenyl)carbamoyl)-i-methyl-iff-pyrrol -3-yl)amino)-4- oxobutoxy)-i4-oxo-6,6a,7,i2-tetrahydrobenzo[5,6][i,4]diazepi no[i,2-b]isoquinoline- 5(i4ff)-carboxylate (115) (135 mg, 0.16 mmol) in a mixture of tetrahydrofuran/ methanol/water (3:1:1) (3 mL) was added lithium hydroxide (19.2 mg, 0.8 mmol). The reaction mixture was stirred at room temperature for 84 h, then water (50 mL) was added and the solution was acidified to pH 1 with 1 M hydrochloric acid aqueous solution and extracted with ethyl acetate. The organic layers were dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure to give the title compound (130 mg, 97%) as a yellow solid. MS (ES+): m/z = 831 (M+H) + ; LCMS

(Method B): t R = 3.75 min.

Allyl (6aS , )-3-(4-((5-((4-(5-((4-aminophenyl)carbamoyl)-i-methyl- iJT- pyrrol-3-yl)phenyl)carbamoyl)-i-methyl-iiT-pyrrol-3-yl)amino )-4- oxobutoxy)-2,6-dimethoxy-i4-oxo-6,6a,7,i2-tetrahydrobenzo[5, 6][i,4]- diazepino[i,2-b]isoquinoline-5(i4H)-carboxylate (117)

117 To a solution of 4-(4-(4-(4-(((6aS , )-5-((allyloxy)carbonyl)-2,6-dimethoxy-i4-oxo-

5,6,6a,7,i2,i4-hexahydrobenzo[5,6][i,4]diazepino[i,2-b]is oquinolin-3-yl)oxy)butan- amido)-i-methyl-iff-pyrrole-2-carboxamido)phenyl)-i-methyl-i ff-pyrrole-2- carboxylic acid (116) (117 mg, 0.14 mmol) in N, L G - d i m et h y 1 f 0 r m a m i d e (1 mL) were added benzene-i, 4-diamine (15.1 mg, 0.14 mmol), N-[(dimethylamino)-iff-i,2,3- triazolo-[4,5-¾] py ri d i n - l-yl m et hyl en e] -L G - met hyl met h a n a m i n i 11 m hexafluoro- phosphate A^-oxide (55.9 mg, 0.147 mmol) and triethylamine (77.9 pL, 0.56 mmol). The reaction mixture was stirred at room temperature for 2 h, then diluted with water (20 mL), washed with brine (2 x 10 mL) and extracted with ethyl acetate (3 x 20 mL). The organic layer was dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica), eluting with methanol/dichloro methane (from 0% to 10%), to give the title compound (102 mg, 80%) as a yellow solid. Ή NMR (400 MHz, DMSO-d 6 ) d 9.91 (s, lH), 9.78 (s, lH), 9.48 (s, lH), 7.69 (d, J= 8.2 Hz, 2H), 7.48 (d, J= 8.6 Hz, 2H), 7.41-7.24 (m, 6H), 7.22 (s, lH), 7.12 (s, lH), 7.00-6.88 (m, lH), 6.53 (d, J= 8.6 Hz, lH), 5.03 (d, J=ii.3 Hz, 2H), 4-87 (br s, 2H), 4.61-4.40 (m, 4H), 4-10-3-97 (m, 3H), 3-88 (s, 2H), 3.82 (s, 6H), 3.40 (s, 3H), 3.15-3.04 (m, 2H), 3-01 (br s, lH), 2.44 (t, J=7- o Hz, 2H),

2.03 (t, J= 6.4 Hz, 2H), 1.99 (s, 2H), 1.20-1.12 (m, 4H); MS (ES+): m/z = 922 (M+H) + ; LCMS (Method A): t R = 6.52 min; LCMS (Method B): t R = 3.47 min.

(S)-JV-(4-Aminophenyl)-4-(4-(4-(4-((2-methoxy-i4-oxo-6a,7 ,i2,i4- tetrahydrobenzo[5,6][i,4]diazepino[i,2-b]isoquinolin-3- yl)oxy)butanamido)-i-methyl-iJT-pyrrole-2-carboxamido)phenyl )-i- methyl-iJT-pyrrole-2-carboxamide (118)

To a solution of allyl (6aS)-3-(4-((5-((4-(5-((4-aminophenyl)carbamoyl)-i-methyl-ii f- pyrrol-3-yl)phenyl)carbamoyl)-i-methyl-iH-pyrrol-3-yl)amino) -4-oxobutoxy)-2,6- dimethoxy- 14-0x0-6, 6a, 7, i2-tetrahydrobenzo[5,6][i,4]diazepino[i,2-b]isoquinoline- 5(i4ff)-carboxylate (117) (100 mg, 0.10 mmol) in dichloro methane (1 mL) were added feZra/a ' s(triphenylphosphine)palladium (o) (5.77 mg, 0.005 mmol) and pyrrolidine (19.7 pL, 0.24 mmol). After 15 min, the reaction mixture was concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica), eluting with methanol/dichloromethane (from 0% to 15%), to give the title compound (59 mg, 73%) as a yellow solid. Ή NMR (400 MHz, DMSO-d 6 ) d 9.90 (s, lH), 9.79 (s, lH), 9.48 (s, lH), 7.70 (d, J=7.8 Hz, 2H), 7.51-7.41 (m, 3H), 7.40-7.20 (m, 8H), 7.08 (s, lH), 6.96 (s, lH), 6.60-6.48 (m, 2H), 4.94-4.73 (m, 2H), 4.60-4.43 (m, lH), 4.20-3.93 (m, 2H), 3.88 (s, 3H), 3 83 (s, 6H), 3-73 (s, lH), 3-26-3.17 (m, 2H), 2.45 (br s, 2H), 2.11-2.01 (m, 2H); MS (ES+): m/z = 805 (M+H) + ; LCMS (Method A): t R = 5.87 min; LCMS (Method B): t R = 3.07 min. (S)-/V-(4-Aminophenyl)-4-(4-(4-(4-((2-methoxy-i4-oxo-5,6,6a, 7,i2,i4- hexahydrobenzo[5,6][i,4]diazepino[i,2-b]isoquinolin-3- yl)oxy)butanamido)-i-methyl-iii-pyrrole-2-carboxamido)phenyl )-i- methyl-iff-pyrrole-2-carboxamide (119)

To a solution of 0S)-iV-(4-aminophenyl)-4-(4-(4-(4-((2-methoxy- 14-0x0-60, 7, 12,14- tetrahydrobenzo[5,6][i,4]diazepino[i,2-b]isoquinolin-3-yl)ox y)butanamido)-i-methyl- iff-pyrrole-2-carboxamido)phenyl)-i-methyl-iff-pyrrole-2-car boxamide (118) (57 mg, 0.07 mmol) in tetrahydrofuran (2.5 mL) were sequentially added ammonium formate (22.1 mg, 0.35 mmol), water (250 pL) and Pd / C (10% w/w, 5.7 mg). The reaction mixture was heated at 70 °C for 1 h. On completion, the reaction mixture was diluted with ethyl acetate and filtered through a syringe driven filter (Millex®-HN 0.45 pm) the filter was washed with ethyl acetate (2 x 4 mL). the filtrate was concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica), eluting with methanol/dichloromethane (from 0% to 15%), to give the title compound (25.5 mg, 45%) as a white solid. Ή NMR (400 MHz, DMSO-d 6 ) d 9.91 (s, lH), 9.80 (s, lH), 9.49 (s, lH), 7.95 (s, lH), 7·73-7·66 (m, J= 9.0 Hz, 2H), 7-51-7-45 (m, J= 8.6 Hz, 2H), 7.41-7.38 (m, lH), 7.36-7.26 (m, 4H), 7.26-7.19 (m, 5H), 7.01-6.92 (m, lH), 6.53 (d, J=8.6 Hz, 2H), 6.36 (s, lH), 5.80 (d, J= 4.3 Hz, lH), 4.88 (br s, 2H), 4.73 (d, J= 15.2 Hz, 2H), 4.62 (d, J=15.6 HZ, 2H), 3.88 (s, 3H), 3.83 (s, 3H), 3.66 (s, 3H), 3.15-3.01 (m, 2H), 2.88-2.80 (m, 2H), 2.44 (t, J=7.2 Hz, 2H), 2.03 (t, J= 6.6 Hz, 2H); MS (ES+): m/z = 807 (M+H) + ; LCMS (Method A): t R = 5.95 min; LCMS (Method B): t R = 3.17 min.

Methyl (S)-4-(4-(4-(4-((2-methoxy-i4-oxo-6a,7,i2,i4-tetrahydrobenzo [5,6]- [i,4]diazepino[i,2-b]isoquinolin-3-yl)oxy)butanamido)-i-meth yl-iJT- pyrrole-2-carboxamido)phenyl)-i-methyl-iiT-pyrrole-2-carboxy late (120)

To a solution of allyl (6aS')-2,6-dimethoxy-3-(4-((5-((4-(5-(methoxycarbonyl)-i- methyl-iff-pyrrol-3-yl)phenyl)carbamoyl)-i-methyl-iff-pyrrol -3-yl)amino)-4- oxobutoxy)-i4-oxo-6,6a,7,i2-tetrahydrobenzo[5,6][i,4]diazepi no[i,2-b]isoquinoline- 5(i4ff)-carboxylate (115) (81 mg, 0.09 mmol) in dichloromethane (1 mL) were added feZra/a ' s(triphenylphosphine)palladium (o) (5.8 mg, 0.005 mmol) and pyrrolidine (17.7 uL, 0.216 mmol). After 8 min, the reaction mixture was concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica), eluting with methanol/ dichloromethane (from 0% to 6%), to give the title compound (41 mg, 62%) as a yellow solid. Ή NMR (400 MHz, DMSO-d 6 ) <59.90 (s, lH), 9.79 (s, lH), 7.68

(d, J=8.6 Hz, 2H), 7.56 (s, lH), 7·53-7·48 (m, 3H), 7·45-7·25 (m, 5H), 7.23-7.18 (m,

2H), 6.95 (s, lH), 6.84 (s, lH), 4.19-4.09 (m, 2H), 4-09-3-92 (m, 2H), 3 89 (s, 3H), 3-83 (d, J=3-5 Hz, 6H), 3.77 (s, 3H), 3-73 (s, lH), 3 2 6 (br s, 2H), 2.44 (t, J=7- 2 Hz, 2H), 2.10-2.01 (m, 2H); MS (ES+): m/z = 729 (M+H) + ; LCMS (Method B): t R = 3.70 min.

Methyl CS)-4-(4-(4-(4-((2-methoxy-i4-oxo-5,6,6a,7,i2,i4-hexahydrobe nzo- [5 ? 6][i,4]diazepino[i,2-b]isoquinolin-3-yl)oxy)butanamido )-i-methyl-iJT- pyrrole-2-carboxamido)phenyl)-i-methyl-iiT-pyrrole-2-carboxy late (121)

120

To a solution of methyl (S)-4-(4-(4-(4-((2-methoxy-i4-oxo-6a,7,i2,i4-tetrahydrobenzo -

[5 6][i,4]diazepino[i,2-b]isoquinolin-3-yl)oxy)butanamido)-i-me thyl-iii-pyrrole-2- carboxamido)phenyl)-i-methyl-iff-pyrrole-2-carboxylate (120) (39 mg, 0.05 mmol) in tetrahydrofuran (2.5 mL) were sequentially added ammonium formate (15.8 mg, 0.25 mmol), water (250 pL) and Pd / C (10% w/w, 3.9 mg). The reaction mixture was heated at 70 °C for 1 h. On completion, the reaction mixture was diluted with ethyl acetate and filtered through a syringe driven filter (Millex®-HN 0.45 mih) the filter was washed with ethyl acetate (2 x 4 mL). The filtrate was concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica), eluting with methanol/dichloro methane (from 0% to 15%), to give the title compound (28.4 mg, 80%) as a yellow solid. Ή NMR (400 MHz, DMSO-d 6 ) d 9.90 (s, lH), 9.79 (s, lH), 7.68 (d, J= 8.6 Hz, 2H), 7.59-7.46 (m, 3H), 7.34-7.14 (m, 6H), 6.95 (d, J=i.6 Hz, lH), 6.36 (s, lH), 5.79 (d, J= 4.7 Hz, lH), 4.73 (d, J=i5-6 Hz, lH), 4.62 (d, J=i5-6 Hz, lH), 3.98-3-91 (m, 2H), 3-89 (s, 3H), 3-83 (s, 3H), 3-78-3-75 (m, 3H), 3-68-3.64 (m, 3H), 3·43-3·35 (m, 2H), 3-15-3-02 (m, 2H), 2.84 (dd, J=7A, 15-6 Hz, 2H), 2.43 (t, J=7- o Hz, 2H), 2.03 (t, J= 6.6 Hz, 2H); «C NMR (101 MHz, DMSO-d 6 ) d 169.3, i68.i, 161.2, 160.0,

152.0, 147-5, 143-0, 141-6, 137-8, 135-9, 135-3, 129-4, 127-8, 127.6, 127.5, 126.9, 126.5, 125.1, 123.2, 123.2, 122.7, 122.5, 120.8, 119.2, 115.5, 114-3, 112.0, 105.2, 102.7, 97.9, 67.7, 56.3, 54.2, 52.9, 51.4, 37.0, 36.6, 32.3, 32.2, 25.3; MS (ES+): m/z = 731 (M+H) + ; LCMS (Method B): £R = 3.80 min.

Allyl (6aS')-2-methoxy-3-(4-((2-((4-(5-(methoxycarbonyl)-i-methyl- iJT- pyrrol-3-yl)phenyl)carbamoyl)-i-methyl-iiT-imidazol-4-yl)ami no)-4- oxobutoxy)-i2-oxo-6-((tetrahydro-2JT-pyran-2-yl)oxy)-6,6a,7, 8,9,io- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine-5(i2H)-carboxyl ate (122)

122

To a solution of 4-(4-(((6a.S)-5-((allyloxy)carbonyl)-2-methoxy-i2-oxo-6-((te trahydro- 2f/-pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydrobenzo[e]pyri do[i,2-a][i,4]diazepin-3- yl)oxy)butanamido)-i-methyl-iff-imidazole-2-carboxylic acid (40) (500 mg, 0.76 mmol) in N, - d i m e t h y 1 f 0 r m a m i d e (5 mL) were added methyl 4-(4-aminophenyl)-i- methyl-iff-pyrrole-2-carboxylate hydrochloride (203 mg, 0.76 mmol), - [(dimethyl - amino)-iif-i,2,3-triazolo-[4,5 -b] pyridin-i-ylmethylene]-iV-methylmethanaminium hexafluorophosphate -oxide (517 mg, 1.36 mmol) and triethylamine (422 pL, 3.04 mmol). The reaction mixture was stirred at room temperature for 24 h, then diluted with water (50 mL), washed with brine (2 x 50 mL) and extracted with ethyl acetate (3 x 30 mL). The organic layer was dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica), eluting with ethyl acetate/petroleum ether (from 0% to 100%), to give the title compound (514 mg, 78%) as an amber solid. Ή NMR (400 MHz, DMSO-de) d 10.42 (s, lH), 9.83 (s, lH), 7.95 (s, lH), 7.71 (d, J=8.6 Hz, 2H), 7-59-7-52 (m, 3H), 7-49 (d, =2.3 Hz, lH), 7.21 (d, J= 1.9 Hz, lH), 7.08-7.04 (m, lH), 5-85-5.70 (m, 2H), 5.11-5.00 (m, 2H), 4-58 (d, J=i4-i Hz, 2H), 4-49 (br s, 2H), 4.16-3.99 (m, 4H), 3.96 (s, 3H), 3-89 (s, 3H), 3-8I (S, 3H), 3-76 (s, 3H), 3·56-3·46 (m, 2H), 3-34 (br s, 2H), 2.09-2.00 (m, 2H), 1.73-1.43 (m, 12H); NMR (101 MHz, DMSO-d 6 ) d 172.4, 170.0,

169-9, 168.5, 168.5, 162.7, I6I.2, 157.2, 155.2, 149.8, 149.1, 136.7, 136.7, 136.6, 134.2, 133.2, 130.2, 127.6, 126.3, 125-3, 123-0, 122.8, 120.6, 114.8, 114.3, 110.8, 68.4, 68.2,

66.0, 63.2, 56.2, 55.3, 51.4, 37.0, 36.2, 35.5, 31.8, 31-2, 30.9, 30.6, 25.4, 25.2, 24.7, 23.1, 21.5; MS (ES+): m/z = 868 (M+H) + ; LCMS (Method A): t R = 8.07 min; LCMS

(Method B): £R = 4.20 min.

4-(4-(4-(4-(((6aS')-5-((Allyloxy)carbonyl)-2-methoxy-i2-o xo-6-((tetra- hydro-2ff-pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydrobenzo[ e]pyrido[i,2- a][i > 4-]diazepin-3-yl)oxy)butanamido)-i-methyl-iii-imidazol e-2- carboxamido)phenyl)-i-methyl-iii-pyrrole-2-carboxylic acid (123)

122

To a solution of allyl (6aS)-2-methoxy-3-(4-((2-((4-(5-(methoxycarbonyl)-i-methyl- iH-pyrrol-3-yl)phenyl)carbamoyl)-i-methyl-iH-imidazol-4-yl)a mino)-4-oxobutoxy)- i2-oxo-6-((tetrahydro-2ff-pyran-2-yl)oxy)-6, 6a, 7,8,9, io-hexahydrobenzo[e]pyri do- [i,2-a][i,4]diazepine-5(i2ff)-carboxylate (122) (497 mg, 0.57 mmol) in a mixture of tetrahydrofuran/methanol/water (3:1:1) (10 mL) was added lithium hydroxide (136 mg, 5.7 mmol) The mixture was stirred at room temperature for 36 h, then water (100 mL) was added and the solution was acidihed to pH 3-4 with glacial acetic acid and extracted with dichloromethane. The organic layers were dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure to give the title compound (359 mg, 73%) as a brown solid. The product was carried through to the next step without any further purihcation. Ή NMR (400 MHz, MeOD) d 7.66 (d, J=y.8 Hz, 2H), 7-58 (br s, lH), 7-53-7-48 (m, 2H), 7-37 (s, lH), 7 21 (s, lH), 7.11 (s, lH), 6.95 (s, lH), 6.19-6.03 (m, lH), 5.80 (br s, lH), 5-15-5-03 (m, 2H), 4.65-4.43 (m, 2H), 4-26- 4.08 (m, 4H), 4.02-3.90 (m, 6H), 3.86 (s, 3H), 3.62-3.51 (m, 2H), 3-42 (br s, lH), 3.02-

2.95 (m, lH), 2.66-2.53 (m, 2H), 2,22 (m, 2H), 1.74-1.45 (m, 12H); «C NMR (100 MHz, DMSO-de) d 161.5, 154.9, 149-4, 132-7, 126.5, 126.3, 125-8, 125.1, 124.9, H9-8, H9-5, 116.3, 114-5, 114-2, 110.6, 104.2, 68.2, 65.8, 64.9, 55-4, 55-2, 38.4, 36.1, 34-9, 30.6, 26.9, 25.3, 24.5, 23.0, 19.4, 18.2; MS (ES+): m/z = 854 (M+H) + ; LCMS (Method A): t R =

7.40 min; LCMS (Method B): t R = 3.97 min. Allyl (6aS')-2-methoxy-3-(4-((i-methyl-2-((4-(i-methyl-5-(phenyl- carbamoyl)-iff-pyrrol-3-yl)phenyl)carbamoyl)-iii-imidazol-4- yl)amino)-4- oxobutoxy)-i2-oxo-6-((tetrahydro-2ff-pyran-2-yl)oxy)-6,6a,7, 8,9,io- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine-5(i2H)-carboxyl ate (124)

To a solution of 4-(4-(4-(4-(((6aS , )-5-((allyloxy)carbonyl)-2-methoxy-i2-oxo-6-((tetra- hydro-2ff-pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydrobenzo[ e]pyrido[i,2-a][i,4]- diazepin-3-yl)oxy)butanamido)-i-methyl-iff-imidazole-2-carbo xamido)phenyl)-i- methyl-iff-pyrrole-2-carboxylic acid (123) (100 mg, 0.11 mmol) in AyV-di methyl - formamide (1 mL) were added aniline (15.4 mg, 0.165 mmol), /V-[(dimethylamino)-ii7- i,2,3-triazolo-[4,5-b] pyridin-i-ylmethylene]-/V-methylmethanaminium hexafluoro- phosphate N-oxide (46.0 mg, 0.121 mmol) and triethylamine (44.5 mg, 0.440 mmol). The reaction mixture was stirred at room temperature for 24 h, then diluted with water (50 mL), washed with brine (2 x 50 mL) and extracted with dichloromethane (3 x 30 mL). The organic layer was dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica), eluting with methanol/ dichloromethane (from 0% to 10%), to give the title compound (73 mg, 71%) as a yellow glassy solid. Ή NMR (400 MHz, DMSO-de) d 10.42 (s, lH), 9.83 (d, J= 3-9 Hz, 2H), 7-95 (s, 2H), 7-77-7-70 (m, 4H), 7-54 (d, J= 8.6 Hz, 2H), 7.49 (dd, J=2.i, 5.7 Hz, 2H), 7.41 (d, 7=1.9 Hz, lH), 7.36-7.29 (m,

2H), 7.09-7-02 (m, 2H), 5.11-4.99 (m, 2H), 4-58 (d, J=i3-7 Hz, lH), 4.48 (br s, lH), 4-15-3-99 (m, 2H), 3.97 (d, J= 1.9 Hz, 4H), 3-9i (S, 3H), 3-82 (d, J= 1.6 Hz, 3H), 3.77 (d, =5.5 Hz, lH), 3·57-3·44 (m, lH), 2.69-2.67 (m, 4H), 2.10-1.97 (m, 2H), 1.91 (br s, 2H), 1.68-1.37 (m, 12H); «C NMR (100 MHz, CDCl 3 ) d 183.0, i8i.8, 169.6, 162.5, 159.9, 148.3, 138.2, 137.8, 135.8, 132.8, 132.0, 129.7, 129-0, 128.9, 128.0, 126.6, 125.4, 125-3,

125.2, 124.0, 123.5, 120.2, 120.0, 114.5, Hi-8, 111.7, 110.7, 109-5, 103-9, 97-1, 81.7, 69.8, 56.0, 52.0, 38.9, 37-0, 36.5, 31-4, 30.7, 27-5, 25.2, 22.9, 21.4, 19.7, 18.1; MS (ES+): m/z = 930 (M+H) + ; LCMS (Method A): t R = 8.25 min; LCMS (Method B): t R = 4.28 min. CS)-4-(4-((2-Methoxy-i2-oxo-6a,7,8,9,io,i2-hexahydrobenzo[e] pyrido[i,2- a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iV-(4-(i-methy l-5-(phenyl- carbamoyl)-iiT-pyrrol-3-yl)phenyl)-iJT-imidazole-2-carboxami de (125)

To a solution of allyl (6aS')-2-methoxy-3-(4-((i-methyl-2-((4-(i-methyl-5-(phenyl- carbamoyl)-iff-pyrrol-3-yl)phenyl)carbamoyl)-iff-imidazol-4- yl)amino)-4-oxo- butoxy)-i2-oxo-6-((tetrahydro-2H-pyran-2-yl)oxy)-6, 6a, 7,8,9, io-hexahydrobenzo[e]- pyrido[i,2-a][i,4]diazepine-5(i2ff)-carboxylate (124) (68 mg, 0.07 mmol) in dichloromethane (1 mL) were added tefrafcis(triphenylphosphine) palladium (o) (4.04 mg, 0.0035 mmol) and pyrrolidine (13.8 pL, 0.168 mmol). After 1 h, the reaction mixture was concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica), eluting with methanol/ dichloromethane (from 0% to 15%), to give the title compound (38.6 mg, 74%) as a yellow solid. Ή NMR (400

MHz, DMSO-d 6 ) d 10.40 (s, lH), 9.82 (s, 2H), 7.89 (d, J= 5.5 Hz, lH), 7.73 (d, J= 3.9 Hz, 2H), 7-71 (d, J= 2.7 Hz, 2H), 7·54-7·48 (m, 3H), 7.46 (s, lH), 7.39 (s, lH), 7.31 (t, J=7- 8 Hz, 2H), 7-25 (s, lH), 7.07-7-00 (m, lH), 6.78 (s, lH), 4-13-3-99 (m, 2H), 3.95 (s, 3H), 3.88 (s, 3H), 3.80 (s, 3H), 3.68-3.65 (m, 2H), 3.10-3.06 (m, lH), 2.51 (br s, 2H), 2.08- 1.97 (m, 3H), 1.88-1.50 (m, 5H); «C NMR (100 MHz, DMSO-d 6 ) d 167.3, 165.1, 157-2

153-9, 151-6, 147-6, 140.1, 136.6, 134-7, 134-2, 130.7, 129.0,126.7, 126.0, 125.1, 123.5,

122.2, 120.7, 120.4, 115-0, 111.0, 90.7, 86.9, 79-2, 72.6, 56.3, 49-0, , 36.9, 36.7, 35-7,

35-5, 31-9, 25.0; MS (ES+): m/z = 743 (M+H) + ; LCMS (Method A): t R = 7.23 min; LCMS (Method B): t R = 3.73 min.

CS)-4-(4-((2-Methoxy-i2-oxo-5,6,6a,7,8,9,io,i2-octahydrob enzo[e]pyrido-

[i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-JV-(4- (i-methyl-5-

(phenylcarbamoyl)-iiT-pyrrol-3-yl)phenyl)-iJT-imidazole-2 -carboxamide

(126)

To a solution of (S)-4-(4-((2-methoxy- 12-0x0-60,7, 8, 9,10, i2-hexahydrobenzo[e]pyrido- [i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-/V-(4-(i- methyl-5-(phenyl- carbamoyl)-iff-pyrrol-3-yl)phenyl)-iff-imidazole-2-carboxami de (125) (34 mg, 0.04 mmol) in tetrahydrofuran (2 mL) were sequentially added ammonium formate (12.6 mg, 0.20 mmol), water (200 pL) and Pd / C (10% w/w, 3.4 mg). The reaction mixture was heated at 70 °C for 2 h. On completion, the reaction mixture was diluted with ethyl acetate and filtered through a syringe driven filter (Millex®-HN 0.45 pm) the filter was washed with dichloro methane (2 x 7 mL). The filtrate was concentrated under reduced pressure to give the title compound (29 mg, 97%) as a white solid. Ή NMR (400 MHz, DMSO-de) d 10.41 (s, lH), 9 83 (s, 2H), 7.76-7.71 (m, 4H), 7-54 (s, lH), 7-52 (s, lH),

7.50 (s, lH), 7.48 (s, lH), 7.47 (d, 7=1.9 Hz, lH), 7.40 (d, 7=1.9 Hz, lH), 7·35-7·29 (m, 2H), 7.08-7.02 (m, lH), 6.36 (s, lH), 5.94 (t, J=3-9 Hz, lH), 4.15-4.07 (m, 2H), 3-96 (s, 3H), 3·95-3·93 (m, lH), 3.92 (s, 3H), 3-66 (s, 3H), 3-57 (d, =3-9 Hz, 2H), 3.25-3.21 (m, 2H), 3.15-3.07 (m, 2H), 2.07-2.00 (m, 2H), 1.71-1.37 (m, 6H); «C NMR (100 MHz,

DMSO-de) d 170.0, 165.9, i6o.i, 157 2, 151 8, 145-9, 141-6, 139-8, 136.6, 136.5, 134.3, 130.7, 129.0, 126.7, 126.0, 125.1, 123.5, 122.2, 120.7, 120.4, 116.7, 111.5, 111.0, 101.8,

67.7, 59-2, 56.4, 51-9, 44-5, 37-0, 35-5, 32.0, 29.6, 25.0; MS (ES+): m/z = 745 (M+H) + ; LCMS (Method A): £R = 7.27 min; LCMS (Method B): £R = 3.80 min.

Allyl (6a-S)-2-methoxy-3-(4-((i-methyl-2-((4-(i-methyl-5-(pyridin- 4- ylcarbamoyl)-iT7-pyrrol-3-yl)phenyl)carbamoyl)-iT7-imidazol- 4-yl)amino)- 4-oxobutoxy)-i2-oxo-6-((tetrahydro-2JT-pyran-2-yl)oxy)-6,6a, 7,8,9,io- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine-5(i277)-carboxy late (127)

To a solution of 4-(4-(4-(4-(((6aS')-5-((allyloxy)carbonyl)-2-methoxy-i2-oxo- 6- ((tetrahydro-277-pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydr obenzo[e]pyrido[i,2- a][i,4]-diazepin-3-yl)oxy)butanamido)-i-methyl-i77-imidazole -2-carboxamido)- phenyl)-i-methyl-i77-pyrrole-2-carboxylic acid (123) (105 mg, 0.12 mmol) in N,N- dimethylformamide (1 mL) were added 4-aminopyridine (15.4 mg, 0.165 mmol), N- [(dimethyl-amino)-iii-i,2,3-triazolo-[4,5-b] py ri d i n - l-yl met hyl e n e]-N- methylmethanaminium hexafluorophosphate N-oxide (50.2 mg, 0.13 mmol) and triethylamine (66.7 pL, 0.48 mmol). The reaction mixture was stirred at room temperature for 48 h, then diluted with water (30 mL), washed with brine (2 x 30 mL) and extracted with dichloromethane (3 x 30 mL). The organic layer was dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica), eluting with methanol/dichloro methane (from 0% to 15%), to give the title compound (30 mg, 27%) as a pale yellow solid. Ή NMR (400 MHz, CDCl 3 ) <5 8.92 (d, J= 10.9 Hz, lH), 8.51 (d,7=5-1 Hz, 3H), 7-64 (d, J= 5-9 Hz, 2H), 7·58-7·47 (m, 2H), 7.46-7.40 (m, lH), 7.34 (d, J= 8.2 Hz, 2H), 7.20 (d, J= 6.2 Hz, lH), 7.15-7.04 (m, 2H), 5.19-4.98 (m, 2H), 4.28 (br s, 2H), 4.17-4.05 (m, 2H), 4-05 (s, 6H), 3.89 (s,3H), 348 (br s, 2H), 3.16-3.02 (m, 2H), 2.70-2.55 (m, 2H), 2.26 (br s, 2H), 2.18 (d, J= 4.7 Hz, 2H), 2.05 (br s, 2H), 1.73-1-53 (m, 12H); MS (ES+): m/z = 931 (M+H) + ; LCMS (Method A): £R = 6.52 min. (S)-4-(4-((2-Methoxy-i2-oxo-6a,7,8,9,io,i2-hexahydrobenzo[e] pyrido[i,2- «][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-/V-(4-(i-meth yl-5-(pyridin- 4-ylcarbamoyl)-iiT-pyrrol-3-yl)phenyl)-iJT-imidazole-2-carbo xamide (128)

To a solution of allyl (6aS)-2-methoxy-3-(4-((i-methyl-2-((4-(i-methyl-5-(pyridin-4 - ylcarbamoyl)-iff-pyrrol-3-yl)phenyl)carbamoyl)-iii-imidazol- 4-yl)amino)-4-oxo- butoxy)-i2-oxo-6-((tetrahydro-2ff-pyran-2-yl)oxy)-6, 6a, 7,8,9, io-hexahydrobenzo[e]- pyrido[i,2-a][i,4]diazepine-5(i2ff)-carboxylate (127) (28 mg, 0.03 mmol) in dichloromethane (1 mL) were added tefrafcis(triphenylphosphine) palladium (o) (1.7 mg, 0.0015 mmol) and pyrrolidine (5.12 pL, 0.72 mmol). After 1 h, the reaction mixture was concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica), eluting with methanol/dichloro methane (from 0% to 25%), to give the title compound (16 mg, 71%) as a pale yellow solid. Ή NMR (400 MHz, DMSO-de) d 10.41 (s, lH), 10.16 (s, lH), 9.85 (s, lH), 8.44 (d, J= 5-9 Hz, 2H), 7.80-7.70 (m, 5H), 7-57-7-53 (m, 3H), 7.51 (s, lH), 7.47 (d, J= 1.9 Hz, lH), 7.16-7.05 (m,

2H), 6.60-6.47 (m, 2H), 6.12 (d, 7=1.6 Hz, 2H), 3-97 (s, 3H), 3-91 (s, 3H), 3-71 (s, 3H), 3.30 (s, 2H), 2.10-1.99 (m, 2H), 1.74-1.47 (m, 6H); «C NMR (100 MHz, DMSO-d 6 ) d 188.5, i8o.o, 172.0, 170.1, 160.6, 157.2, 151.1, 150.6, 146.6, 136.7, 136.6, 134.2, 127.1, 126.0, 125.1, 120.7, 119-8, 114-0, 113-6, 110.3, 106.7, 79-0, 43-1, 37-1, 35-5; 25.0; MS

(ES+): m/z = 744 (M+H) + ; LCMS (Method A): t R = 5.77 min; LCMS (Method B): t R = 2.97 min. CS)-4-(4-((2-Methoxy-i2-oxo-5,6,6a,7,8,9,io,i2-octahydrobenz o[e]pyrido- [i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-/V-(4-(i- methyl-5- (pyridin-4-ylcarbamoyl)-iii-pyrrol-3-yl)phenyl)-iii-imidazol e-2- carboxamide

To a solution of (S)-4-(4-((2-methoxy- 12-oco-όa, 7, 8, 9,10, i2-hexahydrobenzo[e]pyrido- [i,2-a][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iV-(4-(i- methyl-5-(pyridin-4- ylcarbamoyl)-iff-pyrrol-3-yl)phenyl)-iff-imidazole-2-carboxa mide (128) (12 mg,

0.016 mmol) in tetrahydrofuran (2 mL) were sequentially added ammonium formate (5.04 mg, 0.08 mmol), water (200 pL) and Pd / C (10% w/w, 1.2 mg). The reaction mixture was heated at 70 °C for 4 h. On completion, the reaction mixture was diluted with a mixture 1/1 dichloromethane/methanol and filtered through a syringe driven filter (Millex®-HN 0.45 pm) the filter was washed with dichloromethane (2 x 7 mL). the filtrate was concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica), eluting with methanol/dichloromethane (from 0% to 15%), to give the title compound (10.1 mg, 84%) as a white solid. Ή NMR

(400 MHz, DMSO-d 6 ) d 10.41 (s, lH), 10.16 (s, lH), 9.85 (s, lH), 8.44 (d, J= 6.2 Hz, 2H), 7.82-7.67 (m, 4H), 7-55 (d, J= 1.6 Hz, 2H), 7-53 (s, lH), 7-51 (s, lH), 7-49 (s, lH),

7-47 (d, J= 1.9 Hz, lH), 6.37 (s, lH), 5.94 (t, J=3-7 Hz, 2H), 4-12 (d, J=12.9 Hz, 2H), 3-97 (s, 3H), 3-92 (s, 3H), 3 67 (s, 3H), 3-30 (br s, 2H), 3.26-3.07 (m, 4H), 2.12-1.94 (m, 2H),

1.82-1.29 (m, 6H); ^C NMR (100 MHz, DMSO-d 6 ) d 197-9, 165.9, i6o.6, 157.2, 151.8, 150.7, 146.6, 145.9, 141-6, 136.6, 134.2, 130.4, 126.0, 125.1, 120.7, 120.3, 114-0, 111.5, 59.2, 56.4, 51.9, 37.1, 25.0; MS (ES+): m/z = 746 (M+H) + ; LCMS (Method A): t R =

5.67 min; LCMS (Method B): t R = 2.97 min.

Allyl (6aS')-3-(4-((2-((4-(5-((4-acetamidophenyl)carbamoyl)-i-meth yl-iJT- pyrrol-3-yl)phenyl)carbamoyl)-i-methyl-iiT-imidazol-4-yl)ami no)-4- oxobutoxy)-2-methoxy-i2-oxo-6-((tetrahydro-2JT-pyran-2-yl)ox y)- 6,6a,7,8,9,io-hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepine-5 (i2Ji)- carboxylate

To a solution of 4-(4-(4-(4-(((6aS')-5-((allyloxy)carbonyl)-2-methoxy-i2-oxo- 6- ((tetrahydro-2ff-pyran-2-yl)oxy)-5,6,6a,7,8,9,io,i2-octahydr obenzo[e]pyrido[i,2- <3][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iff-imidaz ole-2-carboxamido)phenyl)- i-methyl-iff-pyrrole-2-carboxylic acid (123) (152 mg, 0.17 mmol) in N,N- dimethylformamide (1 mL) were added V-(4-aminophenyl)acetamide (51.1 mg, 0.34 mmol), /V-[(dimethylamino)-ii/-i,2,3-triazolo-[4,5-b] pyridin-i-ylmethylene]-iV- methylmethanaminium hexafluorophosphate V-oxide (137 mg, 0.35 mmol) and triethylamine (89.0 pL, 0.64 mmol). The reaction mixture was stirred at room temperature for 48 h, then diluted with water (30 mL), washed with brine (2 x 30 mL) and extracted with dichloromethane (3 x 30 mL). The organic layer was dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica), eluting with methanol/dichloro methane (from 0% to 15%), to give the title compound (112 mg, 67%) as a yellow solid. Ή NMR (400 MHz, CDCl 3 ) <5 10.42 (s, lH), 9.87 (s, lH), 9.83 (s, lH), 9.79 (s, lH), 7.74 (d, J=8.6 Hz, 2H), 7.66-7.60 (m, 3H), 7·55-7·44 (m, 7H), 7-37 (d,7=1.9 Hz, 2H), 7.08-7.03 (m, 2H), 6.88 (br s, 2), 6.02 (d, J=io.i Hz, 2H), 5.04 (br s,

3H), 4-00 (d, J=7- o Hz, 2H), 3-97-3-95 (s, 3H), 3-92 (s, 3H), 3 89 (s, 2H) 3-81 (s, 3H), 3.31 (s, 2H), 2.03 (br s, 5H), 1.75-1.47 (m, 12H); MS (ES+): m/z = 987 (M+H) + ; LCMS (Method A): t R = 7.55 min; LCMS (Method B): t R = 3.90 min. ( < S)-iV-(4-(5-((4-Acetamidophenyl)carbamoyl)-i-methyl-iT i-pyrrol-3-yl)- phenyl)-4-(4-((2-methoxy-i2-oxo-6a,7,8,9,io,i2- hexahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i- methyl-iJT-imidazole-2-carboxamide (131)

To a solution of allyl (6aS')-3-(4-((2-((4-(5-((4-acetamidophenyl)carbamoyl)-i-meth yl- iff-pyrrol-3-yl)phenyl)carbamoyl)-i-methyl-iff-imidazol-4-yl )amino)-4-oxobutoxy)-2- methoxy-i2-oxo-6-((tetrahydro-2ff-pyran-2-yl)oxy)-6, 6a, 7,8,9, io-hexahydrobenzo[e]- pyrido[i,2-a][i,4]diazepine-5(i2ff)-carboxylate (130) (107 mg, 0.10 mmol) in dichloromethane (5 mL) were added tefrafcis(triphenylphosphine) palladium (o) (5.77 mg, 0.005 mmol) and pyrrolidine (19.7 pL, 0.24 mmol). After 2 h, the reaction mixture was concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica), eluting with methanol/di chloro methane (from 0% to 15%), to give the title compound (47 mg, 59%) as a pale yellow solid. Ή NMR (400

MHz, CDCI 3 ) <5 10.42 (s, lH), 9.87 (s, lH), 9.84 (s, lH), 9.81-9.77 (m, lH), 7.74 (d, J= 9.0 Hz, 2H), 7.66-7.60 (m, 3H), 7-55-7-50 (m, 7H), 7.46 (d, J= 1.9 Hz, lH), 7.37 (d, J=i.6 Hz, 2H), 4-07-4-01 (m, lH), 3.97 (s, 3H), 3-90 (s, 3H), 3 89 (br s, lH), 3.82 (s, lH), 3.74-3.70 (m, 2H), 3-69-3-65 (m, 2H), 2.07-2.01 (m, 5H), 1.87 (br s, 2H), 1.74-1-53 (m, 6H); MS (ES+): m/z = 800 (M+H) + ; LCMS (Method A): t R = 6.45 min; LCMS

(Method B): t R = 3.33 min.

(S)-/V-(4-(5-((4-Acetamidophenyl)carbamoyl)-i-methyl-iiT- pyrrol-3- yl)phenyl)-4-(4-((2-methoxy-i2-oxo-5,6,6a,7,8,9,io,i2- octahydrobenzo[e]pyrido[i,2-a][i,4]diazepin-3-yl)oxy)butanam ido)-i- methyl-iJT-imidazole-2-carboxamide (132)

To a solution -pyrrol-3- yl)phenyl)-4-(4-((2-methoxy- 12-0x0-60,7,8, 9,10, i2-hexahydrobenzo[e]pyrido[i, 2-

<3][i,4]diazepin-3-yl)oxy)butanamido)-i-methyl-iff-imi dazole-2-carboxamide (131) (40 mg, 0.05 mmol) in tetrahydrofuran (3 mL) were sequentially added ammonium formate (15 mg, 0.25 mmol), water (300 pL) and Pd / C (10% w/w, 4.0 mg). The reaction mixture was heated at 70 °C for 5 h. On completion, the reaction mixture was diluted with a mixture 1/1 dichloromethane/ methanol and filtered through a syringe driven filter (Millex®-HN 0.45 pm) the filter was washed with dichloromethane (2 x 7 mL). the filtrate was concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica), eluting with methanol/di chloro methane (from 0% to 20%), to give the title compound (22.2 mg, 55%) as a pale yellow solid.

Ή NMR (400 MHz, DMSO-de) d 10.42 (s, lH), 9.87 (s, lH), 9 84 (s, lH), 9.79 (s, lH), 7-74 (d, J= 8.6 Hz, 2H), 7-63 (d, J= 9.0 Hz, 2H), 7·55-7·5 o (m, 5H), 7-49 (s, lH), 746 (s, lH), 7.38 (s, lH), 6.43-6.32 (m, lH), 5.94 (br s, lH), 4.18-4.06 (m, 2H), 3-97 (s, 3H), 3-94 (t, J= 6.2 Hz, 2H), 3-90 (s, 3H),3-68 (S, 3H), 3-57 (d, =3-9 Hz, 2H), 3 2 3 (br s, lH), 3.18-3.06 (m, 2H), 2.03 (s, 5H), 1.71-1-35 (m, 6H); «C NMR (100 MHz, DMSO-d 6 ) d 170.0, 168.4, 165-9, 159-9, 157-2, 151-8, 145-9, 141-6, 136.6, 136.4, 135-2, 134-9, 134-2, 130.7, 126.8, 125.8, 125.1, 122.2, 120.8, 120.7, 119-7, 116.7, 114-9, 111.5, 110.7, 101.8,

90.2, 67.7, 59-2, 564, 51-9, 44-5, , 36.9, 35-5, 32.0, 29.6, 25.0, 24.4; MS (ES+): m/z = 802 (M+H) + ; LCMS (Method A): £R = 6.60 min; LCMS (Method B): £R = 3.40 min.

FRET DNA Melting

FRET DNA melting studies were undertaken on 15 and 16 using two fluorescently labelled sequences. The sequences (Figure 1) were designed to represent poly AT and poly GC sequences.

The short duplexes used in this FRET study are relatively unstable in the duplex form with a melting temperature below 30°C so that, in the absence of ligand, a large part of the melting occurs below the starting temperature of the experiment. However, it can be observed that 15 stabilizes the duplex form, producing increases in melting temperature with A7i„ values of -30 °C for 5’-AAAAAAAGAAAAAATTT-3’ (Figure 2, top panel), and ~25°C (Figure 2, bottom panel). The melting temperature of each duplex increases significantly in proportion to the concentration of 15 present, providing strong supporting evidence that the compound can effectively interact with both sequences.

When 15 was fragmented into its component parts (28 and 32), it failed to stabilise DNA (Figure 3), suggesting the combination of the alkylating fragment with the DNA- interactive component is critical for DNA stabilisation. The melting temperature of the duplex increases marginally in the presence of each fragment, suggesting limited stabilisation of the DNA.

Surprisingly, compound 16 (which does not contain an alkylating group) produces a concentration-dependent increase in fluorescence when reacted with the poly-AT sequence, suggesting non-covalent interaction and significant stabilisation of the DNA (~20°C). There is little interactivity with the poly-GC sequence, suggesting sequence selectivity of 16. The melting temperature of the duplex (see Figure 4) increases in a concentration dependent manner (top) when reacted with s'-AAAAAAAGAAAAAATTT- 3', suggesting sequence-specific interaction with DNA. There is limited fluorescence in the case of 5'-AATAGGGGGTCGCCTATT-3' (bottom), suggesting limited stabilisation of the DNA in this instance.

In Vitro Cytotoxicity

In vitro cytotoxicity data suggests 16 is potent in the nanomolar range (Table 1).

The in vitro cytotoxicity of a selection of compounds were evaluated in a panel of cell lines using the standard MTT assay for a 72 hour incubation period (Table 2). Free payloads produced cytotoxicities in the nanomolar range.

Table 2: In vitro cytotoxicity of a selection of compounds against three cell -lines

DNA FOOTPRINTING

The DNA sequence selectivity profile of the molecules was investigated using a modification of the previously established DNA footprinting assay [31]. Following an overnight incubation of the ligand- DNA complexes, the mixture was mixed with strand separation buffer containing 10 mM EDTA, 10 mM NaOH, 0.1% bromophenol blue, 80% formamide and incubated at too °C for 3 min. The mixture was then immediately cooled on ice and run on an 8% denaturing gel. Examination of the obtained gel (Figure 5, upper panel) shows footprints produced by the molecules on the DS3 DNA sequence. Interestingly, although the DS3 DNA fragments contains multiple potential binding sites for molecules 15 and 16, there is a distinct difference in binding pattern between both compounds. Compound 15 was found to bind to tracts of DNA where a GC base is present, whereas 16 was found to predominantly target AT rich regions. Molecular modelling studies suggest that this occurs due to the non-covalent van der Waals interactions of the polyamide sidechain with adenine and thymine bases (data not presented). These data also suggest the molecules both act in a highly sequence selective manner with a different sequence selectivity profile for each molecule. The possible adducts formed within the DS3 sequence are shown in Figures 5 (lower panel). A further obtained gel (Figure 6, upper panel) shows footprints produced by 16 on the random“D” DNA sequence. Interestingly, although the“D” DNA fragment contain multiple potential binding sites for 16 (i.e., multiple examples of potential sites), only three preferred sites were observed during this experiment, and all are primarily AT- rich. Molecular modelling studies suggest that this occurs due to the occupation of the molecule in the DNA minor groove, where sequence-interactive H-bonds and non- covalent interactions guide it to a preferred binding site. These data again suggest that the molecules all act in a sequence selective manner, which is a surprising observation given the fact that the molecule does not have the ability to alkylate DNA.

Transcription Factor Plate Array Assay

A transcription factor plate array assay experiment was undertaken to establish which transcription factors are down-regulated through the alkylation of DNA by 16. The study showed that the major transcription factors down-regulated were GR/PR, CDP, ATG2 and Oct-4 (see Figure 7). The consensus sequences of each of these transcription factors corresponds to the DNA footprinting pattern observed for 16.

Compound 16 has been found to bind to XXXXWWW where X is any base and W is A or T. In the case of the transcription factor CDP (consensus site CCAAT), there is an obvious correlation between the DNA footprint derived and the consensus sequence. Similarly, in the case of (consensus site ATGCWAAT where W represents A or T), a binding site (bold and underlined) can be identified, and one of the consensus sequences of ATF2 (5OTGACGTAA-3) also directly corresponds to the DNA footprint. Summary

Taken together, the biophysical data provide strong evidence that 16 effectively stabilises DNA with a degree of sequence-specificity. These data suggest that the population of DNA adduct types derived may account for the cytotoxicity of this family of compounds in cells. Furthermore, DNA Footprinting studies indicate a degree of sequence selectivity for the class, with the DNA-binding site generally corresponding to XXXWWWW where X represents any base and W indicates adenine or thymine. 16 was shown to down-regulate a number of key transcription factors ( e.g ., Oct4 and GATA), and analysis suggests that their binding sites correspond to the main DNA Footprint observed for this class of molecules. Overall, these data suggest that the potent cytotoxicity observed for the class of payloads is directly related to their DNA-binding affinity and sequence selectivity which can result in the inhibition and down-regulation of key transcription factors. The fact that these compounds do not alkylate DNA (either monoalkylated or cross-link) as occurs with the PBD dimers, IGNs and CXI classes suggests that they may produce less overall systemic toxicity [31], and may provide a higher Therapeutic Index in animal models or human clinical trials.

Examples of Conjugation to Antibodies, In vitro and In vivo Efficacy Stochastic Conjugation

Conjugation of 47 to IgGi antibody (forming ADCi)

47 was conjugated to an IgGi antibody (Cetuximab, ADC Biotechnology, lot number 244996) targeted to EGFR Antigen in a stochastic manner.

Antibody QC

The IgGi antibody was of good quality with >90% monomer content (Figure 8) and a varied DAR profile providing a stochastic DAR of 1.9 (Figure 9).

No free toxin linker could be detected in the antibody-drug conjugate IgGi-47 sample (see Figure 10).

FRET Studies Methodology

1. General

1.1. Oligonucleotides

Oligonucleotides were obtained from ATDbio (Southampton, UK) in lyophilised form. They were labelled with a fluorophore molecule (F = fluorescein) at the 5’-end and a quencher molecule (Q = dabcyl) at the 3’-end of the complementary strand. Each oligonucleotide was dissolved in distilled H 2 0 to form stock solutions of 100 mM. Working solutions of 5 pM were prepared by diluting the stock solution with distilled H 2 0.

1.2. Buffers

The following buffers were used: 250 mM phosphate buffer pH 7.4 (consisting of sodium dihydrogen phosphate and sodium phosphate diluted in distilled H 2 0) and 5 M sodium chloride buffer. All buffers and distilled H 2 0 were filtered through a 0.2 pM filter prior to use. 1.3. Compound

For the FRET experiments a stock solution of the relevant compound was prepared by dissolving it in DMSO to give a concentration of 10 mM. From this stock solution, working solutions of the desired concentration were prepared by diluting the stock solution with distilled H 2 0.

1.4. Preparation ofligand-DNA complexes

The reaction mixture was comprised of 4 pL of 250 mM phosphate buffer (final concentration of 50 mM), 4 pL flourophor and 4 pL quencher molecule of the appropriate oligonucleotide for a final concentration of 0.2 pM, 4 pL 5 M sodium chloride (final concentration of 1 M NaCl), and 4 pL of distilled H 2 0. This mixture was heated in an Eppendorf tube at 90 °C for 1 min and slowly cooled down to room temperature. This process was carried out to anneal the single strands to double- stranded DNA. Following this, 4 pL of the ligand was added in the desired

concentration and the mixture incubated overnight either at room temperature or 4 °C. A control sample of DNA only was prepared by mixing 4 pL 250 mM phosphate buffer (final concentration of 50 mM) with 4 pL fluorophore-labelled and 4 pL quencher- labelled oligonucleotides (of the appropriate sequence) to give a final concentration of 0.2 pM, 4 pL 5 M sodium chloride (final concentration of 1 M NaCl) and 4 pL distilled H 2 0. This mixture was analysed without prior annealing.

1.5. Fluorescence melting

Fluorescence melting profiles were measured using a Roche LightCycler using a total reaction volume of 20 pL. Initially, the samples were denatured by heating to 95 °C at a rate of 1 °C min 1 . The samples were then maintained at 95 °C for 5 min before annealing by cooling to 25 °C at 1 °C min 1 . The samples were then held at 25 °C for a further 5 min and finally melted by heating to 95 °C at 1 °C min 1 . Annealing steps and melting steps were all recorded and changes in fluorescence were measured at 520 nm. 1.6. Data analysis

T m values were obtained from the first derivates of the melting profiles using the Roche LightCycler software.

MTT Cytotoxicity Methodology

Tumor cell lines were maintained in RPMI1640 medium supplemented with 10% heat- inactivated fetal bovine serum, 2mM L-glutamine and imM sodium pyruvate. 1800 cells per well were seeded in a volume of i8opl in a 96-well flat bottom polystyrene plate. The cells were allowed to adhere overnight at 37°C in a C0 2 incubator. Ligands were initially formulated in DMSO, and stocks stored at -8o°C. They were then further formulated at tox concentration in RPMI1640 medium. 20ul of diluted samples were added into each treatment well. On each plate, blank wells with no cells, and untreated wells containing cells, were included. Plates were then cultured at 37°C in a C0 2 incubator for 72hrs. Cytotoxicity was evaluated using a tetrazolium salt-based assay, the MTT assay. After 72hours, the supernatant was removed from each well and 200m1 of a sterile filtered 500pg/ml MTT solution in water added to each well. The plates were then incubated at 37°C in a C0 2 incubator for 4hrs. The supernatant was then removed and the formazan crystals formed solubilized by adding I50m1 of DMSO to each well. The plate was then read on a plate reader at 54onm, and percentage cell survival calculated as follows: ((mean absorbance treated wells at concentration x - mean absorbance blank wells) ÷ (mean absorbance untreated wells at concentration x - mean absorbance blank wells)) x too. Data were plotted as concentration in nM vs. % cell survival in Microsoft Excel, and IC 50 values (concentration where cell survival is reduced by a half) were determined from the graph.

DNA Footprinting Methodology

The preparation of the DS3 DNA fragment (Figure 5) and the“D” DNA fragment (Figure 6) has been previously described [32]. Briefly, the sequence which had been cloned into the BamHl site of pUCi8 was obtained by cutting with Hindlll and EcoRI. Radiolabelled DNA fragments were prepared by filling in the 3’-end of the Hindlll site with [a- 32 P]dATP using Klenow DNA polymerase (exo-).

The radiolabelled DNA fragment was separated from the remainder of the plasmid DNA on a 6% non-denaturing polyacrylamide gel. The gel (20 cm long, 0.3 mm thick) was run at 400 V in lx TBE running buffer for about i-2h, until the bromophenol blue had run most of the way down the gel. The glass plates were separated and the position of the labelled DNA fragment was established by short (1 min) exposure to an X-ray film. The relevant band was then cut from the gel and the radiolabelled DNA eluted by adding 300 pL 10 mM Tris-HCl, pH 7.5 containing 0.1 mM EDTA and gently agitating overnight at room temperature. The eluted DNA was finally precipitated with ethanol and re-suspended in a suitable volume of 10 mM Tris-HCl, pH 7.5 containing 0.1 mM EDTA buffer so as to give at least 10 counts per second/ pL on a hand-held Geiger counter. With fresh plasmid and a- 32 P-dATP this process typically generated about 150 pL of radiolabelled fragment DNA. The absolute concentration of the DNA is not important, and it is typically lower than 10 nM.

Footprinting reactions were performed as previously described [31] using the DNA fragments DS3, which contains tracts of AT/GC bases; and using the“D” DNA fragments which represents a random DNA sequence. The DNA fragments were obtained by cutting the parent plasmids with Hindlll and Sad or EcoRI and Pstl, and were labelled at the 3'-end of the sites with [a- 32 P]dATP using reverse transcriptase or exo- Klenow fragment. After gel purification, the radiolabelled DNA was dissolved in 10 mM Tris-HCl pH 7.5 containing 0.1 mM EDTA, at a concentration of about 10 c.p.s per pL as determined on a hand held Geiger counter. 1.5 pL of radiolabelled DNA was mixed with 1.5 pL ligand that had been freshly diluted in 10 mM Tris-HCl pH 7.5, containing 10 mM NaCl. The complexes were left to equilibrate for at least 12 hours before digesting with 2 pL DNase I (final concentration about 0.01 units/mL). The reactions were stopped after 1 minute by adding 4 pL of formamide containing 10 mM EDTA and bromophenol blue (0.1% w/v).

The samples were then heated at too °C for 3 minutes before loading onto 8% denaturing polyacrylamide gels containing 8 M urea. Gels were fixed in 10% acetic acid, transferred to 3MM paper, dried and exposed to a phosphor screen overnight, before analysing with a Typhoon phosphorimager

Transcription Factor Plate Array Assay

The transcription factor plate array assay kit was obtained from Signosis Inc (USA). Briefly, 2 X 106 HeLa cells were treated with toonM compound 16 and incubated for 6 hours before extracting the nuclear protein and carrying out the TF plate array assay. The assay was carried out following the manufacturer’s protocol. In the case of each transcription factor, the RLU value obtained for the cells treated with 16 was deducted from the respective values obtained for the untreated cells to obtain the differences in TF activation/inhibition. Conjugation of Payload to Antibody

All ADC conjugations were completed using a similar methodology, an example of which is provided below. 21.5 mg IgGi antibody (8.0 mg/ml in PBS) were charged with EDTA to a final concentration of 2 mM. Reduction was attained by adding 1.27 molar equivalents TCEP (10 mM in water) and incubating for 2 hours at 20°C. After 1.5 hours, a reduction in-process test conjugation with Mal-vcMMAE was performed, and analyzed by HIC to test for the reduction level. As the target reduction level had not been reached, another 0.1 molar equivalents TCEP were added and the reduction time extended by 1 hour. After 0.5 hours, a second in-process test was run. After

confirmation of the desired reduction level, 20% (v/v) Propylene glycol was added to the reduced antibody followed by 6.4 molar equivalents of compound 16 (10 mM stock in DMSO). The solution was incubated for 1 hour at rt. The reaction was quenched by adding 6.4 molar equivalents N-Acetyl cysteine (10 mM in water). The ADC was buffer exchanged via G25 into PBS and washed by dead-end filtration (Vivaspin-20, 30 kDa MWCO, 0.0006 m 2 ) for 10 DVs. Samples were taken for analysis by HIC, SEC, PLRP, free toxin linker, Endosafe, and the concentration was determined using a SEC calibration curve. Aliquotting was carried out under laminar flow, and the product was stored at -8o°C. Only disposable, sterile and pyrogen/ DNA/RNA-free plasticware was used.

Antimicrobial Characterisation of Compounds 16, 45 and 73

The antimicrobial activity of a selection of compounds were evaluated in a Gram- negative, Gram-positive and multi-resistant bacteria using the standard minimum inhibitory concentration (MIC) assay for an overnight (16-20 hour) incubation period. Commercially marketed antibiotics (Table 3) were used as positive controls during the study.

Compounds 16, 45 and 73 were found to have potent activity against the three bacteria tested. In particular, 16 and 73 were observed to possess potent anti-bacterial effects against the gram negative strain E. coli K12, with potencies of 2 pg/mL determined. This was found to be in line with the commercially available compound Kanamycin (Kanamycin A).

Table 3: Antimicrobial activity of a selection of compounds against Gram-negative (e. coli K12), Gram-positive ( EMRSA 16) and multi-resistant ( klebsiella pneumoniae KP4631) bacterial strains Antimicrobial Characterisation Methodology

Materials

Reagents

Mueller-Hinton broth (MH Broth) (Sigma-Aldrich; Millipore, cat. no. 70192) and Mueller-Hinton agar (MHA) (Sigma-Aldrich; Millipore, cat. no. 70191) were prepared as per manufacturer’s instructions and sterilized by autoclaving. Approximately 20 mL of MHA was poured onto 15 x 100 mm petri dishes and allowed to set. Agar plates were then stored in plastic bags in an inverted position at 4°C.

Bacterial strains

Bacterial strains were provided in the form of a glycerol stock, stored at -8o°C. For the purpose of preparation of bacterial suspension, appropriate strains were removed from -8o°C storage onto dry ice and not allowed to freeze-thaw.

Compounds

Stock solutions of 16, 45 and 73 were provided in DMSO at 1.28 mg/ mL

concentration. From this stock solution, working solutions of the desired concentration were prepared by diluting with MHB.

Method

MIC assay methodology was adapted from previously published work [33].

Preparation of bacterial suspension for colony count

Bacterial isolate to be tested was streaked onto a nutrient-rich MH agar plate and incubated overnight at 37°C in order to obtain single colonies. After overnight incubation, single colonies with the same morphological appearance were chosen from the agar plate. These were touched using a sterile loop and transferred into a tube containing 10 mL of a MH broth. They were then mixed using vortex mixer and incubated at 35-37°C in a shaker at 200 r.p.m. overnight.

Determination ofOD6oo and cfu/mL count

OD600 (the optical density of a sample measured at a wavelength of 600 nm) of the overnight culture was measured. Because of the loss of linearity at OD600 values 41.0, it was necessary to dilute the sample until the OD600 value was below 1.0. For all tested bacteria, a dilution of 1:10 was appropriate (100 pL in 900 pL broth).

The overnight culture was then diluted 1:100 using sterile tubes and MH broth (dilution: 10 2 ). Serial dilution was then performed six times until dilution of io -8 was reached. 100 pL of dilutions icr 7 and icr 8 were plated onto MH agar plates in triplicate using a sterile cell spreader and incubated overnight at 37°C.

After overnight incubation, colonies on plates were counted and cfu per mL calculated. The following formula was applied:

C x 10

where, N = colony-forming unit/millilitre (cfu/mL), C = number of colonies per plater, D = number of the 1:10 dilution.

The average of three plates was calculated and this number was correlated with OD600 measurement. This relationship holds true for subsequent cultures of the same bacterium grown in the same way [33].

For the generation of overnight cultures to be used for preparation of the bacterial suspension for MIC tests, a single colony of bacteria was selected using a sterile loop and a sterile tube with MH broth was inoculated followed by overnight incubation at 35-27°C in a shaker set to 200 r.p.m.

After overnight incubation, the OD600 was measured and the overnight culture was diluted accordingly to obtain 1 x 10 8 cfu/mL.

Compound dilution preparation for MIC assay

Free payload dilutions were prepared as per Table 4.

202 Stage Antimicrobial Source Volume Volume Antimicrobial Final concentration of of concentration concentration

(mg/L) antibiotic sterile obtained in test

stock broth

solution (pL)

(pL)

1 1280 Stock 100 900 128 64

2 128 Stage 1 too too 64 32

3 128 Stage 1 too 300 32 16

4 128 Stage 1 too 700 16 8

5 16 Stage too too 8 4

4

6 16 Stage too 300 4 2

4

7 16 Stage too 700 2 1

4

8 2 Stage too too 1 0.5

7

9 2 Stage too 300 0.5 0.25

7

10 2 Stage too 700 0.25 0.125

11 0.25 Stage too too 0.125 0.06

10

12 0.25 Stage too 300 0.06 0.03

10

Table 4: Scheme for preparing dilutions of 16, 45 and 73 and positive controls for MIC assay.

MIC assay preparation

96-well sterile microtiter plates were used for MIC assay. The plate was filled with the respective free payload or positive control antibiotic concentrations in triplicate. 100 pL of broth was pipetted in the sterility control well (SC, column 12) and 50 pL in the growth control well (GC, column 11). l x 10 8 cfu/ mL bacterial suspension was diluted 1:100 and 50 pL added to each well apart from SC. Bacterial suspension was at the same time diluted to achieve 1 x to 4 and a x to 3 cfu/mL which was plated onto fresh MH agar plates for colony count by pipetting too uL onto agar plate and spread using a sterile cell spreader. The 96-well plates together with the MH agar plates were then incubated overnight (16-20 h) at 37°C.

Results

The 96-well plates were read using a SpectraMax plate reader at an absorbance of 600 nm. The results for compounds 16, 45 and 73 together with the data for antibiotics kanamycin, vancomycin and gentamicin are presented in Figure 16A, 16B, 16C, 16D, 17A, 17B, 17C, 17D, 18A, 18B, 18C and 18D.

REFERENCES

1. Antonow, D., and Thurston, D. E. (2011) Chem Rev 111, 2815-2864.

2. Cipolla, L., Araujo, A. C., Airoldi, C., and Bini, D. (2009) Anticancer Agents Med Chem 9, 1-31.

3. Gerratana, B. (2012) Med Res Rev 32, 254-293.

4. Hartley, J. A. (2011) Expert Opin Investig Drugs 20, 733-744.

5. Kamal, A., Reddy, K. L., Devaiah, V., Shankaraiah, N., and Reddy, D. R. (2006)

Mini Rev Med Chem 6, 53-69.

6. Hurley, L. H., Reck, T., Thurston, D. E., Langley, D. R., Holden, K. G.,

Hertzberg, R. P., Hoover, J. R., Gallagher, G., Jr., Faucette, L. F., Mong, S. M., (1988) Chem Res Toxicol 1, 258-268.

7. Wells, G., Martin, C. R., Howard, P. W., Sands, Z. A., Laughton, C. A.,

Tiberghien, A., Woo, C. K., Masterson, L. A., Stephenson, M. J., Hartley, J. A., Jenkins, T. C., Shnyder, S. D., Loadman, P. M., Waring, M. J., and Thurston, D. E. (2006) J Med Chem 49, 5442-5461.

8. Brucoli, F., Hawkins, R. M., James, C. H., Jackson, P. J., Wells, G., Jenkins, T. C., Ellis, T., Kotecha, M., Hochhauser, D., Hartley, J. A., Howard, P. W., and Thurston,

D. E. (2013) JMed Chem 56, 6339-6351.

9. Kotecha, M., Kluza, J., Wells, G., O'Hare, C. C., Forni, C., Mantovani, R., Howard, P. W., Morris, P., Thurston, D. E., Hartley, J. A., and Hochhauser, D. (2008) Mol Cancer Ther 7, 1319-1328.

10. Puwada, M. S., Hartley, J. A., Jenkins, T. C., and Thurston, D. E. (1993) Nucleic

Acids Res 21, 3671-3675.

11. Clingen, P. H., De Silva, I. U., McHugh, P. J., Ghadessy, F. J., Tilby, M. J., Thurston, D. E., and Hartley, J. A. (2005) Nucleic Acids Res 33, 3283-3291.

12. Puwada, M. S., Forrow, S. A., Hartley, J. A., Stephenson, P., Gibson, L, Jenkins, T. C., and Thurston, D. E. (1997) Biochemistry 36, 2478-2484.

13. Barkley, M. D., Cheatham, S., Thurston, D. E., and Hurley, L. H. (1986)

Biochemistry 25, 3021-3031.

14. Seifert, J., Pezeshki, S., Kamal, A., and Weisz, K. (2012) Organic &

Biomolecular Chemistry 10, 6850-6860.

15. Smellie, M., Bose, D. S., Thompson, A. S., Jenkins, T. C., Hartley, J. A., and

Thurston, D. E. (2003) Biochemistry 42, 8232-8239.

16. Kopka, M. L., Goodsell, D. S., Baikalov, L, Grzeskowiak, K., Cascio, D., and Dickerson, R. E. (1994) Biochemistry 33, 13593-13610.

17. Kizu, R., Draves, P. H., and Hurley, L. H. (1993) Biochemistry 32, 8712-8722. 18. Leimgruber, W., Stefanovic, V., Schenker, F., Karr, A., and Berger, J. (1965) J Am Chem Soc 87, 5791-5793.

19. Arima, K., Kosaka, M., Tamura, G., Imanaka, H., and Sakai, H. (1972) JAntibiot (Tokyo) 25, 437-444·

20. Sato, S., Iwata, F., Yamada, S., Kawahara, H., and Katayama, M. (2011) Bioorg Med Chem Lett 21, 7099-7101.

21. Thurston D.E. and Bose D.S., Chem Rev (1994); 94:433-465.

22. Damayanthi, Y., et al.; Journal of Organic Chemistry (1999), 64, 290-292;

23. Kumar, et al., Heterocyclic Communications (2002) 8, 19-26.

24. Kumar, R, Lown, J. W.; Oncology Research, (2003) 13, 221-233.

25. Baraldi, P. G. et al., Journal of Medicinal Chemistry (1999) 42, 5131-5141.

26. Wells, G., et al., Proc. Am. Assoc. Cane. Res. (2003) 44, 452.

27. Thurston, D. E.; Howard, P. W. WO 2004/043963.

28. Reddy, B. S., Sharma, S. K., Lown, J. W., Current Medicinal Chemistry (2001)

8, 475-508.

29. Antonow, D., Jenkins, T. C., Howard, P. W., Thurston, D. E., Bioorganic & Medicinal Chemistry (2007) 15, 3041-3053.

30. Blencowe, C. A., Russell, A. T., Greco, F., Hayes, W., Thornthwaite, D.W., Polymer Chemistry (2011) 2, 773-790

31. Hampshire, A. J., Rusling, D. A., Broughton-Head, V. J., Fox, K. R., Methods (2007), 42, 128-140.

32. Drew, H. R., Travers, A. A., Cell (1984) 37, 491-502.

33. Wiegand, L, Hilpert, K., Hancock, R. E., Nature protocols 2008, 3, 163-175.

All publications mentioned in the above specification are herein incorporated by reference. Although illustrative embodiments of the invention have been disclosed in detail herein, with reference to the accompanying drawings, it is understood that the invention is not limited to the precise embodiment and that various changes and modifications can be effected therein by one skilled in the art without departing from the scope of the invention as defined by the appended claims and their equivalents.