Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PYRROLOBENZODIAZEPINES AND CONJUGATES THEREOF AS ANTITUMOUR AGENTS
Document Type and Number:
WIPO Patent Application WO/2019/197602
Kind Code:
A1
Abstract:
A compound with the formula I: (I) and salts and solvates thereof, wherein: R" is a group of formula II: (II) where each of n and m are independently selected from 1, 2 and 3.

Inventors:
HOWARD PHILIP (GB)
DICKINSON NIALL (GB)
Application Number:
PCT/EP2019/059404
Publication Date:
October 17, 2019
Filing Date:
April 12, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MEDIMMUNE LTD (GB)
International Classes:
C07D519/00; A61K31/551; A61P35/00; C07K5/062
Domestic Patent References:
WO2014159981A22014-10-02
WO2013041606A12013-03-28
Foreign References:
US20140155590A12014-06-05
Attorney, Agent or Firm:
MEWBURN ELLIS LLP (GB)
Download PDF:
Claims:
Claims

1 . A compound with the formula I:

and salts and solvates thereof, wherein:

R" is a group of formula II:

where each of n and m are independently selected from 1 , 2 and 3;

R° is selected from the group consisting of H, methyl, ethyl, iso-propyl and benzyl;

Qc is selected from N and CH;

Y and Y’ are selected from O, S, or NH;

when there is a double bond present between C2 and C3, R2 is selected from the group consisting of:

(ia) C5-10 aryl group, optionally substituted by one or more substituents selected from the group comprising: halo, nitro, cyano, ether, carboxy, ester, C1-7 alkyl, C3-7 heterocyclyl and bis-oxy-Ci-3 alkylene;

(ib) C1-5 saturated aliphatic alkyl;

(ic) C3-6 saturated cycloalkyl;

, wherein each of R11, R12 and R13 are independently selected from H, C1-3 saturated alkyl, C2-3 alkenyl, C2-3 alkynyl and cyclopropyl, where the total number of carbon atoms in the R2 group is no more than 5;

(ie) , wherein one of R15a and R15b is H and the other is selected from: phenyl, which phenyl is optionally substituted by a group selected from halo, methyl, methoxy; pyridyl; and thiophenyl; and , where R14 is selected from: H; C1-3 saturated alkyl; C2-3 alkenyl; C2-3 alkynyl; cyclopropyl; phenyl, which phenyl is optionally substituted by a group selected from halo, methyl, methoxy; pyridyl; and thiophenyl;

when there is a single bond present between C2 and C3,

R2 is H or , where R16a and R16b are independently selected from H, F, Ci-4 saturated alkyl, C2-3 alkenyl, which alkyl and alkenyl groups are optionally substituted by a group selected from C1-4 alkyl amido and Ci-4 alkyl ester; or, when one of R16a and R16b is H, the other is selected from nitrile and a Ci-4 alkyl ester;

when there is a double bond present between C2’ and C3’, R2 is selected from the group consisting of:

(iia) C5-10 aryl group, optionally substituted by one or more substituents selected from the group comprising: halo, nitro, cyano, ether, carboxy, ester, C1-7 alkyl, C3-7 heterocyclyl and bis-oxy-Ci-3 alkylene;

(iib) C1-5 saturated aliphatic alkyl;

(iic) C3-6 saturated cycloalkyl;

wherein each of R21, R22 and R23 are independently selected from H, C1-3 saturated alkyl, C2-3 alkenyl, C2-3 alkynyl and cyclopropyl, where the total number of carbon atoms in the R2 group is no more than 5;

,25b

R

(iie) , wherein one of R25a and R25b is H and the other is selected from: phenyl, which phenyl is optionally substituted by a group selected from halo, methyl, methoxy;

pyridyl; and thiophenyl; and

(iif) R' , where R24 is selected from: H; C1-3 saturated alkyl; C2-3 alkenyl; C2-3 alkynyl; cyclopropyl; phenyl, which phenyl is optionally substituted by a group selected from halo, methyl, methoxy; pyridyl; and thiophenyl;

when there is a single bond present between C2’ and C3’,

R2’ is H or , where R26a and R26b are independently selected from H, F, Ci-4 saturated alkyl, C2-3 alkenyl, which alkyl and alkenyl groups are optionally substituted by a group selected from Ci-4 alkyl amido and Ci-4 alkyl ester; or, when one of R26a and R26b is H, the other is selected from nitrile and a Ci-4 alkyl ester;

R6 and R9 are independently selected from H, R, OH, OR, SH, SR, NH2, NHR, NRR’, nitro, MesSn and halo;

where R and R’ are independently selected from optionally substituted C1-12 alkyl, C3-20 heterocyclyl and C5-20 aryl groups;

R7 is selected from H, R, OH, OR, SH, SR, NH2, NHR, NHRR’, nitro, MesSn and halo;

R6 , R7 , R9 are selected from the same groups as R6, R7 and R9 respectively;

R11a is selected from OH, ORA, where RA is Ci-4 alkyl, and SOzM, where z is 2 or 3 and M is a monovalent pharmaceutically acceptable cation;

and either

(a) R30 is H, and R31 is OH, ORA, where RA is Ci-4 alkyl;

(b) R30 and R31 form a nitrogen-carbon double bond between the nitrogen and carbon atoms to which they are bound; or

(c) R30 is H and R31 is SOzM, where z is 2 or 3 and M is a monovalent pharmaceutically acceptable cation, wherein if R11a and R31 are SOzM, M may represent a divalent pharmaceutically acceptable cation

RL is a linker for connection to a cell binding agent, which is selected from:

(ilia):

wherein

Q is:

, where Qx is such that Q is an amino-acid residue, a dipeptide residue or a tripeptide residue;

X is:

where a = 0 to 5, b = 0 to 16, c = 0 or 1 , d = 0 to 5;

GL is a linker for connecting to a Ligand Unit; and

(iiib): where RL1 and RL2 are independently selected from H and methyl, or together with the carbon atom to which they are bound form a cyclopropylene or cyclobutylene group;

and e is 0 or 1.

2. A compound according to claim 1 , wherein Y and Y’ are O. 3. A compound according to either claim 1 or claim 2, wherein R° is H.

4. A compound according to either claim 1 or claim 2, wherein R° is methyl.

5. A compound according to claim 1 , wherein R" is of formula I la:

where R°1 is selected from the group consisting of H and methyl.

6. A compound according to any one of claims 1 to 5, wherein R6, R9, R6’ and R9 are H, and R7 and R7 are methoxy.

7. A compound according to any one of claims 1 to 6, wherein R11a is OH. 8. A compound according to any one of claims 1 to 7, wherein R30 and R31 form a nitrogen-carbon double bond between the nitrogen and carbon atoms to which they are bound.

9. A compound according to claim 1 of:

(i) formula la:

where R2a and R2a are the same and are selected from:

(iii) formula lc:

wherein R7a and R7a are the same and are selected from methoxy and benzyloxy.

10. A compound according to any one of claims 1 to 9, wherein RL is of formula Ilia, and Q is a dipeptide residue selected from:

co-Phe-Lys-NH,

co-Val-Ala-NH,

co-Val-Lys-NH,

co-Ala-Lys-NH,

co-Val-Cit-NH,

co-Phe-Cit-NH,

co-Leu-Cit-NH,

co-lle-Cit-NH,

co-Phe-Arg-NH, and co-Trp-Cit-NH.

1 1. A compound according to any one of claims 1 to 10, wherein RL is of formula Ilia and, a is 0, c is 1 and d is 2, and b is from 0 to 8.

12. A compound according to any one of claims 1 to 11 , wherein RL is of formula Ilia and GL is selected from

where Ar represents a C5-6 arylene group, e.g. phenylene, and X represents C1-4 alkyl.

13. A compound according to any one of claims 1 to 9, wherein RL is of formula I Mb. 14. A conjugate comprising a compound of formula I according to any one of claims 1 to

13, or a pharmaceutically acceptable salt or solvate thereof, linked to a Ligand unit.

15. A conjugate of formula IV:

L - (DL)P (IV)

or a pharmaceutically acceptable salt or solvate thereof, wherein L is a Ligand unit , DL is of formula III:

wherein R2, R6, R7, R9, R11a, Y, R”, Y’, R2’, R6’, R7’, R9’, R30 and R31 are as defined in any one of claims 1 to 8;

RLL is a linker for connection to a cell binding agent, which is selected from:

(iiia): where Q and X are as defined in any one of claims 1 , 10 and 1 1 and GLL is a linker connected to a Ligand Unit; and

(iiib): mb'

where RL1 and RL2 are as defined in claim 1 ;

wherein p is an integer of from 1 to 20.

16. A conjugate according to claim 15, wherein GLL is selected from:

17. A conjugate according to claim 15, wherein DL is of: (i) formula Ilia:

where R2a and R2a are the same and are selected from:

(iii) formula lllc:

where R7a and R7a are the same and are selected from methoxy and benzyloxy.

18. A conjugate according to any one of claims 14 to 17, wherein the Ligand Unit is an antibody or an active fragment thereof.

19. The conjugate according to claim 18, wherein the antibody or antibody fragment is an antibody which binds to one or more tumor-associated antigens or cell-surface receptors selected from (1 )-(88):

(1 ) BMPR1 B;

(2) E16;

(3) STEAP1 ;

(4) 0772P;

(5) MPF;

(6) Napi3b;

(7) Serna 5b;

(8) PSCA hlg;

(9) ETBR;

(10) MSG783;

(11 ) STEAP2;

(12) TrpM4;

(13) CRIPTO;

(14) CD21 ;

(15) CD79b;

(16) FcRH2;

(17) HER2;

(18) NCA;

(19) MDP;

(20) IL20R-alpha;

(21 ) Brevican;

(22) EphB2R;

(23) ASLG659;

(24) PSCA;

(25) GEDA;

(26) BAFF-R;

(27) CD22;

(28) CD79a;

(29) CXCR5;

(30) HLA-DOB;

(31 ) P2X5;

(32) CD72; (33) LY64;

(34) FcRM ;

(35) IRTA2;

(36) TENB2;

(37) PSMA - FOLH1 ;

(38) SST;

(38.1 ) SSTR2;

(38.2) SSTR5;

(38.3) SSTR1 ;

(38.4) SSTR3;

(38.5) SSTR4;

(39) ITGAV;

(40) ITGB6;

(41) CEACAM5;

(42) MET;

(43) MUC1 ;

(44) CA9;

(45) EGFRvlll;

(46) CD33;

(47) CD19;

(48) IL2RA;

(49) AXL;

(50) CD30 - TNFRSF8;

(51) BCMA - TNFRSF17;

(52) CT Ags - CTA;

(53) CD174 (Lewis Y) - FUT3;

(54) CLEC14A;

(55) GRP78 - HSPA5;

(56) CD70;

(57) Stem Cell specific antigens;

(58) ASG-5;

(59) ENPP3;

(60) PRR4;

(61) GCC - GUCY2C;

(62) Liv-1 - SLC39A6;

(63) 5T4; (64) CD56 - NCMA1 ;

(65) CanAg;

(66) FOLR1 ;

(67) GPNMB;

(68) TIM-1 - HAVCR1 ;

(69) RG-1/Prostate tumor target Mindin - Mindin/RG-1 ;

(70) B7-H4 - VTCN1 ;

(71) PTK7;

(72) CD37;

(73) CD138 - SDC1 ;

(74) CD74;

(75) Claudins - CLs;

(76) EGFR;

(77) Her3;

(78) RON - MST1 R;

(79) EPHA2;

(80) CD20 - MS4A1 ;

(81) Tenascin C - TNC;

(82) FAP;

(83) DKK-1 ;

(84) CD52;

(85) CS1 - SLAMF7;

(86) Endoglin - ENG;

(87) Annexin A1 - ANXA1 ;

(88) V-CAM (CD106) - VCAM1.

20. A composition comprising a mixture of conjugates according to any one of claims 15 to 19, wherein the average p in the mixture of conjugate compounds is about 1 to about 8.

21. The conjugate according to any one of claims 14 to 19, for use in therapy.

22. A pharmaceutical composition comprising the conjugate of any one of claims 14 to 19 and a pharmaceutically acceptable diluent, carrier or excipient.

23. The conjugate according to any one of claims 14 to 19 or the pharmaceutical composition according to claim 22, for use in the treatment of a proliferative disease in a subject. 24. A compound with the formula REL:

and salts and solvates thereof, wherein R2, R6, R7, R9, Y, R”, Y’, R2’, R6’, R7’, R9’, R30 and R31 are as defined in any one of claims 1 to 8.

Description:
PYRROLOBENZODIAZEPINES AND CONJUGATES THEREOF AS

ANTITUMOUR AGENTS

The present invention relates to pyrrolobenzodiazepines (PBDs), and their inclusion in targeted conjugates. The PBDs of the present invention are dimer where the group linking the two PBD moieties comprises a benzene or pyridine ring substituted by a carboxy or ester group.

Background to the invention

Pyrrolobenzodiazepines

Some pyrrolobenzodiazepines (PBDs) have the ability to recognise and bond to specific sequences of DNA; the preferred sequence is PuGPu. The first PBD antitumour antibiotic, anthramycin, was discovered in 1965 (Leimgruber, et al., J. Am. Chem. Soc., 87, 5793-5795 (1965); Leimgruber, et al., J. Am. Chem. Soc., 87, 5791-5793 (1965)). Since then, a number of naturally occurring PBDs have been reported, and over 10 synthetic routes have been developed to a variety of analogues (Thurston, et al., Chem. Rev. 1994, 433-465 (1994)). Family members include abbeymycin (Hochlowski, et al., J. Antibiotics, 40, 145-148 (1987)), chicamycin (Konishi, et ai., J. Antibiotics, 37, 200-206 (1984)), DC-81 (Japanese Patent 58- I SO 487; Thurston, et ai., Chem. Brit., 26, 767-772 (1990); Bose, et ai., Tetrahedron, 48, 751-758 (1992)), mazethramycin (Kuminoto, et ai., J. Antibiotics, 33, 665-667 (1980)), neothramycins A and B (Takeuchi, et al., J. Antibiotics, 29, 93-96 (1976)), porothramycin (Tsunakawa, et al., J. Antibiotics, 41 , 1366-1373 (1988)), prothracarcin (Shimizu, et al, J. Antibiotics, 29, 2492-2503 (1982); Langley and Thurston, J. Org. Chem., 52, 91-97 (1987)), sibanomicin (DC-102)(Hara, et al., J. Antibiotics, 41 , 702-704 (1988); Itoh, et al., J.

Antibiotics, 41 , 1281-1284 (1988)), sibiromycin (Leber, et al., J. Am. Chem. Soc., 110, 2992- 2993 (1988)) and tomamycin (Arima, et al., J. Antibiotics, 25, 437-444 (1972)). PBDs are of the general structure:

They differ in the number, type and position of substituents, in both their aromatic A rings and pyrrolo C rings, and in the degree of saturation of the C ring. In the B-ring there is either an imine (N=C), a carbinolamine(NH-CH(OH)), or a carbinolamine methyl ether (NH- CH(OMe)) at the N10-C1 1 position which is the electrophilic centre responsible for alkylating DNA. All of the known natural products have an (S)-configuration at the chiral C11 a position which provides them with a right-handed twist when viewed from the C ring towards the A ring. This gives them the appropriate three-dimensional shape for isohelicity with the minor groove of B-form DNA, leading to a snug fit at the binding site (Kohn, In Antibiotics III.

Springer-Verlag, New York, pp. 3-1 1 (1975); Hurley and Needham-VanDevanter, Acc.

Chem. Res., 19, 230-237 (1986)). Their ability to form an adduct in the minor groove, enables them to interfere with DNA processing, hence their use as antitumour agents. A particularly advantageous pyrrolobenzodiazepine compound is described by Gregson et al. (Chem. Commun. 1999, 797-798) as compound 1 , and by Gregson et al. ( J . Med. Chem. 2001 , 44, 1 161-1174) as compound 4a. This compound, also known as SG2000, is shown below:

WO 2007/085930 describes the preparation of dimer PBD compounds having linker groups for connection to a cell binding agent, such as an antibody. The linker is present in the bridge linking the monomer PBD units of the dimer.

Dimer PBD compounds having linker groups for connection to a cell binding agent, such as an antibody, are described in WO 201 1/130598. The linker in these compounds is attached to one of the available N10 positions, and are generally cleaved by action of an enzyme on the linker group. More recently, the warhead:

has been used in drug linkers and antibody-drug conjugates. WO 2014/057074 discloses:

WO2015/052322 discloses

A number of disclosures, such as US2014/155590, WO2014/159981 , WO2014/140862, and WO 2016/038383 disclose PBD dimers linked from an aromatic group in the tether, such as compound 33 in WO 2014/159981

as well as intermediates in their synthesis.

Disclosure of the Invention

The present invention provides PBD dimers where the group linking the two PBD moieties comprises a benzene or pyridine ring substituted by a carboxy or ester group, as drug linkers, and conjugates made from these drug linkers, as well as the released drugs.

The presence of the carboxy group or ester group should reduce the activity of the drug linker and warhead compared to the analogous drug linkers and warheads being unsubstituted. However, the conjugate with an ester group shows good activity in vitro and in vivo of a level which is comparable to the analogous unsubstituted conjugates. Without wishing to be bound by theory, it is thought that the carboxy group may limit the ability of the warhead to enter the cell when not conjugated.

A first aspect of the present invention comprises a compound with the formula I:

and salts and solvates thereof, wherein:

R" is a group of formula II:

where each of n and m are independently selected from 1 , 2 and 3;

R° is selected from the group consisting of H, methyl, ethyl, iso-propyl and benzyl;

Q c is selected from N and CH;

Y and Y’ are selected from O, S, or NH;

when there is a double bond present between C2 and C3, R 2 is selected from the group consisting of:

(ia) C 5-10 aryl group, optionally substituted by one or more substituents selected from the group comprising: halo, nitro, cyano, ether, carboxy, ester, C 1-7 alkyl, C 3-7 heterocyclyl and bis-oxy-Ci-3 alkylene;

(ib) C 1-5 saturated aliphatic alkyl;

(ic) C3-6 saturated cycloalkyl;

, wherein each of R 11 , R 12 and R 13 are independently selected from H, C1-3 saturated alkyl, C2-3 alkenyl, C2-3 alkynyl and cyclopropyl, where the total number of carbon atoms in the R 2 group is no more than 5;

(ie) , wherein one of R 15a and R 15b is H and the other is selected from: phenyl, which phenyl is optionally substituted by a group selected from halo, methyl, methoxy; pyridyl; and thiophenyl; and

(if) , where R 14 is selected from: H; C1-3 saturated alkyl; C2-3 alkenyl; C2-3 alkynyl; cyclopropyl; phenyl, which phenyl is optionally substituted by a group selected from halo, methyl, methoxy; pyridyl; and thiophenyl; when there is a single bond present between C2 and C3,

R 2 , where R 16a and R 16b are independently selected from H, F, Ci -4 saturated alkyl, C2-3 alkenyl, which alkyl and alkenyl groups are optionally substituted by a group selected from Ci -4 alkyl amido and Ci -4 alkyl ester; or, when one of R 16a and R 16b is H, the other is selected from nitrile and a Ci -4 alkyl ester;

when there is a double bond present between C2’ and C3’, R 2 is selected from the group consisting of:

(iia) C 5-10 aryl group, optionally substituted by one or more substituents selected from the group comprising: halo, nitro, cyano, ether, carboxy, ester, C 1-7 alkyl, C 3-7 heterocyclyl and bis-oxy-Ci-3 alkylene;

(iib) C 1-5 saturated aliphatic alkyl;

(iic) C 3-6 saturated cycloalkyl;

(iid) , wherein each of R 21 , R 22 and R 23 are independently selected from H,

C 1-3 saturated alkyl, C 2-3 alkenyl, C 2-3 alkynyl and cyclopropyl, where the total number of carbon atoms in the R 2 group is no more than 5;

(iie) , wherein one of R 25a and R 25b is H and the other is selected from: phenyl, which phenyl is optionally substituted by a group selected from halo, methyl, methoxy; pyridyl; and thiophenyl; and

(iif) , where R 24 is selected from: H; C1-3 saturated alkyl; C2-3 alkenyl; C2-3 alkynyl; cyclopropyl; phenyl, which phenyl is optionally substituted by a group selected from halo, methyl, methoxy; pyridyl; and thiophenyl;

when there is a single bond present between C2’ and C3’,

R 2’ is H or , where R 26a and R 26b are independently selected from H, F, Ci -4 saturated alkyl, C2-3 alkenyl, which alkyl and alkenyl groups are optionally substituted by a group selected from Ci -4 alkyl amido and Ci -4 alkyl ester; or, when one of R 26a and R 26b is H, the other is selected from nitrile and a Ci -4 alkyl ester;

R 6 and R 9 are independently selected from H, R, OH, OR, SH, SR, NH2, NHR, NRR’, nitro, MesSn and halo;

where R and R’ are independently selected from optionally substituted C1-12 alkyl, C3-20 heterocyclyl and C5-20 aryl groups;

R 7 is selected from H, R, OH, OR, SH, SR, NH2, NHR, NHRR’, nitro, MesSn and halo;

R 6 , R 7 , R 9 are selected from the same groups as R 6 , R 7 and R 9 respectively;

R 11a is selected from OH, OR A , where R A is Ci -4 alkyl, and SO z M, where z is 2 or 3 and M is a monovalent pharmaceutically acceptable cation;

and either

(a) R 30 is H, and R 31 is OH, OR A , where R A is Ci -4 alkyl;

(b) R 30 and R 31 form a nitrogen-carbon double bond between the nitrogen and carbon atoms to which they are bound; or

(c) R 30 is H and R 31 is SO z M, where z is 2 or 3 and M is a monovalent pharmaceutically acceptable cation, wherein if R 11a and R 31 are SO z M, M may represent a divalent pharmaceutically acceptable cation

R L is a linker for connection to a cell binding agent, which is selected from:

(ilia):

wherein

Q is:

, where Q x is such that Q is an amino-acid residue, a dipeptide residue or a tripeptide residue;

X is:

where a = 0 to 5, b = 0 to 16, c = 0 or 1 , d = 0 to 5;

G L is a linker for connecting to a Ligand Unit; and (iiib): where R L1 and R L2 are independently selected from H and methyl, or together with the carbon atom to which they are bound form a cyclopropylene or cyclobutylene group;

and e is O oM .

A second aspect of the present invention provides a method of making a compound of the first aspect of the invention, comprising at least one of the method steps set out below. In a third aspect, the present invention relates to Conjugates comprising dimers of PBDs linked to a Ligand unit/targeting agent, wherein the PBD dimer is a derivative of formula I, or a pharmaceutically acceptable salt or solvate thereof.

In some embodiments, the Conjugates having the following formula IV:

L - (D L ) P (IV)

or a pharmaceutically acceptable salt or solvate thereof, wherein L is a Ligand unit (i.e., a targeting agent), D L is a Drug Linker unit that is a PBD dimer of formula III:

wherein R 2 , R 6 , R 7 , R 9 , R 11a , Y, R”, Y’, R 2’ , R 6’ , R 7’ , R 9’ , R 30 and R 31 are as defined in the first aspect of the invention;

R LL is a linker for connection to a cell binding agent, which is selected from:

(iiia):

where Q and X are as defined in the first aspect and G LL is a linker connected to a Ligand

Unit; and

(iiib): where R L1 and R L2 are as defined in the first aspect;

wherein p is an integer of from 1 to 20.

Accordingly, the Conjugates comprise a Ligand unit covalently linked to at least one Drug unit by a Linker unit. The Ligand unit, described more fully below, is a targeting agent that binds to a target moiety. The Ligand unit can, for example, specifically bind to a cell component (a Cell Binding Agent) or to other target molecules of interest. Accordingly, the present invention also provides methods for the treatment of, for example, various cancers and autoimmune disease. These methods encompass the use of the Conjugates wherein the Ligand unit is a targeting agent that specifically binds to a target molecule. The Ligand unit can be, for example, a protein, polypeptide or peptide, such as an antibody, an antigen- binding fragment of an antibody, or other binding agent, such as an Fc fusion protein.

The drug loading is represented by p, the number of drug molecules per Ligand unit (e.g., an antibody). Drug loading may range from 1 to 20 Drug units (D) per Ligand unit (e.g., Ab or mAb). For compositions, p represents the average drug loading of the Conjugates in the composition, and p ranges from 1 to 20.

A fourth aspect of the present invention provides the use of a conjugate of the third aspect of the invention in the manufacture of a medicament for treating a proliferative disease. The fourth aspect also provides a conjugate of the third aspect of the invention for use in the treatment of a proliferative disease.

One of ordinary skill in the art is readily able to determine whether or not a candidate compound treats a proliferative condition for any particular cell type. For example, assays which may conveniently be used to assess the activity offered by a particular compound are described in the examples below. A further aspect of the invention provides compounds with the formula REL:

and salts and solvates thereof, where all the substituents are as defined above.

Definitions

Pharmaceutically acceptable cations

Examples of pharmaceutically acceptable monovalent and divalent cations are discussed in Berge, et al., J. Pharm. Sci., 66, 1-19 (1977), which is incorporated herein by reference. The pharmaceutically acceptable cation may be inorganic or organic.

Examples of pharmaceutically acceptable monovalent inorganic cations include, but are not limited to, alkali metal ions such as Na + and K + . Examples of pharmaceutically acceptable divalent inorganic cations include, but are not limited to, alkaline earth cations such as Ca 2+ and Mg 2+ . Examples of pharmaceutically acceptable organic cations include, but are not limited to, ammonium ion (i.e. NH 4 + ) and substituted ammonium ions (e.g. NH 3 R + , NhhF^, NHR3 + , NR 4 + ). 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 + .

Substituents

The phrase“optionally substituted” as used herein, pertains to a parent group which may be unsubstituted or which may be substituted.

Unless otherwise specified, the term“substituted” as used herein, pertains to a parent group which bears one or more substituents. The term“substituent” is used herein in the conventional sense and refers to a chemical moiety which is covalently attached to, or if appropriate, fused to, a parent group. A wide variety of substituents are well known, and methods for their formation and introduction into a variety of parent groups are also well known.

Examples of substituents are described in more detail below.

C-i- 12 alkyl: The term“C 1-12 alkyl” as used herein, pertains to a monovalent moiety obtained by removing a hydrogen atom from a carbon atom of a hydrocarbon compound having from 1 to 12 carbon atoms, which may be aliphatic or alicyclic, and which may be saturated or unsaturated (e.g. partially unsaturated, fully unsaturated). The term“Ci -n alkyl” as used herein, pertains to a monovalent moiety obtained by removing a hydrogen atom from a carbon atom of a hydrocarbon compound having from 1 to n carbon atoms, which may be aliphatic or alicyclic, and which may be saturated or unsaturated (e.g. partially unsaturated, fully unsaturated). Thus, the term“alkyl” includes the sub-classes alkenyl, alkynyl, cycloalkyl, etc., discussed below.

Examples of saturated alkyl groups include, but are not limited to, methyl (C 1 ), ethyl (C 2 ), propyl (C3), butyl (C 4 ), pentyl (C5), hexyl (OQ) and heptyl (C7).

Examples of saturated linear alkyl groups include, but are not limited to, methyl (C 1 ), ethyl (C 2 ), n-propyl (C 3 ), n-butyl (C 4 ), n-pentyl (amyl) (C 5 ), n-hexyl (O Q ) and n-heptyl (C 7 ).

Examples of saturated branched alkyl groups include iso-propyl (C 3 ), iso-butyl (C 4 ), sec-butyl (C 4 ), tert-butyl (C 4 ), iso-pentyl (C 5 ), and neo-pentyl (C 5 ).

C 2-12 Alkenyl: The term“C 2-12 alkenyl” as used herein, pertains to an alkyl group having one or more carbon-carbon double bonds.

Examples of unsaturated alkenyl groups include, but are not limited to, ethenyl (vinyl, - CH=CH 2 ), 1 -propenyl (-CH=CH-CH 3 ), 2-propenyl (allyl, -CH-CH=CH 2 ), isopropenyl (1 - methylvinyl, -C(CH 3 )=CH 2 ), butenyl (C 4 ), pentenyl (C 5 ), and hexenyl (Ce).

C 2-12 alkynyl: The term“C 2-12 alkynyl” as used herein, pertains to an alkyl group having one or more carbon-carbon triple bonds.

Examples of unsaturated alkynyl groups include, but are not limited to, ethynyl (-CºCH) and 2-propynyl (propargyl, -CH2-CºCH). C3-12 cycloalkyl: The term“C3-12 cycloalkyl” as used herein, pertains to an alkyl group which is also a cyclyl group; that is, a monovalent moiety obtained by removing a hydrogen atom from an alicyclic ring atom of a cyclic hydrocarbon (carbocyclic) compound, which moiety has from 3 to 7 carbon atoms, including from 3 to 7 ring atoms.

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

saturated monocyclic hydrocarbon compounds:

cyclopropane (C3), cyclobutane (C 4 ), cyclopentane (C5), cyclohexane {Ce), cycloheptane (C7), methylcyclopropane (C 4 ), dimethylcyclopropane (C5), methylcyclobutane (C5), dimethylcyclobutane {Ce), methylcyclopentane {Ce), dimethylcyclopentane (C7) and methylcyclohexane (C7);

unsaturated monocyclic hydrocarbon compounds:

cyclopropene (C3), cyclobutene (C 4 ), cyclopentene (C5), cyclohexene {Ce),

methylcyclopropene (C 4 ), dimethylcyclopropene (C5), methylcyclobutene (C5),

dimethylcyclobutene {Ce), methylcyclopentene {Ce), dimethylcyclopentene (C7) and methylcyclohexene (C7); and

saturated polycyclic hydrocarbon compounds:

norcarane (C7), norpinane (C7), norbornane (C7).

C3-20 heterocyclyl: The term“C3-20 heterocyclyl” as used herein, pertains to a monovalent moiety obtained by removing a hydrogen atom from a ring atom of a heterocyclic compound, which moiety has from 3 to 20 ring atoms, of which from 1 to 10 are ring heteroatoms.

Preferably, each ring has from 3 to 7 ring atoms, of which from 1 to 4 are ring heteroatoms.

In this context, the prefixes (e.g. C3-20, C3-7, C5-6, etc.) denote the number of ring atoms, or range of number of ring atoms, whether carbon atoms or heteroatoms. For example, the term“Cs-eheterocyclyl”, as used herein, pertains to a heterocyclyl group having 5 or 6 ring atoms.

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

Ni: aziridine (C3), azetidine (C 4 ), pyrrolidine (tetrahydropyrrole) (C5), pyrroline (e.g.,

3-pyrroline, 2,5-dihydropyrrole) (C5), 2H-pyrrole or 3H-pyrrole (isopyrrole, isoazole) (C5), piperidine {Ce), dihydropyridine {Ce), tetrahydropyridine {Ce), azepine (C7); Oi: oxirane (C3), oxetane (C 4 ), oxolane (tetrahydrofuran) (C5), oxole (dihydrofuran) (C5), oxane (tetrahydropyran) (Ce), dihydropyran (Ce), pyran (Ce), oxepin (C7);

Si: thiirane (C3), thietane (C 4 ), thiolane (tetrahydrothiophene) (C5), thiane

(tetrahydrothiopyran) (Ce), thiepane (C7);

0 2 : dioxolane (C5), dioxane (Ce), and dioxepane (C7);

O3: trioxane (Ce) ' ,

N 2 : imidazolidine (C5), pyrazolidine (diazolidine) (C5), imidazoline (C5), pyrazoline

(dihydropyrazole) (C5), piperazine (Ce) ' ,

N 1 O 1 : tetrahydrooxazole (C 5 ), dihydrooxazole (C 5 ), tetrahydroisoxazole (C 5 ),

dihydroisoxazole (C5), morpholine (Ce), tetrahydrooxazine (Ce), dihydrooxazine (Ce), oxazine (Ce);

N1S1: thiazoline (C5), thiazolidine (C5), thiomorpholine (Ce) ' ,

N2O1: oxadiazine (Ce) ' ,

O1S1: oxathiole (C5) and oxathiane (thioxane) (Ce) ' , and,

N1O1S1: oxathiazine (O Q ).

Examples of substituted monocyclic heterocyclyl groups include those derived from saccharides, in cyclic form, for example, furanoses (C5), such as arabinofuranose, lyxofuranose, ribofuranose, and xylofuranse, and pyranoses (Ce), such as allopyranose, altropyranose, glucopyranose, mannopyranose, gulopyranose, idopyranose,

galactopyranose, and talopyranose.

C5-20 aryl: The term“C5-20 aryl”, as used herein, pertains to a monovalent moiety obtained by removing a hydrogen atom from an aromatic ring atom of an aromatic compound, which moiety has from 3 to 20 ring atoms. The term“C5-7 aryl”, as used herein, pertains to a monovalent moiety obtained by removing a hydrogen atom from an aromatic ring atom of an aromatic compound, which moiety has from 5 to 7 ring atoms and the term“C5-10 aryl”, as used herein, pertains to a monovalent moiety obtained by removing a hydrogen atom from an aromatic ring atom of an aromatic compound, which moiety has from 5 to 10 ring atoms. Preferably, each ring has from 5 to 7 ring atoms.

In this context, the prefixes (e.g. C3-20, C5-7, C5-6, C5-10, etc.) denote the number of ring atoms, or range of number of ring atoms, whether carbon atoms or heteroatoms. For example, the term“Cs-e aryl” as used herein, pertains to an aryl group having 5 or 6 ring atoms.

The ring atoms may be all carbon atoms, as in“carboaryl groups”. Examples of carboaryl groups include, but are not limited to, those derived from benzene (i.e. phenyl) (Ce), naphthalene (C10), azulene (Cio), anthracene (CM), phenanthrene (CM), naphthacene (Cis), and pyrene (OIQ).

Examples of aryl groups which comprise fused rings, at least one of which is an aromatic ring, include, but are not limited to, groups derived from indane (e.g. 2,3-dihydro-1 H-indene) (Cg), indene (Cg), isoindene (Cg), tetraline (1 ,2,3,4-tetrahydronaphthalene (Cio),

acenaphthene (C12), fluorene (C13), phenalene (C13), acephenanthrene (C15), and aceanthrene (OIQ).

Alternatively, the ring atoms may include one or more heteroatoms, as in“heteroaryl groups”. Examples of monocyclic heteroaryl groups include, but are not limited to, those derived from:

Ni: pyrrole (azole) (C5), pyridine (azine) {Ce),

O1: furan (oxole) (C5);

Si: thiophene (thiole) (C5);

N1O1: oxazole (C5), isoxazole (C5), isoxazine (Ce);

N2O1: oxadiazole (furazan) (C 5 );

N3O1 : oxatriazole (C5);

N1S1: thiazole (C5), isothiazole (C5);

N2: imidazole (1 ,3-diazole) (C5), pyrazole (1 ,2-diazole) (C5), pyridazine (1 ,2-diazine) (Ce), pyrimidine (1 ,3-diazine) (Ce) (e.g., cytosine, thymine, uracil), pyrazine (1 ,4-diazine) (Ce);

N3: triazole (C5), triazine (Ce); and,

N 4 : tetrazole (C 5 ).

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

Cg (with 2 fused rings) derived from benzofuran (O1), isobenzofuran (O1), indole (N1), isoindole (N1), indolizine (N1), indoline (N1), isoindoline (N1), purine (N 4 ) (e.g., adenine, guanine), benzimidazole (N2), indazole (N2), benzoxazole (N1O1), benzisoxazole (N1O1), benzodioxole (O2), benzofurazan (N2O1), benzotriazole (N 3 ), benzothiofuran (Si), benzothiazole (N1S1), benzothiadiazole (N2S);

Cio (with 2 fused rings) derived from chromene (O1), isochromene (O1), chroman (O1), isochroman (O1), benzodioxan (O2), quinoline (N1), isoquinoline (N1), quinolizine (N1), benzoxazine (N1O1), benzodiazine (N2), pyridopyridine (N2), quinoxaline (N2), quinazoline (N2), cinnoline (N2), phthalazine (N2), naphthyridine (N2), pteridine (N 4 );

C11 (with 2 fused rings) derived from benzodiazepine (N2); Ci 3 (with 3 fused rings) derived from carbazole (Ni), dibenzofuran (Oi), dibenzothiophene (Si), carboline (N 2 ), perimidine (N 2 ), pyridoindole (N 2 ); and,

Ci 4 (with 3 fused rings) derived from acridine (N 1 ), xanthene (O 1 ), thioxanthene (Si), oxanthrene (O 2 ), phenoxathiin (O 1 S 1 ), phenazine (N 2 ), phenoxazine (N 1 O 1 ), phenothiazine (N 1 S 1 ), thianthrene (S 2 ), phenanthridine (N 1 ), phenanthroline (N 2 ), phenazine (N 2 ).

The above groups, whether alone or part of another substituent, may themselves optionally be substituted with one or more groups selected from themselves and the additional substituents listed below.

Halo: -F, -Cl, -Br, and -I.

Hydroxy: -OH.

Ether: -OR, wherein R is an ether substituent, for example, a C 1-7 alkyl group (also referred to as a Ci- 7 alkoxy group, discussed below), a C 3-20 heterocyclyl group (also referred to as a C 3-20 heterocyclyloxy group), or a Cs- 2 o aryl group (also referred to as a Cs- 2 o aryloxy group), preferably a C^alkyl group.

Alkoxy: -OR, wherein R is an alkyl group, for example, a C 1-7 alkyl group. Examples of C 1-7 alkoxy groups include, but are not limited to, -OMe (methoxy), -OEt (ethoxy), -O(nPr) (n- propoxy), -O(iPr) (isopropoxy), -O(nBu) (n-butoxy), -O(sBu) (sec-butoxy), -O(iBu)

(isobutoxy), and -O(tBu) (tert-butoxy).

Acetal: -CH(OR 1 )(OR 2 ), wherein R 1 and R 2 are independently acetal substituents, for example, a C 1-7 alkyl group, a C 3-20 heterocyclyl group, or a Cs- 2 o aryl group, preferably a C 1-7 alkyl group, or, in the case of a“cyclic” acetal group, R 1 and R 2 , taken together with the two oxygen atoms to which they are attached, and the carbon atoms to which they are attached, form a heterocyclic ring having from 4 to 8 ring atoms. Examples of acetal groups include, but are not limited to, -CH(OMe) 2 , -CH(OEt) 2 , and -CH(OMe)(OEt).

Hemiacetal: -CH(OH)(OR 1 ), wherein R 1 is a hemiacetal substituent, for example, a C 1-7 alkyl group, a C 3-20 heterocyclyl group, or a Cs- 2 o aryl group, preferably a C 1-7 alkyl group.

Examples of hemiacetal groups include, but are not limited to, -CH(OH)(OMe) and - CH(OH)(OEt). Ketal: -CR(OR 1 )(OR 2 ), where R 1 and R 2 are as defined for acetals, and R is a ketal substituent other than hydrogen, for example, a Cw alkyl group, a C 3-20 heterocyclyl group, or a C 5-2 o aryl group, preferably a C alkyl group. Examples ketal groups include, but are not limited to, -C(Me)(OMe) 2 , -C(Me)(OEt) 2 , -C(Me)(OMe)(OEt), -C(Et)(OMe) 2 , -C(Et)(OEt) 2 , and -C(Et)(OMe)(OEt).

Hemiketal: -CR(OH)(OR 1 ), where R 1 is as defined for hemiacetals, and R is a hemiketal substituent other than hydrogen, for example, a Cw alkyl group, a C 3-20 heterocyclyl group, or a C 5-2 o aryl group, preferably a Cw alkyl group. Examples of hemiacetal groups include, but are not limited to, -C(Me)(OH)(OMe), -C(Et)(OH)(OMe), -C(Me)(OH)(OEt), and

-C(Et)(OH)(OEt).

Oxo (keto, -one): =0.

Thione (thioketone): =S.

Imino (imine): =NR, wherein R is an imino substituent, for example, hydrogen, Cw alkyl group, a C 3-20 heterocyclyl group, or a Cs- 2 o aryl group, preferably hydrogen or a Cw alkyl group. Examples of ester groups include, but are not limited to, =NH, =NMe, =NEt, and =NPh.

Formyl (carbaldehyde, carboxaldehyde): -C(=0)H.

Acyl (keto): -C(=0)R, wherein R is an acyl substituent, for example, a Cw alkyl group (also referred to as Ci- 7 alkylacyl or Ci- 7 alkanoyl), a C 3-20 heterocyclyl group (also referred to as C 3-20 heterocyclylacyl), or a Cs- 2 o aryl group (also referred to as Cs- 2 o arylacyl), preferably a C 1-7 alkyl group. Examples of acyl groups include, but are not limited to, -C(=0)CH 3 (acetyl), -C(=0)CH 2 CH 3 (propionyl), -C(=0)C(CH 3 ) 3 (t-butyryl), and -C(=0)Ph (benzoyl, phenone).

Carboxy (carboxylic acid): -C(=0)0H.

Thiocarboxy (thiocarboxylic acid): -C(=S)SH.

Thiolocarboxy (thiolocarboxylic acid): -C(=0)SH.

Thionocarboxy (thionocarboxylic acid): -C(=S)OH. Imidic acid: -C(=NH)OH.

Hydroxamic acid: -C(=NOH)OH.

Ester (carboxylate, carboxylic acid ester, oxycarbonyl): -C(=0)0R, wherein R is an ester substituent, for example, a C alkyl group, a C3-2o heterocyclyl group, or a Cs-2o aryl group, preferably a C alkyl group. Examples of ester groups include, but are not limited to, -C(=0)0CH 3 , -C(=0)0CH 2 CH 3 , -C(=0)0C(CH 3 ) 3 , and -C(=0)0Ph.

Acyloxy (reverse ester): -0C(=0)R, wherein R is an acyloxy substituent, for example, a C alkyl group, a C 3- 2o heterocyclyl group, or a Cs-2o aryl group, preferably a C alkyl group. Examples of acyloxy groups include, but are not limited to, -0C(=0)CH 3 (acetoxy),

-0C(=0)CH 2 CH 3 , -0C(=0)C(CH 3 ) 3 , -0C(=0)Ph, and -0C(=0)CH 2 Ph.

Oxycarboyloxy: -0C(=0)0R, wherein R is an ester substituent, for example, a C1-7 alkyl group, a C 3-2 o heterocyclyl group, or a Cs-2o aryl group, preferably a C alkyl group.

Examples of ester groups include, but are not limited to, -0C(=0)0CH 3 , -0C(=0)0CH 2 CH 3 , -0C(=0)0C(CH 3 ) 3 , and -0C(=0)0Ph.

Amino: -NR 1 R 2 , wherein R 1 and R 2 are independently amino substituents, for example, hydrogen, a C alkyl group (also referred to as Ci-7 alkylamino or di-Ci-7 alkylamino), a C 3-2 o heterocyclyl group, or a Cs-2o aryl group, preferably H or a C alkyl group, or, in the case of a “cyclic” amino group, R 1 and R 2 , taken together with the nitrogen atom to which they are attached, form a heterocyclic ring having from 4 to 8 ring atoms. Amino groups may be primary (-NH 2 ), secondary (-NHR 1 ), or tertiary (-NHR 1 R 2 ), and in cationic form, may be quaternary (- + NR 1 R 2 R 3 ). Examples of amino groups include, but are not limited to, -NH 2 , -NHCH 3 , -NHC(CH 3 ) 2 , -N(CH 3 ) 2 , -N(CH 2 CH 3 ) 2 , and -NHPh. Examples of cyclic amino groups include, but are not limited to, aziridino, azetidino, pyrrolidino, piperidino, piperazino, morpholino, and thiomorpholino.

Amido (carbamoyl, carbamyl, aminocarbonyl, carboxamide): -C(=0)NR 1 R 2 , wherein R 1 and R 2 are independently amino substituents, as defined for amino groups. Examples of amido groups include, but are not limited to, -C(=0)NH 2 , -C(=0)NHCH 3 , -C(=0)N(CH 3 ) 2 ,

-C(=0)NHCH 2 CH 3 , and -C(=0)N(CH 2 CH 3 ) 2 , as well as amido groups in which R 1 and R 2 , together with the nitrogen atom to which they are attached, form a heterocyclic structure as in, for example, piperidinocarbonyl, morpholinocarbonyl, thiomorpholinocarbonyl, and piperazinocarbonyl.

Thioamido (thiocarbamyl): -C(=S)NR 1 R 2 , wherein R 1 and R 2 are independently amino substituents, as defined for amino groups. Examples of amido groups include, but are not limited to, -C(=S)NH 2 , -C(=S)NHCH 3 , -C(=S)N(CH 3 ) 2 , and -C(=S)NHCH 2 CH 3 .

Acylamido (acylamino): -NR 1 C(=0)R 2 , wherein R 1 is an amide substituent, for example, hydrogen, a C alkyl group, a C 3- 2o heterocyclyl group, or a Cs-2o aryl group, preferably hydrogen or a C alkyl group, and R 2 is an acyl substituent, for example, a C alkyl group, a C 3- 2o heterocyclyl group, or a Cs-2oaryl group, preferably hydrogen or a C alkyl group. Examples of acylamide groups include, but are not limited to, -NHC(=0)CH 3 ,

-NHC(=0)CH 2 CH 3 , and -NHC(=0)Ph. R 1 and R 2 may together form a cyclic structure, as in, for example, succinimidyl, maleimidyl, and phthalimidyl:

succinimidyl maleimidyl phthalimidyl

Aminocarbonyloxy: -0C(=0)NR 1 R 2 , wherein R 1 and R 2 are independently amino

substituents, as defined for amino groups. Examples of aminocarbonyloxy groups include, but are not limited to, -0C(=0)NH 2 , -0C(=0)NHMe, -0C(=0)NMe 2 , and -0C(=0)NEt 2 .

Ureido: -N(R 1 )CONR 2 R 3 wherein R 2 and R 3 are independently amino substituents, as defined for amino groups, and R 1 is a ureido substituent, for example, hydrogen, a C alkyl group, a C 3- 2o heterocyclyl group, or a Cs-2o aryl group, preferably hydrogen or a C alkyl group. Examples of ureido groups include, but are not limited to, -NHCONH2, -NHCONHMe, -NHCONHEt, -NHCONMe 2 , -NHCONEt 2 , -NMeCONhh, -NMeCONHMe, -NMeCONHEt, - NMeCONMe 2 , and -NMeCONEt 2 .

Guanidino: -NH-C(=NH)NH 2 .

Tetrazolyl: a five membered aromatic ring having four nitrogen atoms and one carbon atom,

Imino: =NR, wherein R is an imino substituent, for example, for example, hydrogen, a C alkyl group, a C 3-20 heterocyclyl group, or a Cs- 2 o aryl group, preferably H or a Ci- 7 alkyl group. Examples of imino groups include, but are not limited to, =NH, =NMe, and =NEt.

Amidine (amidino): -C(=NR)NR 2 , wherein each R is an amidine substituent, for example, hydrogen, a C 1-7 alkyl group, a C 3-20 heterocyclyl group, or a Cs- 2 o aryl group, preferably H or a C 1-7 alkyl group. Examples of amidine groups include, but are not limited to, -C(=NH)NH 2 , -C(=NH)NMe 2 , and -C(=NMe)NMe 2 .

Nitro: -NO2.

Nitroso: -NO.

Azido: -N3.

Cyano (nitrile, carbonitrile): -CN.

Isocyano: -NC.

Cyanato: -OCN.

Isocyanato: -NCO.

Thiocyano (thiocyanato): -SCN.

Isothiocyano (isothiocyanato): -NCS.

Sulfhydryl (thiol, mercapto): -SH.

Thioether (sulfide): -SR, wherein R is a thioether substituent, for example, a C 1-7 alkyl group (also referred to as a C alkylthio group), a C 3-20 heterocyclyl group, or a Cs- 2 o aryl group, preferably a C1-7 alkyl group. Examples of C1-7 alkylthio groups include, but are not limited to, -SCHs and -SCH2CH3.

Disulfide: -SS-R, wherein R is a disulfide substituent, for example, a C alkyl group, a C3-20 heterocyclyl group, or a Cs-2o aryl group, preferably a C alkyl group (also referred to herein as C1-7 alkyl disulfide). Examples of C alkyl disulfide groups include, but are not limited to, -SSCHs and -SSCH2CH3.

Sulfine (sulfinyl, sulfoxide): -S(=0)R, wherein R is a sulfine substituent, for example, a C1-7 alkyl group, a C3-20 heterocyclyl group, or a Cs-2o aryl group, preferably a C1-7 alkyl group. Examples of sulfine groups include, but are not limited to, -S(=0)CH 3 and -S(=0)CH 2 CH 3 .

Sulfone (sulfonyl): -S(=0) 2 R, wherein R is a sulfone substituent, for example, a C1-7 alkyl group, a C3-20 heterocyclyl group, or a Cs-2o aryl group, preferably a C alkyl group, including, for example, a fluorinated or perfluorinated C1-7 alkyl group. Examples of sulfone groups include, but are not limited to, -S(=0) 2 CH 3 (methanesulfonyl, mesyl), -S(=0)2CF3 (triflyl), -S(=0) 2 CH 2 CH 3 (esyl), -S(=0) 2 C 4 F 9 (nonaflyl), -S(=0) 2 CH 2 CF 3 (tresyl), -S(=0)2CH 2 CH 2 NH2 (tauryl), -S(=0) 2 Ph (phenylsulfonyl, besyl), 4-methylphenylsulfonyl (tosyl),

4-chlorophenylsulfonyl (closyl), 4-bromophenylsulfonyl (brosyl), 4-nitrophenyl (nosyl), 2-naphthalenesulfonate (napsyl), and 5-dimethylamino-naphthalen-1 -ylsulfonate (dansyl).

Sulfinic acid (sulfino): -S(=0)0H, -SO2H.

Sulfonic acid (sulfo): -S(=0) 2 0H, -SO3H .

Sulfinate (sulfinic acid ester): -S(=0)0R; wherein R is a sulfinate substituent, for example, a C1-7 alkyl group, a C3-20 heterocyclyl group, or a Cs-2o aryl group, preferably a C1-7 alkyl group. Examples of sulfinate groups include, but are not limited to, -S(=0)0CH 3 (methoxysulfinyl; methyl sulfinate) and -S(=0)0CH 2 CH 3 (ethoxysulfinyl; ethyl sulfinate).

Sulfonate (sulfonic acid ester): -S(=0) 2 0R, wherein R is a sulfonate substituent, for example, a C1-7 alkyl group, a C3-20 heterocyclyl group, or a Cs-2o aryl group, preferably a C1-7 alkyl group. Examples of sulfonate groups include, but are not limited to, -S(=0) 2 0CH 3

(methoxysulfonyl; methyl sulfonate) and -S(=0) 2 0CH 2 CH 3 (ethoxysulfonyl; ethyl sulfonate). Sulfinyloxy: -0S(=0)R, wherein R is a sulfinyloxy substituent, for example, a C alkyl group, a C3-20 heterocyclyl group, or a Cs-2o aryl group, preferably a C alkyl group. Examples of sulfinyloxy groups include, but are not limited to, -OS(=0)CH3 and -0S(=0)CH2CH3.

Sulfonyloxy: -OS(=0) 2 R, wherein R is a sulfonyloxy substituent, for example, a C alkyl group, a C3-20 heterocyclyl group, or a Cs-2o aryl group, preferably a C alkyl group.

Examples of sulfonyloxy groups include, but are not limited to, -0S(=0) 2 CH 3 (mesylate) and -0S(=0) 2 CH 2 CH 3 (esylate).

Sulfate: -0S(=0) 2 0R; wherein R is a sulfate substituent, for example, a C alkyl group, a C3-20 heterocyclyl group, or a Cs-2o aryl group, preferably a C alkyl group. Examples of sulfate groups include, but are not limited to, -0S(=0)20CH3 and -S0(=0)20CH2CH3.

Sulfamyl (sulfamoyl; sulfinic acid amide; sulfinamide): -S(=0)NR 1 R 2 , wherein R 1 and R 2 are independently amino substituents, as defined for amino groups. Examples of sulfamyl groups include, but are not limited to, -S(=0)NH2, -S(=0)NH(CH3), -S(=0)N(CH3)2,

-S(=0)NH(CH 2 CH 3 ), -S(=0)N(CH 2 CH 3 ) 2 , and -S(=0)NHPh.

Sulfonamido (sulfinamoyl; sulfonic acid amide; sulfonamide): -S(=0) 2 NR 1 R 2 , wherein R 1 and R 2 are independently amino substituents, as defined for amino groups. Examples of sulfonamido groups include, but are not limited to, -S(=0)2NH2, -S(=0)2NH(CH3),

-S(=0) 2 N(CH 3 ) 2 , -S(=0) 2 NH(CH 2 CH 3 ), -S(=0) 2 N(CH 2 CH 3 )2, and -S(=0) 2 NHPh.

Sulfamino: -NR 1 S(=0) 2 0H, wherein R 1 is an amino substituent, as defined for amino groups. Examples of sulfamino groups include, but are not limited to, -NHS(=0) 2 0H and

-N(CH 3 )S(=0) 2 0H.

Sulfonamino: -NR 1 S(=0) 2 R, wherein R 1 is an amino substituent, as defined for amino groups, and R is a sulfonamino substituent, for example, a C alkyl group, a C3-20 heterocyclyl group, or a Cs-2o aryl group, preferably a C alkyl group. Examples of sulfonamino groups include, but are not limited to, -NHS(=0)2CH3 and -N(CH3)S(=0)2C6H 5 .

Sulfinamino: -NR 1 S(=0)R, wherein R 1 is an amino substituent, as defined for amino groups, and R is a sulfinamino substituent, for example, a C alkyl group, a C3-20 heterocyclyl group, or a C5-2o aryl group, preferably a C alkyl group. Examples of sulfinamino groups include, but are not limited to, -NHS(=0)CH3 and -N(CH3)S(=0)C6H 5 . Phosphino (phosphine): -PR2, wherein R is a phosphino substituent, for example, -H, a C1-7 alkyl group, a C3-20 heterocyclyl group, or a Cs-2o aryl group, preferably -H, a C alkyl group, or a C5-2o aryl group. Examples of phosphino groups include, but are not limited to, -PH2, -P(CH 3 ) 2 , -P(CH 2 CH 3 )2, -P(t-Bu) 2 , and -P(Ph) 2 .

Phospho: -P(=0)2.

Phosphinyl (phosphine oxide): -P(=0)R 2 , wherein R is a phosphinyl substituent, for example, a C1-7 alkyl group, a C 3- 2o heterocyclyl group, or a Cs-2o aryl group, preferably a C1-7 alkyl group or a Cs-2o aryl group. Examples of phosphinyl groups include, but are not limited to, -P(=0)(CH 3 ) 2 , -P(=0)(CH 2 CH 3 ) 2 , -P(=0)(t-Bu) 2 , and -P(=0)(Ph) 2 .

Phosphonic acid (phosphono): -P(=0)(OH) 2 .

Phosphonate (phosphono ester): -P(=0)(OR) 2 , where R is a phosphonate substituent, for example, -H, a C1-7 alkyl group, a C 3- 2o heterocyclyl group, or a Cs-2o aryl group, preferably -H, a C1-7 alkyl group, or a Cs-2o aryl group. Examples of phosphonate groups include, but are not limited to, -P(=0)(0CH 3 ) 2 , -P(=0)(0CH 2 CH 3 ) 2 , -P(=0)(0-t-Bu) 2 , and -P(=0)(OPh) 2 .

Phosphoric acid (phosphonooxy): -OP(=0)(OH) 2 .

Phosphate (phosphonooxy ester): -OP(=0)(OR) 2 , where R is a phosphate substituent, for example, -H, a C1-7 alkyl group, a C 3- 2o heterocyclyl group, or a Cs-2o aryl group, preferably -H, a C1-7 alkyl group, or a Cs-2o aryl group. Examples of phosphate groups include, but are not limited to, -0P(=0)(0CH 3 ) 2 , -0P(=0)(0CH 2 CH 3 ) 2 , -0P(=0)(0-t-Bu) 2 , and -OP(=0)(OPh) 2 .

Phosphorous acid: -OP(OH)2.

Phosphite: -OP(OR)2, where R is a phosphite substituent, for example, -H, a C1-7 alkyl group, a C 3- 2o heterocyclyl group, or a Cs-2o aryl group, preferably -H, a C1-7 alkyl group, or a Cs-2o aryl group. Examples of phosphite groups include, but are not limited to, -OP(OCH 3 )2,

-OP(OCH 2 CH 3 ) 2 , -OP(0-t-Bu) 2 , and -OP(OPh) 2 .

Phosphoramidite: -OP(OR 1 )-NR 2 2, where R 1 and R 2 are phosphoramidite substituents, for example, -H, a (optionally substituted) C1-7 alkyl group, a C 3- 2o heterocyclyl group, or a C5-20 aryl group, preferably -H, a C 1-7 alkyl group, or a Cs- 2 o aryl group. Examples of phosphoramidite groups include, but are not limited to, -OP(OCH2CH3)-N(CH3)2,

-OP(OCH 2 CH3)-N(i-Pr)2, and -OP(OCH 2 CH 2 CN)-N(i-Pr)2.

Phosphoramidate: -0P(=0)(0R 1 )-NR 2 2 , where R 1 and R 2 are phosphoramidate substituents, for example, -H, a (optionally substituted) C 1-7 alkyl group, a C 3-20 heterocyclyl group, or a C 5-2 o aryl group, preferably -H, a C 1-7 alkyl group, or a Cs- 2 o aryl group. Examples of phosphoramidate groups include, but are not limited to, -0P(=0)(0CH2CH3)-N(CH3)2, -0P(=0)(0CH 2 CH 3 )-N(i-Pr)2, and -0P(=0)(0CH 2 CH 2 CN)-N(i-Pr)2.

Alkylene

C 3-12 alkylene: The term“C 3-12 alkylene”, as used herein, pertains to a bidentate moiety obtained by removing two hydrogen atoms, either both from the same carbon atom, or one from each of two different carbon atoms, of a hydrocarbon compound having from 3 to 12 carbon atoms (unless otherwise specified), which may be aliphatic or alicyclic, and which may be saturated, partially unsaturated, or fully unsaturated. Thus, the term“alkylene” includes the sub-classes alkenylene, alkynylene, cycloalkylene, etc., discussed below.

Examples of linear saturated C 3-12 alkylene groups include, but are not limited to, -(CH 2 ) n - where n is an integer from 3 to 12, for example, -CH 2 CH 2 CH 2 - (propylene),

-CH2CH2CH2CH2- (butylene), -CH2CH2CH2CH2CH2- (pentylene) and

-CH2CH2CH2CH-2CH2CH2CH2- (heptylene).

Examples of branched saturated C 3-12 alkylene groups include, but are not limited to, -CH(CH 3 )CH 2 -, -CH(CH 3 )CH 2 CH 2 -, -CH(CH3)CH 2 CH 2 CH2-, -CH 2 CH(CH 3 )CH 2 -,

-CH 2 CH(CH3)CH 2 CH2-, -CH(CH 2 CH 3 )-, -CH(CH 2 CH 3 )CH 2 -, and -CH 2 CH(CH 2 CH3)CH2-.

Examples of linear partially unsaturated C 3 -i 2 alkylene groups (C 3-12 alkenylene, and alkynylene groups) include, but are not limited to, -CH=CH-CH2-, -CH2-CH=CH2-,

-CH=CH-CH 2 -CH 2 -, -CH=CH-CH 2 -CH 2 -CH 2 -, -CH=CH-CH=CH-, -CH=CH-CH=CH-CH 2 -, - CH=CH-CH=CH-CH 2 -CH 2 -, -CH=CH-CH 2 -CH=CH-, -CH=CH-CH 2 -CH 2 -CH=CH-, and -CH 2 - CºC-CH 2 -.

Examples of branched partially unsaturated C 3 -i 2 alkylene groups (C 3-12 alkenylene and alkynylene groups) include, but are not limited to, -C(CH 3 )=CH-, -C(CH 3 )=CH-CH 2 -,

-CH=CH-CH(CH 3 )- and -CºC-CH(CH 3 )-. Examples of alicyclic saturated C3-i2 alkylene groups (C3-12 cycloalkylenes) include, but are not limited to, cyclopentylene (e.g. cyclopent-1 ,3-ylene), and cyclohexylene

(e.g. cyclohex-1 ,4-ylene).

Examples of alicyclic partially unsaturated C3-i2 alkylene groups (C3-12 cycloalkylenes) include, but are not limited to, cyclopentenylene (e.g. 4-cyclopenten-1 ,3-ylene),

cyclohexenylene (e.g. 2-cyclohexen-1 ,4-ylene; 3-cyclohexen-1 ,2-ylene; 2,5-cyclohexadien- 1 ,4-ylene).

Bis-oxy-Ci-3 alkylene: The term“Bis-oxy-C- 1-3 alkylene”, as used herein, pertains to a bidentate moiety of formula -0-(CH 2 ) q -0-, where q is from 1 to 3, i.e. -OCH2CH2CH2O-, - OCH2CH2O-, -OCH2O-.

Carbamate nitrogen protecting group: the term“carbamate nitrogen protecting group” pertains to a moiety which masks the nitrogen in the imine bond, and these are well known in the art. These groups have the following structure:

wherein R’ 10 is R as defined above. A large number of suitable groups are described on pages 503 to 549 of Greene, T.W. and Wuts, G.M., Protective Groups in Organic Synthesis, 3 rd Edition, John Wiley & Sons, Inc., 1999, which is incorporated herein by reference.

Hemi-aminal nitrogen protecting group: the term“hemi-aminal nitrogen protecting group” pertains to a group having the following structure:

wherein R’ 10 is R as defined above. A large number of suitable groups are described on pages 633 to 647 as amide protecting groups of Greene, T.W. and Wuts, G.M., Protective Groups in Organic Synthesis, 3 rd Edition, John Wiley & Sons, Inc., 1999, which is incorporated herein by reference.

The groups Carbamate nitrogen protecting group and Hemi-aminal nitrogen protecting group may be jointly termed a“nitrogen protecting group for synthesis”. Includes 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 + HR 1 R 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.

Salts

It may be convenient or desirable to prepare, purify, and/or handle a corresponding salt of the active compound, for example, a pharmaceutically-acceptable salt. Examples of pharmaceutically acceptable salts are discussed in Berge, et al., J. Pharm. Sci., 66, 1 -19 (1977).

For example, if the compound is anionic, or has a functional group which may be anionic (e.g. -COOH may be -COO ), then a salt may be formed with a suitable cation. Examples of suitable inorganic cations include, but are not limited to, alkali metal ions such as Na + and K + , alkaline earth cations such as Ca 2+ and Mg 2+ , and other cations such as AG 3 . Examples of suitable organic cations include, but are not limited to, ammonium ion (i.e. NH 4 + ) and substituted ammonium ions (e.g. NH 3 R + , NH2R2 + , NHR3 + , NR 4 + ). 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 + .

If the compound is cationic, or has a functional group which may be cationic (e.g. -NFh may be -NH 3 + ), then a 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, hydrobromic, hydroiodic, sulfuric, sulfurous, nitric, nitrous, phosphoric, and phosphorous. 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, triflu oroacetic acid 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.

Solvates

It may be convenient or desirable to prepare, purify, and/or handle a corresponding solvate of the active compound. The term“solvate” is used herein in the conventional sense to refer to a complex of solute (e.g. active compound, salt of active compound) and solvent. If the solvent is water, the solvate may be conveniently referred to as a hydrate, for example, a mono-hydrate, a di-hydrate, a tri-hydrate, etc.

The invention includes compounds where a solvent adds across the imine bond of the PBD moiety, which is illustrated below where the solvent is water or an alcohol (R A OH, where R A is C1-4 alkyl):

These forms can be called the carbinolamine and carbinolamine ether forms of the PBD (as described in the section relating to R 10 above). The balance of these equilibria depends on the conditions in which the compounds are found, as well as the nature of the moiety itself.

These particular compounds may be isolated in solid form, for example, by lyophilisation.

Isomers

Certain compounds of the invention 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 l-forms; (+) and (-) forms; keto-, enol-, and enolate-forms; syn- and anti-forms; synclinal- and anticlinal-forms; a- and b-forms; axial and equatorial forms; boat-, chair-, twist-, envelope-, and halfchair-forms; and combinations thereof, hereinafter collectively referred to as “isomers” (or“isomeric forms”).

The term“chiral” refers to molecules which have the property of non-superimposability of the mirror image partner, while the term“achiral” refers to molecules which are superimposable on their mirror image partner.

The term“stereoisomers” refers to compounds which have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space.

“Diastereomer” refers to a stereoisomer with two or more centers of chirality and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g. melting points, boiling points, spectral properties, and reactivities. Mixtures of diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography.

“Enantiomers” refer to two stereoisomers of a compound which are non-superimposable mirror images of one another.

Stereochemical definitions and conventions used herein generally follow S. P. Parker, Ed., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; and Eliel, E. and Wilen, S.,“Stereochemistry of Organic Compounds”, John Wiley & Sons, Inc., New York, 1994. The compounds of the invention may contain asymmetric or chiral centers, and therefore exist in different stereoisomeric forms. It is intended that all stereoisomeric forms of the compounds of the invention, including but not limited to, diastereomers, enantiomers and atropisomers, as well as mixtures thereof such as racemic mixtures, form part of the present invention. Many organic compounds exist in optically active forms, i.e., they have the ability to rotate the plane of plane-polarized light. In describing an optically active compound, the prefixes D and L, or R and S, are used to denote the absolute configuration of the molecule about its chiral center(s). The prefixes d and I or (+) and (-) are employed to designate the sign of rotation of plane-polarized light by the compound, with (-) or I meaning that the compound is levorotatory. A compound prefixed with (+) or d is dextrorotatory. For a given chemical structure, these stereoisomers are identical except that they are mirror images of one another. A specific stereoisomer may also be referred to as an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture. A 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which may occur where there has been no stereoselection or stereospecificity in a chemical reaction or process. The terms“racemic mixture” and“racemate” refer to an equimolar mixture of two enantiomeric species, devoid of optical activity.

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, -CH2OH. Similarly, a reference to ortho-chlorophenyl is not to be construed as a reference to its structural isomer, meta- chlorophenyl. However, a reference to a class of structures may well include structurally isomeric forms falling within that class (e.g. C1- 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 pertain to tautomeric forms, for example, keto-, enol-, and enolate-forms, as in, for example, the following tautomeric pairs: keto/enol (illustrated below), imine/enamine, amide/imino alcohol, amidine/amidine, nitroso/oxime,

thioketone/enethiol, N-nitroso/hyroxyazo, and nitro/aci-nitro.

keto enol enolate

The term“tautomer” or“tautomeric form” refers to structural isomers of different energies which are interconvertible via a low energy barrier. For example, proton tautomers (also known as prototropic tautomers) include interconversions via migration of a proton, such as keto-enol and imine-enamine isomerizations. Valence tautomers include interconversions by reorganization of some of the bonding electrons.

Note that specifically included in the term“isomer” are compounds with one or more isotopic substitutions. For example, H may be in any isotopic form, including 1 H, 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 16 0 and 18 0; and the like. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, and chlorine, such as, but not limited to 2 H (deuterium, D), 3 H (tritium), 11 C, 13 C, 14 C, 15 N, 18 F, 31 P, 32 P, 35 S, 36 CI, and 125 l. Various isotopically labeled compounds of the present invention, for example those into which radioactive isotopes such as 3H, 13C, and 14C are incorporated. Such isotopically labelled compounds may be useful in metabolic studies, reaction kinetic studies, detection or imaging techniques, such as positron emission tomography (PET) or single- photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays, or in radioactive treatment of patients. Deuterium labelled or substituted therapeutic compounds of the invention may have improved DMPK (drug metabolism and pharmacokinetics) properties, relating to distribution, metabolism, and excretion (ADME). Substitution with heavier isotopes such as deuterium may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements. An 18F labeled compound may be useful for PET or SPECT studies. Isotopically labeled compounds of this invention and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations described below by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent. Further, substitution with heavier isotopes, particularly deuterium (i.e., 2H or D) may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements or an improvement in therapeutic index. It is understood that deuterium in this context is regarded as a substituent. The concentration of such a heavier isotope, specifically deuterium, may be defined by an isotopic enrichment factor. In the compounds of this invention any atom not specifically designated as a particular isotope is meant to represent any stable isotope of that atom.

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. Ligand Unit

The Ligand Unit may be of any kind, and include a protein, polypeptide, peptide and a non- peptidic agent that specifically binds to a target molecule. In some embodiments, the Ligand unit may be a protein, polypeptide or peptide. In some embodiments, the Ligand unit may be a cyclic polypeptide. These Ligand units can include antibodies or a fragment of an antibody that contains at least one target molecule-binding site, lymphokines, hormones, growth factors, or any other cell binding molecule or substance that can specifically bind to a target.

The terms“specifically binds” and“specific binding” refer to the binding of an antibody or other protein, polypeptide or peptide to a predetermined molecule (e.g., an antigen).

Typically, the antibody or other molecule binds with an affinity of at least about 1x10 7 M 1 , and binds to the predetermined molecule with an affinity that is at least two-fold greater than its affinity for binding to a non-specific molecule (e.g., BSA, casein) other than the predetermined molecule or a closely-related molecule.

Examples of Ligand units include those agents described for use in WO 2007/085930, which is incorporated herein.

In some embodiments, the Ligand unit is a Cell Binding Agent that binds to an extracellular target on a cell. Such a Cell Binding Agent can be a protein, polypeptide, peptide or a non- peptidic agent. In some embodiments, the Cell Binding Agent may be a protein, polypeptide or peptide. In some embodiments, the Cell Binding Agent may be a cyclic polypeptide. The Cell Binding Agent also may be antibody or an antigen-binding fragment of an antibody. Thus, in one embodiment, the present invention provides an antibody-drug conjugate (ADC).

Cell Binding Agent

A cell binding agent may be of any kind, and include peptides and non-peptides. These can include antibodies or a fragment of an antibody that contains at least one binding site, lymphokines, hormones, hormone mimetics, vitamins, growth factors, nutrient-transport molecules, or any other cell binding molecule or substance.

Peptides

In one embodiment, the cell binding agent is a linear or cyclic peptide comprising 4-30, preferably 6-20, contiguous amino acid residues. In this embodiment, it is preferred that one cell binding agent is linked to one monomer or dimer pyrrolobenzodiazepine compound. In one embodiment the cell binding agent comprises a peptide that binds integrin a n b 6 . The peptide may be selective for a n b q over XYS.

In one embodiment the cell binding agent comprises the A20FMDV-Cys polypeptide. The A20FMDV-Cys has the sequence: NAVPNLRGDLQVLAQKVARTC. Alternatively, a variant of the A20FMDV-Cys sequence may be used wherein one, two, three, four, five, six, seven, eight, nine or ten amino acid residues are substituted with another amino acid residue.

Furthermore, the polypeptide may have the sequence NAVXXXXXXXXXXXXXXXRTC.

Antibodies

The term“antibody” herein is used in the broadest sense and specifically covers monoclonal antibodies, polyclonal antibodies, dimers, multimers, multispecific antibodies (e.g., bispecific antibodies), multivalent antibodies and antibody fragments, so long as they exhibit the desired biological activity (Miller et al (2003) Jour of Immunology 170:4854-4861 ).

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 multiple antibodies. 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. lgG1 , lgG2, lgG3, lgG4, lgA1 and lgA2) or subclass of immunoglobulin molecule. The immunoglobulins can be derived from any species, including human, murine, or rabbit origin.

"Antibody fragments" comprise a portion of a full-length antibody, generally 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-ld) antibodies, CDR (complementary determining region), and epitope-binding fragments of any of the above which immunospecifically bind to cancer cell antigens, viral antigens or microbial antigens, single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.

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 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 monoclonal antibodies herein specifically include chimeric antibodies, humanized antibodies and human antibodies.

Examples of cell binding agents include those agents described for use in WO 2007/085930, which is incorporated herein.

Tumour-associate antigens and cognate antibodies for use in embodiments of the present invention are listed below, and are described in more detail on pages 14 to 86 of WO

2017/186894, which is incorporated herein.

(1) BMPR1 B (bone morphogenetic protein receptor-type IB)

(2) E16 (LAT 1 , SLC7A5)

(3) STEAP1 (six transmembrane epithelial antigen of prostate)

(4) 0772P (CA125, MUC16) (5) MPF (MPF, MSLN, SMR, megakaryocyte potentiating factor, mesothelin)

(6) Napi3b (NAPI-3B, NPTIIb, SLC34A2, solute carrier family 34 (sodium phosphate), member 2, type II sodium-dependent phosphate transporter 3b)

(7) Serna 5b (FLJ10372, KIAA1445, Mm.42015, SEMA5B, SEMAG, Semaphorin 5b Hlog,

25 sema domain, seven thrombospondin repeats (type 1 and type 1-like), transmembrane domain (TM) and short cytoplasmic domain, (semaphorin) 5B)

(8) PSCA hlg (2700050C12Rik, C530008016Rik, RIKEN cDNA 2700050C12, RIKEN cDNA 2700050C12 gene)

(9) ETBR (Endothelin type B receptor)

(10) MSG783 (RNF124, hypothetical protein FLJ20315)

(11) STEAP2 (HGNC_8639, IPCA-1 , PCANAP1 , STAMP1 , STEAP2, STMP, prostate cancer associated gene 1 , prostate cancer associated protein 1 , six transmembrane epithelial antigen of prostate 2, six transmembrane prostate protein)

(12) TrpM4 (BR22450, FLJ20041 , TRPM4, TRPM4B, transient receptor potential cation 5 channel, subfamily M, member 4)

(13) CRIPTO (CR, CR1 , CRGF, CRIPTO, TDGF1 , teratocarcinoma-derived growth factor)

(14) CD21 (CR2 (Complement receptor 2) or C3DR (C3d/Epstein Barr virus receptor) or Hs.73792)

(15) CD79b (CD79B, CD793, IGb (immunoglobulin-associated beta), B29)

(16) FcRH2 (IFGP4, IRTA4, SPAP1A (SH2 domain containing phosphatase anchor protein 1 a), SPAP1 B, SPAP1 C)

(17) HER2 (ErbB2)

(18) NCA (CEACAM6)

(19) MDP (DPEP1 )

(20) IL20R-alpha (IL20Ra, ZCYTOR7)

(21) Brevican (BCAN, BEHAB)

(22) EphB2R (DRT, ERK, Hek5, EPHT3, Tyro5)

(23) ASLG659 (B7h)

(24) PSCA (Prostate stem cell antigen precursor)

(25) GEDA

(26) BAFF-R (B cell -activating factor receptor, BLyS receptor 3, BR3)

(27) CD22 (B-cell receptor CD22-B isoform, BL-CAM, Lyb-8, Lyb8, SIGLEC-2, FLJ22814) (27a) CD22 (CD22 molecule)

(28) CD79a (CD79A, CD79alpha), 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: 19q 13.2).

(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 10 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: 11 q23.3,

(30) HLA-DOB (Beta subunit of MHC class II molecule (la antigen) that binds peptides and 20 presents them to CD4+ T lymphocytes); 273 aa, pi: 6.56, MW: 30820. TM: 1 [P] Gene Chromosome: 6p21.3)

(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).

(32) CD72 (B-cell differentiation antigen CD72, Lyb-2); 359 aa, pi: 8.66, MW: 40225, TM: 1 5 [P] Gene Chromosome: 9p13.3).

(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: 5q12).

(34) FcRH1 (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

20 differentiation); 429 aa, pi: 5.28, MW: 46925 TM: 1 [P] Gene Chromosome: 1 q21-1q22)

(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: 1q21 )

(36) TENB2 (TMEFF2, tomoregulin, TPEF, HPP1 , TR, putative transmembrane

35 proteoglycan, related to the EGF/heregulin family of growth factors and follistatin); 374 aa)

(37) PSMA - FOLH1 (Folate hydrolase (prostate-specific membrane antigen) 1 )

(38) SST (Somatostatin Receptor; note that there are5 subtypes)

(38.1 ) SSTR2 (Somatostatin receptor 2)

(38.2) SSTR5 (Somatostatin receptor 5)

(38.3) SSTR1

(38.4) SSTR3

(38.5) SSTR4 AvB6 - Both subunits (39+40)

(39) ITGAV (Integrin, alpha V)

(40) ITGB6 (Integrin, beta 6)

(41) CEACAM5 (Carcinoembryonic antigen-related cell adhesion molecule 5)

(42) MET (met proto-oncogene; hepatocyte growth factor receptor)

(43) MUC1 (Mucin 1 , cell surface associated)

(44) CA9 (Carbonic anhydrase IX)

(45) EGFRvlll (Epidermal growth factor receptor (EGFR), transcript variant 3,

(46) CD33 (CD33 molecule)

(47) CD19 (CD19 molecule)

(48) IL2RA (Interleukin 2 receptor, alpha); NCBI Reference Sequence: NM_000417.2);

(49) AXL (AXL receptor tyrosine kinase)

(50) CD30 - TNFRSF8 (Tumor necrosis factor receptor superfamily, member 8)

(51) BCMA (B-cell maturation antigen) - TNFRSF17 (Tumor necrosis factor receptor superfamily, member 17)

(52) CT Ags - CTA (Cancer Testis Antigens)

(53) CD174 (Lewis Y) - FUT3 (fucosyltransferase 3 (galactoside 3(4)-L-fucosyltransferase, Lewis blood group)

(54) CLEC14A (C-type lectin domain family 14, member A; Genbank accession no. NM175060)

(55) GRP78 - HSPA5 (heat shock 70kDa protein 5 (glucose-regulated protein, 78kDa)

(56) CD70 (CD70 molecule) L08096

(57) Stem Cell specific antigens. For example:

• 5T4 (see entry (63) below)

• CD25 (see entry (48) above)

• CD32

• LGR5/GPR49

• Prominin/CD133

(58) ASG-5

(59) ENPP3 (Ectonucleotide pyrophosphatase/phosphodiesterase 3)

(60) PRR4 (Proline rich 4 (lacrimal))

(61) GCC - GUCY2C (guanylate cyclase 2C (heat stable enterotoxin receptor)

(62) Liv-1 - SLC39A6 (Solute carrier family 39 (zinc transporter), member 6)

(63) 5T4, Trophoblast glycoprotein, TPBG - TPBG (trophoblast glycoprotein)

(64) CD56 - NCMA1 (Neural cell adhesion molecule 1 ) (65) CanAg (Tumor associated antigen CA242)

(66) FOLR1 (Folate Receptor 1 )

(67) GPNMB (Glycoprotein (transmembrane) nmb)

(68) TIM-1 - HAVCR1 (Hepatitis A virus cellular receptor 1 )

(69) RG-1/Prostate tumor target Mindin - Mindin/RG-1

(70) B7-H4 - VTCN1 (V-set domain containing T cell activation inhibitor 1

(71) PTK7 (PTK7 protein tyrosine kinase 7)

(72) CD37 (CD37 molecule)

(73) CD138 - SDC1 (syndecan 1 )

(74) CD74 (CD74 molecule, major histocompatibility complex, class II invariant chain)

(75) Claudins - CLs (Claudins)

(76) EGFR (Epidermal growth factor receptor)

(77) Her3 (ErbB3) - ERBB3 (v-erb-b2 erythroblastic leukemia viral oncogene homolog 3 (avian))

(78) RON - MST1 R (macrophage stimulating 1 receptor (c-met-related tyrosine kinase))

(79) EPHA2 (EPH receptor A2)

(80) CD20 - MS4A1 (membrane-spanning 4-domains, subfamily A, member 1 )

(81) Tenascin C - TNC (Tenascin C)

(82) FAP (Fibroblast activation protein, alpha)

(83) DKK-1 (Dickkopf 1 homolog (Xenopus laevis)

(84) CD52 (CD52 molecule)

(85) CS1 - SLAMF7 (SLAM family member 7)

(86) Endoglin - ENG (Endoglin)

(87) Annexin A1 - ANXA1 (Annexin A1 )

(88) V-CAM (CD106) - VCAM1 (Vascular cell adhesion molecule 1 )

The cell binding agent may be labelled, for example to aid detection or purification of the agent either prior to incorporation as a conjugate, or as part of the conjugate. The label may be a biotin label. In another embodiment, the cell binding agent may be labelled with a radioisotope.

Connection of Linker unit to Ligand unit

The Ligand unit may be connected to the Linker unit through a disulfide bond. In one embodiment, the connection between the Ligand unit and the Drug Linker is formed between a thiol group of a cysteine residue of the Ligand unit and a maleimide group of the Drug Linker unit.

The cysteine residues of the Ligand unit may be available for reaction with the functional group of the Linker unit to form a connection. In other embodiments, for example where the Ligand unit is an antibody, the thiol groups of the antibody may participate in interchain disulfide bonds. These interchain bonds may be converted to free thiol groups by e.g.

treatment of the antibody with DTT prior to reaction with the functional group of the Linker unit.

In some embodiments, the cysteine residue is an introduced into the heavy or light chain of an antibody. Positions for cysteine insertion by substitution in antibody heavy or light chains include those described in Published U.S. Application No. 2007-0092940 and International Patent Publication W02008070593, which are incorporated herein.

Methods of Treatment

The compounds of the present invention may be used in a method of therapy. Also provided is a method of treatment, comprising administering to a subject in need of treatment a therapeutically-effective amount of a conjugate of formula II. The term“therapeutically effective amount” is an amount sufficient to show benefit to a patient. Such benefit may be at least amelioration of at least one symptom. The actual amount administered, and rate and time-course of administration, will depend on the nature and severity of what is being treated. Prescription of treatment, e.g. decisions on dosage, is within the responsibility of general practitioners and other medical doctors.

A conjugate may be administered alone or in combination with other treatments, either simultaneously or sequentially dependent upon the condition to be treated. Examples of treatments and therapies include, but are not limited to, chemotherapy (the administration of active agents, including, e.g. drugs; surgery; and radiation therapy.

Pharmaceutical compositions according to the present invention, and for use in accordance with the present invention, may comprise, in addition to the active ingredient, i.e. a conjugate of formula I, a pharmaceutically acceptable excipient, carrier, buffer, stabiliser or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient. The precise nature of the carrier or other material will depend on the route of administration, which may be oral, or by injection, e.g. cutaneous, subcutaneous, or intravenous.

Pharmaceutical compositions for oral administration may be in tablet, capsule, powder or liquid form. A tablet may comprise a solid carrier or an adjuvant. Liquid pharmaceutical compositions generally comprise a liquid carrier such as water, petroleum, animal or vegetable oils, mineral oil or synthetic oil. Physiological saline solution, dextrose or other saccharide solution or glycols such as ethylene glycol, propylene glycol or polyethylene glycol may be included. A capsule may comprise a solid carrier such a gelatin.

For intravenous, cutaneous or subcutaneous injection, or injection at the site of affliction, the active ingredient will be in the form of a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability. Those of relevant skill in the art are well able to prepare suitable solutions using, for example, isotonic vehicles such as Sodium Chloride Injection, Ringer's Injection, Lactated Ringer's Injection. Preservatives, stabilisers, buffers, antioxidants and/or other additives may be included, as required.

The Conjugates can be used to treat proliferative disease and autoimmune disease. The term“proliferative disease” pertains 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, gastrointestinal cancer, bowel cancer, colon cancer, breast carinoma, ovarian carcinoma, prostate cancer, testicular cancer, liver cancer, kidney cancer, bladder cancer, pancreatic cancer, brain cancer, sarcoma, osteosarcoma, Kaposi's sarcoma, melanoma), leukemias, psoriasis, bone diseases, fibroproliferative disorders (e.g. of connective tissues), and atherosclerosis. Other cancers of interest include, but are not limited to,

haematological; malignancies such as leukemias and lymphomas, such as non-Hodgkin lymphoma, and subtypes such as DLBCL, marginal zone, mantle zone, and follicular, Hodgkin lymphoma, AML, and other cancers of B or T cell origin. Any type of cell may be treated, including but not limited to, lung, gastrointestinal (including, e.g. bowel, colon), breast (mammary), ovarian, prostate, liver (hepatic), kidney (renal), bladder, pancreas, brain, and skin. Examples of autoimmune disease include the following: rheumatoid arthritis, autoimmune demyelinative diseases (e.g., multiple sclerosis, allergic encephalomyelitis), psoriatic arthritis, endocrine ophthalmopathy, uveoretinitis, systemic lupus erythematosus,

myasthenia gravis, Graves’ disease, glomerulonephritis, autoimmune hepatological disorder, inflammatory bowel disease (e.g., Crohn’s disease), anaphylaxis, allergic reaction, Sjogren’s syndrome, type I diabetes mellitus, primary biliary cirrhosis, Wegener’s granulomatosis, fibromyalgia, polymyositis, dermatomyositis, multiple endocrine failure, Schmidt’s syndrome, autoimmune uveitis, Addison’s disease, adrenalitis, thyroiditis, Hashimoto’s thyroiditis, autoimmune thyroid disease, pernicious anemia, gastric atrophy, chronic hepatitis, lupoid hepatitis, atherosclerosis, subacute cutaneous lupus erythematosus, hypoparathyroidism, Dressler’s syndrome, autoimmune thrombocytopenia, idiopathic thrombocytopenic purpura, hemolytic anemia, pemphigus vulgaris, pemphigus, dermatitis herpetiformis, alopecia areata, pemphigoid, scleroderma, progressive systemic sclerosis, CREST syndrome (calcinosis, Raynaud’s phenomenon, esophageal dysmotility, sclerodactyly, and telangiectasia), male and female autoimmune infertility, ankylosing spondolytis, ulcerative colitis, mixed connective tissue disease, polyarteritis nedosa, systemic necrotizing vasculitis, atopic dermatitis, atopic rhinitis, Goodpasture’s syndrome, Chagas’ disease, sarcoidosis, rheumatic fever, asthma, recurrent abortion, anti-phospholipid syndrome, farmer’s lung, erythema multiforme, post cardiotomy syndrome, Cushing’s syndrome, autoimmune chronic active hepatitis, bird-fancier’s lung, toxic epidermal necrolysis, Alport’s syndrome, alveolitis, allergic alveolitis, fibrosing alveolitis, interstitial lung disease, erythema nodosum, pyoderma gangrenosum, transfusion reaction, Takayasu’s arteritis, polymyalgia rheumatica, temporal arteritis, schistosomiasis, giant cell arteritis, ascariasis, aspergillosis, Sampter’s syndrome, eczema, lymphomatoid granulomatosis, Behcet’s disease, Caplan’s syndrome, Kawasaki’s disease, dengue, encephalomyelitis, endocarditis, endomyocardial fibrosis, endophthalmitis, erythema elevatum et diutinum, psoriasis, erythroblastosis fetalis, eosinophilic faciitis, Shulman’s syndrome, Felty’s syndrome, filariasis, cyclitis, chronic cyclitis, heterochronic cyclitis, Fuch’s cyclitis, IgA nephropathy, Henoch-Schonlein purpura, graft versus host disease, transplantation rejection, cardiomyopathy, Eaton-Lambert syndrome, relapsing polychondritis, cryoglobulinemia, Waldenstrom’s macroglobulemia, Evan’s syndrome, and autoimmune gonadal failure.

In some embodiments, the autoimmune disease is a disorder of B lymphocytes (e.g., systemic lupus erythematosus, Goodpasture’s syndrome, rheumatoid arthritis, and type I diabetes), Th1 -lymphocytes (e.g., rheumatoid arthritis, multiple sclerosis, psoriasis, Sjogren’s syndrome, Hashimoto’s thyroiditis, Graves’ disease, primary biliary cirrhosis, Wegener’s granulomatosis, tuberculosis, or graft versus host disease), or Th2-lymphocytes (e.g., atopic dermatitis, systemic lupus erythematosus, atopic asthma, rhinoconjunctivitis, allergic rhinitis, Omenn’s syndrome, systemic sclerosis, or chronic graft versus host disease). Generally, disorders involving dendritic cells involve disorders of Th1 -lymphocytes or Th2-lymphocytes. In some embodiments, the autoimmunie disorder is a T cell-mediated immunological disorder.

Methods of Treatment

The conjugates of the present invention may be used in a method of therapy. Also provided is a method of treatment, comprising administering to a subject in need of treatment a therapeutically-effective amount of a conjugate compound of the invention. The term “therapeutically effective amount” is an amount sufficient to show benefit to a patient. Such benefit may be at least amelioration of at least one symptom. The actual amount

administered, and rate and time-course of administration, will depend on the nature and severity of what is being treated. Prescription of treatment, e.g. decisions on dosage, is within the responsibility of general practitioners and other medical doctors.

A compound of the invention may be administered alone or in combination with other treatments, either simultaneously or sequentially dependent upon the condition to be treated. Examples of treatments and therapies include, but are not limited to, chemotherapy (the administration of active agents, including, e.g. drugs, such as chemotherapeutics); surgery; and radiation therapy.

A“chemotherapeutic agent” is a chemical compound useful in the treatment of cancer, regardless of mechanism of action. Classes of chemotherapeutic agents include, but are not limited to: alkylating agents, antimetabolites, spindle poison plant alkaloids,

cytotoxic/antitumor antibiotics, topoisomerase inhibitors, antibodies, photosensitizers, and kinase inhibitors. Chemotherapeutic agents include compounds used in“targeted therapy” and conventional chemotherapy.

Examples of chemotherapeutic agents include: erlotinib (TARCEVA®, Genentech/OSI Pharm.), docetaxel (TAXOTERE®, Sanofi-Aventis), 5-FU (fluorouracil, 5-fluorouracil, CAS No. 51-21-8), gemcitabine (GEMZAR®, Lilly), PD-0325901 (CAS No. 391210-10-9, Pfizer), cisplatin (cis-diamine, dichloroplatinum(ll), CAS No. 15663-27-1 ), carboplatin (CAS No. 41575-94-4), paclitaxel (TAXOL®, Bristol-Myers Squibb Oncology, Princeton, N.J.), trastuzumab (HERCEPTIN®, Genentech), temozolomide (4-methyl-5-oxo- 2, 3, 4,6,8- pentazabicyclo [4.3.0] nona-2,7,9-triene- 9-carboxamide, CAS No. 85622-93-1 ,

TEMODAR®, TEMODAL®, Schering Plough), tamoxifen ((Z)-2-[4-(1 ,2-diphenylbut-1- enyl)phenoxy]-/V,/V-dimethylethanamine, NOLVADEX®, ISTUBAL®, VALODEX®), and doxorubicin (ADRIAMYCIN®), Akti-1/2, HPPD, and rapamycin.

More examples of chemotherapeutic agents include: oxaliplatin (ELOXATIN®, Sanofi), bortezomib (VELCADE®, Millennium Pharm.), sutent (SUNITINIB®, SU1 1248, Pfizer), letrozole (FEMARA®, Novartis), imatinib mesylate (GLEEVEC®, Novartis), XL-518 (Mek inhibitor, Exelixis, WO 2007/044515), ARRY-886 (Mek inhibitor, AZD6244, Array BioPharma, Astra Zeneca), SF-1126 (PI3K inhibitor, Semafore Pharmaceuticals), BEZ-235 (PI3K inhibitor, Novartis), XL-147 (PI3K inhibitor, Exelixis), PTK787/ZK 222584 (Novartis), fulvestrant (FASLODEX®, AstraZeneca), leucovorin (folinic acid), rapamycin (sirolimus, RAPAMUNE®, Wyeth), lapatinib (TYKERB®, GSK572016, Glaxo Smith Kline), lonafarnib (SARASAR™, SCH 66336, Schering Plough), sorafenib (NEXAVAR®, BAY43-9006, Bayer Labs), gefitinib (IRESSA®, AstraZeneca), irinotecan (CAMPTOSAR®, CPT-1 1 , Pfizer), tipifarnib (ZARNESTRA™, Johnson & Johnson), ABRAXANE™ (Cremophor-free), albumin- engineered nanoparticle formulations of paclitaxel (American Pharmaceutical Partners, Schaumberg, II), vandetanib (rINN, ZD6474, ZACTIMA®, AstraZeneca), chloranmbucil, AG1478, AG1571 (SU 5271 ; Sugen), temsirolimus (TORISEL®, Wyeth), pazopanib

(GlaxoSmithKline), canfosfamide (TELCYTA®, Telik), thiotepa and cyclosphosphamide (CYTOXAN®, NEOSAR®); 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); a camptothecin (including the synthetic analog topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogs); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogs, 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, calicheamicin gammal l, calicheamicin ornegaH (Angew Chem. Inti. Ed. Engl. (1994) 33:183-186); dynemicin, dynemicin A; bisphosphonates, such as clodronate; an

esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, morpholino-doxorubicin,

cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, nemorubicin, 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 and 5-fluorouracil (5-FU); folic acid analogs 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; podophyllinic acid; 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; dacarbazine; mannomustine; mitobronitol; mitolactol;

pipobroman; gacytosine; arabinoside (“Ara-C”); cyclophosphamide; thiotepa; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin;

vinblastine; etoposide (VP-16); ifosfamide; mitoxantrone; vincristine; vinorelbine

(NAVELBINE®); novantrone; teniposide; edatrexate; daunomycin; aminopterin; capecitabine (XELODA®, Roche); ibandronate; CPT-11 ; topoisomerase inhibitor RFS 2000;

difluoromethylornithine (DMFO); retinoids such as retinoic acid; and pharmaceutically acceptable salts, acids and derivatives of any of the above.

Also included in the definition of“chemotherapeutic agent” are: (i) anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including

NOLVADEX®; tamoxifen citrate), raloxifene, droloxifene, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and FARESTON® (toremifine citrate); (ii) aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, MEGASE® (megestrol acetate), AROMASIN® (exemestane; Pfizer), formestanie, fadrozole, RIVISOR® (vorozole), FEMARA® (letrozole; Novartis), and ARIMIDEX® (anastrozole; AstraZeneca);

(iii) anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; as well as troxacitabine (a 1 ,3-dioxolane nucleoside cytosine analog); (iv) protein kinase inhibitors such as MEK inhibitors (WO 2007/044515); (v) lipid kinase inhibitors; (vi) antisense oligonucleotides, particularly those which inhibit expression of genes in signaling pathways implicated in aberrant cell proliferation, for example, PKC-alpha, Raf and H-Ras, such as oblimersen (GENASENSE®, Genta Inc.); (vii) ribozymes such as VEGF expression inhibitors (e.g., ANGIOZYME®) and HER2 expression inhibitors; (viii) vaccines such as gene therapy vaccines, for example, ALLOVECTIN®, LEUVECTIN®, and VAXID®; PROLEUKIN® rlL-2; topoisomerase 1 inhibitors such as LURTOTECAN®; ABARELIX® rmRH; (ix) anti- angiogenic agents such as bevacizumab (AVASTIN®, Genentech); and pharmaceutically acceptable salts, acids and derivatives of any of the above.

Also included in the definition of“chemotherapeutic agent” are therapeutic antibodies such as alemtuzumab (Campath), bevacizumab (AVASTIN®, Genentech); cetuximab

(ERBITUX®, Imclone); panitumumab (VECTIBIX®, Amgen), rituximab (RITUXAN®, Genentech/Biogen Idee), pertuzumab (OMNITARG™, 2C4, Genentech), trastuzumab (HERCEPTIN®, Genentech), tositumomab (Bexxar, Corixia), and the antibody drug conjugate, gemtuzumab ozogamicin (MYLOTARG®, Wyeth).

Humanized monoclonal antibodies with therapeutic potential as chemotherapeutic agents in combination with the conjugates of the invention include: alemtuzumab, apolizumab, aselizumab, atlizumab, bapineuzumab, bevacizumab, bivatuzumab mertansine, cantuzumab mertansine, cedelizumab, certolizumab pegol, cidfusituzumab, cidtuzumab, daclizumab, eculizumab, efalizumab, epratuzumab, erlizumab, felvizumab, fontolizumab, gemtuzumab ozogamicin, inotuzumab ozogamicin, ipilimumab, labetuzumab, lintuzumab, matuzumab, mepolizumab, motavizumab, motovizumab, natalizumab, nimotuzumab, nolovizumab, numavizumab, ocrelizumab, omalizumab, palivizumab, pascolizumab, peefusituzumab, pectuzumab, pertuzumab, pexelizumab, ralivizumab, ranibizumab, reslivizumab, reslizumab, resyvizumab, rovelizumab, ruplizumab, sibrotuzumab, siplizumab, sontuzumab,

tacatuzumab tetraxetan, tadocizumab, talizumab, tefibazumab, tocilizumab, toralizumab, trastuzumab, tucotuzumab celmoleukin, tucusituzumab, umavizumab, urtoxazumab, and visilizumab. Pharmaceutical compositions according to the present invention, and for use in accordance with the present invention, may comprise, in addition to the active ingredient, i.e. a conjugate compound, a pharmaceutically acceptable excipient, carrier, buffer, stabiliser or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient. The precise nature of the carrier or other material will depend on the route of administration, which may be oral, or by injection, e.g. cutaneous, subcutaneous, or intravenous.

Pharmaceutical compositions for oral administration may be in tablet, capsule, powder or liquid form. A tablet may comprise a solid carrier or an adjuvant. Liquid pharmaceutical compositions generally comprise a liquid carrier such as water, petroleum, animal or vegetable oils, mineral oil or synthetic oil. Physiological saline solution, dextrose or other saccharide solution or glycols such as ethylene glycol, propylene glycol or polyethylene glycol may be included. A capsule may comprise a solid carrier such a gelatin.

For intravenous, cutaneous or subcutaneous injection, or injection at the site of affliction, the active ingredient will be in the form of a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability. Those of relevant skill in the art are well able to prepare suitable solutions using, for example, isotonic vehicles such as Sodium Chloride Injection, Ringer's Injection, Lactated Ringer's Injection. Preservatives, stabilisers, buffers, antioxidants and/or other additives may be included, as required.

Formulations

While it is possible for the conjugate compound to be used (e.g., administered) alone, it is often preferable to present it as a composition or formulation.

In one embodiment, the composition is a pharmaceutical composition (e.g., formulation, preparation, medicament) comprising a conjugate compound, as described herein, and a pharmaceutically acceptable carrier, diluent, or excipient.

In one embodiment, the composition is a pharmaceutical composition comprising at least one conjugate compound, as described herein, together with one or more other

pharmaceutically acceptable ingredients well known to those skilled in the art, including, but not limited to, pharmaceutically acceptable carriers, diluents, excipients, adjuvants, fillers, buffers, preservatives, anti-oxidants, lubricants, stabilisers, solubilisers, surfactants (e.g., wetting agents), masking agents, colouring agents, flavouring agents, and sweetening agents.

In one embodiment, the composition further comprises other active agents, for example, other therapeutic or prophylactic agents.

Suitable carriers, diluents, excipients, etc. can be found in standard pharmaceutical texts. See, for example, Handbook of Pharmaceutical Additives, 2nd Edition (eds. M. Ash and I. Ash), 2001 (Synapse Information Resources, Inc., Endicott, New York, USA), Remington's Pharmaceutical Sciences, 20th edition, pub. Lippincott, Williams & Wilkins, 2000; and

Handbook of Pharmaceutical Excipients, 2nd edition, 1994.

Another aspect of the present invention pertains to methods of making a pharmaceutical composition comprising admixing at least one [ 11 C]-radiolabelled conjugate or conjugate-like compound, as defined herein, together with one or more other pharmaceutically acceptable ingredients well known to those skilled in the art, e.g., carriers, diluents, excipients, etc. If formulated as discrete units (e.g., tablets, etc.), each unit contains a predetermined amount (dosage) of the active compound.

The term“pharmaceutically acceptable,” as used herein, pertains to compounds,

ingredients, materials, compositions, dosage forms, etc., which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of the subject in question (e.g., human) without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. Each carrier, diluent, excipient, etc. must also be“acceptable” in the sense of being compatible with the other ingredients of the formulation.

The formulations may be prepared by any methods well known in the art of pharmacy. Such methods include the step of bringing into association the active compound with a carrier which constitutes one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association the active compound with carriers (e.g., liquid carriers, finely divided solid carrier, etc.), and then shaping the product, if necessary.

The formulation may be prepared to provide for rapid or slow release; immediate, delayed, timed, or sustained release; or a combination thereof. Formulations suitable for parenteral administration (e.g., by injection), include aqueous or non-aqueous, isotonic, pyrogen-free, sterile liquids (e.g., solutions, suspensions), in which the active ingredient is dissolved, suspended, or otherwise provided (e.g., in a liposome or other microparticulate). Such liquids may additional contain other pharmaceutically acceptable ingredients, such as anti-oxidants, buffers, preservatives, stabilisers,

bacteriostats, suspending agents, thickening agents, and solutes which render the formulation isotonic with the blood (or other relevant bodily fluid) of the intended recipient. Examples of excipients include, for example, water, alcohols, polyols, glycerol, vegetable oils, and the like. Examples of suitable isotonic carriers for use in such formulations include Sodium Chloride Injection, Ringer's Solution, or Lactated Ringer's Injection. Typically, the concentration of the active ingredient in the liquid is from about 1 ng/ml to about 10 pg/ml, for example from about 10 ng/ml to about 1 pg/ml. The formulations may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilised) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules, and tablets.

Dosage

It will be appreciated by one of skill in the art that appropriate dosages of the conjugate compound, and compositions comprising the conjugate compound, can vary from patient to patient. Determining the optimal dosage will generally involve the balancing of the level of therapeutic benefit against any risk or deleterious side effects. The selected dosage level will depend on a variety of factors including, but not limited to, the activity of the particular compound, the route of administration, the time of administration, the rate of excretion of the compound, the duration of the treatment, other drugs, compounds, and/or materials used in combination, the severity of the condition, and the species, sex, age, weight, condition, general health, and prior medical history of the patient. The amount of compound and route of administration will ultimately be at the discretion of the physician, veterinarian, or clinician, although generally the dosage will be selected to achieve local concentrations at the site of action which achieve the desired effect without causing substantial harmful or deleterious side-effects.

Administration can be effected in one dose, continuously or intermittently (e.g., in divided doses at appropriate intervals) throughout the course of treatment. Methods of determining the most effective means and dosage of administration are well known to those of skill in the art 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 pattern being selected by the treating physician, veterinarian, or clinician.

In general, a suitable dose of the active compound is in the range of about 100 ng to about 25 mg (more typically about 1 pg to about 10 mg) per kilogram body weight of the subject per day. Where the active compound is a salt, an ester, an amide, a prodrug, or the like, the amount administered is calculated on the basis of the parent compound and so the actual weight to be used is increased proportionately.

In one embodiment, the active compound is administered to a human patient according to the following dosage regime: about 100 mg, 3 times daily.

In one embodiment, the active compound is administered to a human patient according to the following dosage regime: about 150 mg, 2 times daily.

In one embodiment, the active compound is administered to a human patient according to the following dosage regime: about 200 mg, 2 times daily.

However in one embodiment, the conjugate compound is administered to a human patient according to the following dosage regime: about 50 or about 75 mg, 3 or 4 times daily.

In one embodiment, the conjugate compound is administered to a human patient according to the following dosage regime: about 100 or about 125 mg, 2 times daily.

The dosage amounts described above may apply to the conjugate (including the PBD moiety and the linker to the antibody) or to the effective amount of PBD compound provided, for example the amount of compound that is releasable after cleavage of the linker.

For the prevention or treatment of disease, the appropriate dosage of an ADC of the invention will depend on the type of disease to be treated, as defined above, the severity and course of the disease, whether the molecule is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody, and the discretion of the attending physician. The molecule is suitably

administered to the patient at one time or over a series of treatments. Depending on the type and severity of the disease, about 1 mV^V to 15 mg/kg (e.g. 0.1-20 mg/kg) of molecule is an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion. A typical daily dosage might range from about 1 mV^V to 100 mg/kg or more, depending on the factors mentioned above. An exemplary dosage of ADC to be administered to a patient is in the range of about 0.1 to about 10 mg/kg of patient weight. For repeated administrations over several days or longer, depending on the condition, the treatment is sustained until a desired suppression of disease symptoms occurs. An exemplary dosing regimen comprises a course of administering an initial loading dose of about 4 mg/kg, followed by additional doses every week, two weeks, or three weeks of an ADC. Other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.

Drug loading

The drug loading (p) is the average number of PBD drugs per cell binding agent, e.g.

antibody. Where the compounds of the invention are bound to cysteines, drug loading may range from 1 to 8 drugs (D) per cell binding agent, i.e. where 1 , 2, 3, 4, 5, 6, 7, and 8 drug moieties are covalently attached to the cell binding agent. Compositions of conjgates include collections of cell binding agents, e.g. antibodies, conjugated with a range of drugs, from 1 to 8. Where the compounds of the invention are bound to lysines, drug loading may range from 1 to 80 drugs (D) per cell binding agent, although an upper limit of 40, 20, 10 or 8 may be preferred. Compositions of conjgates include collections of cell binding agents, e.g. antibodies, conjugated with a range of drugs, from 1 to 80, 1 to 40, 1 to 20, 1 to 10 or 1 to 8.

The average number of drugs per antibody in preparations of ADC from conjugation reactions may be characterized by conventional means such as UV, reverse phase HPLC, HIC, mass spectroscopy, ELISA assay, and electrophoresis. The quantitative distribution of ADC in terms of p may also be determined. By ELISA, the averaged value of p in a particular preparation of ADC may be determined (Hamblett et al (2004) Clin. Cancer Res.

10:7063-7070; Sanderson et al (2005) Clin. Cancer Res. 1 1 :843-852). However, the distribution of p (drug) values is not discernible by the antibody-antigen binding and detection limitation of ELISA. Also, ELISA assay for detection of antibody-drug conjugates does not determine where the drug moieties are attached to the antibody, such as the heavy chain or light chain fragments, or the particular amino acid residues. In some instances, separation, purification, and characterization of homogeneous ADC where p is a certain value from ADC with other drug loadings may be achieved by means such as reverse phase HPLC or electrophoresis. Such techniques are also applicable to other types of conjugates. For some antibody-drug conjugates, p may be limited by the number of attachment sites on the antibody. For example, an antibody may have only one or several cysteine thiol groups, or may have only one or several sufficiently reactive thiol groups through which a linker may be attached. Higher drug loading, e.g. p >5, may cause aggregation, insolubility, toxicity, or loss of cellular permeability of certain antibody-drug conjugates.

Typically, fewer than the theoretical maximum of drug moieties are conjugated to an antibody during a conjugation reaction. An antibody may contain, for example, many lysine residues that do not react with the Drug Linker. Only the most reactive lysine groups may react with an amine-reactive linker reagent. Also, only the most reactive cysteine thiol groups may react with a thiol-reactive linker reagent. Generally, antibodies do not contain many, if any, free and reactive cysteine thiol groups which may be linked to a drug moiety. Most cysteine thiol residues in the antibodies of the compounds exist as disulfide bridges and must be reduced with a reducing agent such as dithiothreitol (DTT) or TCEP, under partial or total reducing conditions. The loading (drug/antibody ratio) of an ADC may be controlled in several different manners, including: (i) limiting the molar excess of Drug Linker relative to antibody, (ii) limiting the conjugation reaction time or temperature, and (iii) partial or limiting reductive conditions for cysteine thiol modification.

Certain antibodies have reducible interchain disulfides, i.e. cysteine bridges. Antibodies may be made reactive for conjugation with linker reagents by treatment with a reducing agent such as DTT (dithiothreitol). Each cysteine bridge will thus form, theoretically, two reactive thiol nucleophiles. Additional nucleophilic groups can be introduced into antibodies through the reaction of lysines with 2-iminothiolane (Traut’s reagent) resulting in conversion of an amine into a thiol. Reactive thiol groups may be introduced into the antibody (or fragment thereof) by engineering one, two, three, four, or more cysteine residues (e.g., preparing mutant antibodies comprising one or more non-native cysteine amino acid residues). US 7521541 teaches engineering antibodies by introduction of reactive cysteine amino acids.

Cysteine amino acids may be engineered at reactive sites in an antibody and which do not form intrachain or intermolecular disulfide linkages (Junutula, et al., 2008b Nature Biotech., 26(8):925-932; Dornan et al (2009) Blood 114(13):2721 -2729; US 7521541 ; US 7723485; W02009/052249). The engineered cysteine thiols may react with linker reagents or the drug-linker reagents of the present invention which have thiol-reactive, electrophilic groups such as maleimide or alpha-halo amides to form ADC with cysteine engineered antibodies and the PBD drug moieties. The location of the drug moiety can thus be designed, controlled, and known. The drug loading can be controlled since the engineered cysteine thiol groups typically react with thiol-reactive linker reagents or drug-linker reagents in high yield. Engineering an IgG antibody to introduce a cysteine amino acid by substitution at a single site on the heavy or light chain gives two new cysteines on the symmetrical antibody. A drug loading near 2 can be achieved with near homogeneity of the conjugation product ADC.

Where more than one nucleophilic or electrophilic group of the antibody reacts with a drug- linker intermediate, or linker reagent followed by drug moiety reagent, then the resulting product is a mixture of ADC compounds with a distribution of drug moieties attached to an antibody, e.g. 1 , 2, 3, etc. Liquid chromatography methods such as polymeric reverse phase (PLRP) and hydrophobic interaction (HIC) may separate compounds in the mixture by drug loading value. Preparations of ADC with a single drug loading value (p) may be isolated, however, these single loading value ADCs may still be heterogeneous mixtures because the drug moieties may be attached, via the linker, at different sites on the antibody.

Thus the antibody-drug conjugate compositions of the invention include mixtures of antibody-drug conjugate compounds where the antibody has one or more PBD drug moieties and where the drug moieties may be attached to the antibody at various amino acid residues.

In one embodiment, the average number of dimer pyrrolobenzodiazepine groups per cell binding agent is in the range 1 to 20. In some embodiments the range is selected from 1 to 8, 2 to 8, 2 to 6, 2 to 4, and 4 to 8.

In some embodiments, there is one dimer pyrrolobenzodiazepine group per cell binding agent.

General synthetic routes

The synthesis of PBD compounds is extensively discussed in the following references, which discussions are incorporated herein by reference:

a) WO 00/12508 (pages 14 to 30);

b) WO 2005/023814 (pages 3 to 10);

c) WO 2004/043963 (pages 28 to 29); and

d) WO 2005/085251 (pages 30 to 39). Synthesis route

Compounds of the present invention of formula I may be synthesised from compounds of formula IP:

where R L pre represents a precursor to the group R L . Where R° is H, the carboxy group may be protected during synthesis (for example a tert-butyl ester), which can be removed in a final step. For example, when R L is a group:

, or a protected version thereof. The addition of G L may be achieved by conventional means. R30 and R31 may also be a nitrogen protecting group and OProt° respectively, when Prot° represents an oxygen protecting group for synthesis, which are removed as appropriate.

Alternatively, compounds of formula IP may be synthesised by coupling compounds of formulae 2 and 3:

where Hal is selected from I, Cl, and Br, and Prot° represents an oxygen protecting group for synthesis, following by removal of the Prot° group under standard conditions.

The coupling can be achieved, for example, in refluxing acetone with a base, such as K2CO3.

Compounds of formula 2 can be synthesised from compounds of formula 4:

By coupling a compound of Formula 5:

Hal-R”-Q Formula 5 where Q is selected from I, Cl, and Br, the reaction can be achieved, for example, in refluxing acetone with a base, such as K2CO 3 . An excess of the compound of Formula 5 is required to achieve the desired product.

The compound of formula 4 may be synthesised from a compound of Formula 6:

where Prot r is a protecting group for Y’ that is orthogonal to the other protecting groups in the compound. The synthesis is achieved by deprotection of Y’, under standard conditions. Compounds of formula 3 can be synthesised from compounds of formula 7:

where Prot Y is a protecting group for Y that is orthogonal to the other protecting groups in the compound. The synthesis is achieved by deprotection of Y, under standard conditions. Compounds of formula 7 can be synthesised from compounds of formula 8:

by protecting the OH group with Prot°, under standard conditions.

Compounds of formula 8 can be synthesised from compounds of formula 9:

by oxidation. The oxidation may be carried out, for example, with Dess-Martin periodinane (or alternatively TPAP/NMO, TFAA/DMSO, SO3. Pyridine complex/DMSO, PDC, PCC, BAIB/TEMPO or under Swern conditions).

Compounds of formula 9 can be synthesised from compounds of formula 10:

by deprotection of the OH group under standard conditions. Compounds of Formula 10 can also be used to make compounds of Formula 1 1 :

which may be coupled to a compound of Formula 2, following by cyclisation.

The coupling of the two PBD (or pre-PBD) moieities to the group comprising R” may be carried out in either order. Synthesis of Drug Conjugates

Conjugates can be prepared as previously described. Antibodies can be conjugated to the Drug Linker compound as described in Doronina et al., Nature Biotechnology, 2003, 21 , 778- 784). Briefly, antibodies (4-5 mg/mL) in PBS containing 50 mM sodium borate at pH 7.4 are reduced with tris(carboxyethyl)phosphine hydrochloride (TCEP) at 37 °C. The progress of the reaction, which reduces interchain disulfides, is monitored by reaction with 5,5’- dithiobis(2-nitrobenzoic acid) and allowed to proceed until the desired level of thiols/mAb is achieved. The reduced antibody is then cooled to 0°C and alkylated with 1.5 equivalents of maleimide drug-linker per antibody thiol. After 1 hour, the reaction is quenched by the addition of 5 equivalents of N-acetyl cysteine. Quenched drug-linker is removed by gel filtration over a PD-10 column. The ADC is then sterile-filtered through a 0.22 pm syringe filter. Protein concentration can be determined by spectral analysis at 280 nm and 329 nm, respectively, with correction for the contribution of drug absorbance at 280 nm. Size exclusion chromatography can be used to determine the extent of antibody aggregation, and RP-HPLC can be used to determine the levels of remaining NAC-quenched drug-linker.

Further Preferences

The following preferences may apply to all aspects of the invention as described above, or may relate to a single aspect. The preferences may be combined together in any

combination.

R 6’ , R 7’ , R 9’ , and Y’ are selected from the same groups as R 6 , R 7 , R 9 and Y respectively. In some embodiments, R 6’ , R 7’ , R 9’ , and Y’ are the same as R 6 , R 7 , R 9 and Y respectively.

In some embodiments, R 2 is the same as R 2 .

Dimer link

Y and Y’ are preferably O.

In some embodiments, n is 1. In other embodiments, n is 2. In other embodiments, n is 3.

In some embodiments, m is 1. In other embodiments, m is 2. In other embodiments, m is 3. In certain embodiments, n and m are the same. In some of these embodiments, n and m are 1. In other of these embodiments, n and m are 2. In other of these embodiments, n and m are 3.

In some embodiments, Q is N. In other embodiments, Q is CH.

In some embodiments, R° is H. In other embodiments, R° is methyl. In other

embodiments, R° is ethyl. In other embodiments, R° is iso-propyl. In other embodiments, R° is benzyl.

In particular embodiments, R" is of formula I la:

lla

where R° 1 is selected from the group consisting of H and methyl.

R 6 to R 9

In some embodiments, R 9 is H.

In some embodiments, R 6 is selected from H, OH, OR, SH, NH2, nitro and halo, and may be selected from H or halo. In some of these embodiments R 6 is H.

In some embodiments, R 7 is selected from H, OH, OR, SH, SR, NH2, NHR, NRR’, and halo. In some of these embodiments R 7 is selected from H, OH and OR, where R is selected from optionally substituted C alkyl, C3-10 heterocyclyl and C5-10 aryl groups. R may be more preferably a C1-4 alkyl group, which may or may not be substituted. A substituent of interest is a C5-6 aryl group (e.g. phenyl). Particularly preferred substituents at the 7- positions are OMe and OCH2PI7. Other substituents of particular interest are dimethylamino (i.e. -NMe2); -(OC 2 H 4 ) q OMe, where q is from 0 to 2; nitrogen-containing O Q heterocyclyls, including morpholino, piperidinyl and N-methyl-piperazinyl.

These embodiments and preferences apply to R 9’ , R 6 and R 7 respectively. R 2

When there is a double bond present between C2 and C3, R 2 is selected from:

(a) C 5-10 aryl group, optionally substituted by one or more substituents selected from the group comprising: halo, nitro, cyano, ether, C alkyl, C 3-7 heterocyclyl and bis-oxy-Ci- 3 alkylene;

(b) C 1-5 saturated aliphatic alkyl;

(c) C3-6 saturated cycloalkyl;

, wherein each of R 11 , R 12 and R 13 are independently selected from H, C1-3 saturated alkyl, C2-3 alkenyl, C2-3 alkynyl and cyclopropyl, where the total number of carbon atoms in the R 2 group is no more than 5;

(e) , wherein one of R 15a and R 15b is H and the other is selected from:

phenyl, which phenyl is optionally substituted by a group selected from halo methyl, methoxy; pyridyl; and thiophenyl; and

(f) , where R 14 is selected from: H; C1-3 saturated alkyl; C2-3 alkenyl; C2-3 alkynyl; cyclopropyl; phenyl, which phenyl is optionally substituted by a group selected from halo methyl, methoxy; pyridyl; and thiophenyl.

When R 2 is a C 5-10 aryl group, it may be a C 5-7 aryl group. A C 5-7 aryl group may be a phenyl group or a C 5-7 heteroaryl group, for example furanyl, thiophenyl and pyridyl. In some embodiments, R 2 is preferably phenyl. In other embodiments, R 2 is preferably thiophenyl, for example, thiophen-2-yl and thiophen-3-yl.

When R 2 is a C 5-10 aryl group, it may be a Cs-io aryl, for example a quinolinyl or isoquinolinyl group. The quinolinyl or isoquinolinyl group may be bound to the PBD core through any available ring position. For example, the quinolinyl may be quinolin-2-yl, quinolin-3-yl, quinolin-4yl, quinolin-5-yl, quinolin-6-yl, quinolin-7-yl and quinolin-8-yl. Of these quinolin-3-yl and quinolin-6-yl may be preferred. The isoquinolinyl may be isoquinolin-1 -yl, isoquinolin-3- yl, isoquinolin-4yl, isoquinolin-5-yl, isoquinolin-6-yl, isoquinolin-7-yl and isoquinolin-8-yl. Of these isoquinolin-3-yl and isoquinolin-6-yl may be preferred. When R 2 is a C5-10 aryl group, it may bear any number of substituent groups. It preferably bears from 1 to 3 substituent groups, with 1 and 2 being more preferred, and singly substituted groups being most preferred. The substituents may be any position.

Where R 2 is C5-7 aryl group, a single substituent is preferably on a ring atom that is not adjacent the bond to the remainder of the compound, i.e. it is preferably b or y to the bond to the remainder of the compound. Therefore, where the C5-7 aryl group is phenyl, the substituent is preferably in the meta- or para- positions, and more preferably is in the para- position.

Where R 2 is a Ce-io aryl group, for example quinolinyl or isoquinolinyl, it may bear any number of substituents at any position of the quinoline or isoquinoline rings. In some embodiments, it bears one, two or three substituents, and these may be on either the proximal and distal rings or both (if more than one substituent).

R 2 substituents, when R 2 is a C 5-10 aryl group

If a substituent on R 2 when R 2 is a C5-10 aryl group is halo, it is preferably F or Cl, more preferably Cl.

If a substituent on R 2 when R 2 is a C5-10 aryl group is ether, it may in some embodiments be an alkoxy group, for example, a C1-7 alkoxy group (e.g. methoxy, ethoxy) or it may in some embodiments be a C5-7 aryloxy group (e.g phenoxy, pyridyloxy, furanyloxy). The alkoxy group may itself be further substituted, for example by an amino group (e.g. dimethylamino).

If a substituent on R 2 when R 2 is a C5-10 aryl group is C1-7 alkyl, it may preferably be a C1-4 alkyl group (e.g. methyl, ethyl, propryl, butyl).

If a substituent on R 2 when R 2 is a C5-10 aryl group is C3-7 heterocyclyl, it may in some embodiments be C 6 nitrogen containing heterocyclyl group, e.g. morpholino, thiomorpholino, piperidinyl, piperazinyl. These groups may be bound to the rest of the PBD moiety via the nitrogen atom. These groups may be further substituted, for example, by Ci -4 alkyl groups.

If the C 6 nitrogen containing heterocyclyl group is piperazinyl, the said further substituent may be on the second nitrogen ring atom. If a substituent on R 2 when R 2 is a C 5-10 aryl group is bis-oxy-C- 1-3 alkylene, this is preferably bis-oxy-methylene or bis-oxy-ethylene.

If a substituent on R 2 when R 2 is a C 5-10 aryl group is ester, this is preferably methyl ester or ethyl ester.

Particularly preferred substituents when R 2 is a C 5-10 aryl group include methoxy, ethoxy, fluoro, chloro, cyano, bis-oxy-methylene, methyl-piperazinyl, morpholino and methyl- thiophenyl. Other particularly preferred substituents for R 2 are dimethylaminopropyloxy and carboxy.

Particularly preferred substituted R 2 groups when R 2 is a C 5-10 aryl group include, but are not limited to, 4-methoxy-phenyl, 3-methoxyphenyl, 4-ethoxy-phenyl, 3-ethoxy-phenyl, 4-fluoro- phenyl, 4-chloro-phenyl, 3,4-bisoxymethylene-phenyl, 4-methylthiophenyl, 4-cyanophenyl, 4- phenoxyphenyl, quinolin-3-yl and quinolin-6-yl, isoquinolin-3-yl and isoquinolin-6-yl, 2-thienyl, 2-furanyl, methoxynaphthyl, and naphthyl. Another possible substituted R 12 group is 4- nitrophenyl. R 12 groups of particular interest include 4-(4-methylpiperazin-1 -yl)phenyl and 3,4-bisoxymethylene-phenyl.

When R 2 is C 1-5 saturated aliphatic alkyl, it may be methyl, ethyl, propyl, butyl or pentyl. In some embodiments, it may be methyl, ethyl or propyl (n-pentyl or isopropyl). In some of these embodiments, it may be methyl. In other embodiments, it may be butyl or pentyl, which may be linear or branched.

When R 2 is C 3-6 saturated cycloalkyl, it may be cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl. In some embodiments, it may be cyclopropyl.

When , each of R 11 , R 12 and R 13 are independently selected from H, C 1-3 saturated alkyl, C 2-3 alkenyl, C 2-3 alkynyl and cyclopropyl, where the total number of carbon atoms in the R 2 group is no more than 5. In some embodiments, the total number of carbon atoms in the R 2 group is no more than 4 or no more than 3. In some embodiments, one of R 11 , R 12 and R 13 is H, with the other two groups being selected from H, C 1-3 saturated alkyl, C 2-3 alkenyl, C 2-3 alkynyl and cyclopropyl.

In other embodiments, two of R 11 , R 12 and R 13 are H, with the other group being selected from H, C 1-3 saturated alkyl, C 2-3 alkenyl, C 2-3 alkynyl and cyclopropyl.

In some embodiments, the groups that are not H are selected from methyl and ethyl. In some of these embodiments, the groups that re not H are methyl.

In some embodiments, R 1 1 is H.

In some embodiments, R 12 is H.

In some embodiments, R 13 is H.

In some embodiments, R 1 1 and R 12 are H.

In some embodiments, R 1 1 and R 13 are H.

In some embodiments, R 12 and R 13 are H.

An R 2 group of particular interest is:

When R 2 is , one of R 15a and R 15b is H and the other is selected from: phenyl, which phenyl is optionally substituted by a group selected from halo, methyl, methoxy; pyridyl; and thiophenyl. In some embodiments, the group which is not H is optionally substituted phenyl. If the phenyl optional substituent is halo, it is preferably fluoro. In some embodiment, the phenyl group is unsubstituted.

When R 2 is R 14 is selected from: H; C 1-3 saturated alkyl; C 2-3 alkenyl; C 2-3 alkynyl; cyclopropyl; phenyl, which phenyl is optionally substituted by a group selected from halo methyl, methoxy; pyridyl; and thiophenyl. If the phenyl optional substituent is halo, it is preferably fluoro. In some embodiment, the phenyl group is unsubstituted.

In some embodiments, R 14 is selected from H, methyl, ethyl, ethenyl and ethynyl. In some of these embodiments, R 14 is selected from H and methyl.

When there is a single bond present between C2 and C3,

R 2 is H or , where R 16a and R 16b are independently selected from H, F, Ci -4 saturated alkyl, C2-3 alkenyl, which alkyl and alkenyl groups are optionally substituted by a group selected from C1-4 alkyl amido and Ci -4 alkyl ester; or, when one of R 16a and R 16b is H, the other is selected from nitrile and a Ci -4 alkyl ester.

In some embodiments, R 2 is H.

In some embodiments,

In some embodiments, it is preferred that R 16a and R 16b are both H.

In other embodiments, it is preferred that R 16a and R 16b are both methyl.

In further embodiments, it is preferred that one of R 16a and R 16b is H, and the other is selected from Ci -4 saturated alkyl, C2-3 alkenyl, which alkyl and alkenyl groups are optionally substituted. In these further embodiment, it may be further preferred that the group which is not H is selected from methyl and ethyl.

R 2’

The above preferences for R 2 apply equally to R 2’ .

R 11a

In some embodiments, R 11a is OH.

In some embodiments, R 11a is OR A . In some of these embodiments, R A is methyl. In some embodiments, R 11a is selected from OH, OR A , where R A is Ci -4 alkyl, and SO z M, where z is 2 or 3 and M is a monovalent pharmaceutically acceptable cation.

R 30 and R 31

In some embodiments, R 30 is H, and R 31 is OH. In other embodiments, R 30 is H, and R 31 is OR A . In some of these embodiments, R A is methyl.

In other embodiment, R 30 and R 31 form a nitrogen-carbon double bond between the nitrogen and carbon atoms to which they are bound.

In other embodiments, R 30 is H and R 31 is SO z M, where z is 2 or 3 and M is a monovalent pharmaceutically acceptable cation.

M and z

It is preferred that M is a monovalent pharmaceutically acceptable cation, and is more preferably Na + . z is preferably 3.

Linker (R L )

In some embodiments, R L is of formula Ilia.

In some embodiments, R LL is of formula Ilia’.

G L

G L may be selected from

where Ar represents a C 5-6 arylene group, e.g. phenylene, and X represents C 1-4 alkyl. In some embodiments, G L is selected from G L1 1 and G L1 2 . In some of these embodiments, G L is G L1 - 1 .

G LL G LL may be selected from:

In some embodiments, G LL is selected from G LL1 1 and G LL1 2 . In some of these

embodiments, G LL is G LL1 1 .

where a = 0 to 5, b = 0 to 16, c = 0 or 1 , d = 0 to 5. a may be 0, 1 , 2, 3, 4 or 5. In some embodiments, a is 0 to 3. In some of these

embodiments, a is 0 or 1. In further embodiments, a is 0. b may be 0, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15 or 16. In some embodiments, b is 0 to 12. In some of these embodiments, b is 0 to 8, and may be 0, 2, 4 or 8. c may be 0 or 1 . d may be 0, 1 , 2, 3, 4 or 5. In some embodiments, d is 0 to 3. In some of these

embodiments, d is 1 or 2. In further embodiments, d is 2.

In some embodiments of X, a is 0, c is 1 and d is 2, and b may be from 0 to 8. In some of these embodiments, b is 0, 4 or 8.

Q

In one embodiment, Q is an amino acid residue. The amino acid may a natural amino acids or a non-natural amino acid. In one embodiment, Q is selected from: Phe, Lys, Val, Ala, Cit, Leu, lie, Arg, and Trp, where Cit is citrulline.

In one embodiment, Q comprises a dipeptide residue. The amino acids in the dipeptide may be any combination of natural amino acids and non-natural amino acids. In some embodiments, the dipeptide comprises natural amino acids. Where the linker is a cathepsin labile linker, the dipeptide is the site of action for cathepsin-mediated cleavage. The dipeptide then is a recognition site for cathepsin.

In one embodiment, Q is selected from:

c o -Phe-Lys- NH ,

c o -Val-Ala- NH ,

c o -Val-Lys- NH ,

c o -Ala-Lys- NH ,

c o -Val-Cit- NH ,

c o -Phe-Cit- NH ,

c o -Leu-Cit- NH ,

c o -lle-Cit- NH ,

c o -Phe-Arg- NH , and

c o -Trp-Cit- NH ;

where Cit is citrulline.

Preferably, Q is selected from:

c o -Phe-Lys- NH ,

c o -Val-Ala- NH ,

c o -Val-Lys- NH ,

c o -Ala-Lys- NH ,

c o -Val-Cit- NH .

Most preferably, Q is selected from co -Phe-Lys- NH , co -Val-Cit- NH and co -Val-Ala- NH .

Other dipeptide combinations of interest include:

c o -Gly-Gly- NH ,

c°-Pro-Pro- NH , and

c o -Val-Glu- NH . Other dipeptide combinations may be used, including those described by Dubowchik et al., Bioconjugate Chemistry, 2002, 13,855-869, which is incorporated herein by reference.

In some embodiments, Q x is a tripeptide residue. The amino acids in the tripeptide may be any combination of natural amino acids and non-natural amino acids. In some

embodiments, the tripeptide comprises natural amino acids. Where the linker is a cathepsin labile linker, the tripeptide is the site of action for cathepsin-mediated cleavage. The tripeptide then is a recognition site for cathepsin.

In one embodiment, the amino acid side chain is chemically protected, where appropriate. The side chain protecting group may be a group as discussed below. Protected amino acid sequences are cleavable by enzymes. For example, a dipeptide sequence comprising a Boc side chain-protected Lys residue is cleavable by cathepsin.

Protecting groups for the side chains of amino acids are well known in the art and are described in the Novabiochem Catalog, and as described above.

In some embodiments, R L is of formula II lb.

In some embodiments, R LL is is formula 11 lb’.

R L1 and R L2 are independently selected from H and methyl, or together with the carbon atom to which they are bound form a cyclopropylene or cyclobutylene group.

In some embodiments, both R L1 and R L2 are H.

In some embodiments, R L1 is H and R L2 is methyl.

In some embodiments, both R L1 and R L2 are methyl.

In some embodiments, R L1 and R L2 together with the carbon atom to which they are bound form a cyclopropylene group.

In some embodiments, R L1 and R L2 together with the carbon atom to which they are bound form a cyclobutylene group. In the group lllb, in some embodiments, e is 0. In other embodiments, e is 1 and the nitro group may be in any available position of the ring. In some of these embdoiments, it is in the ortho position. In others of these embodiments, it is in the para position.

In one particular embodiment, the first aspect of the invention comprises a compound of

In some embodiments of the first aspect of the present invention are of formula la, lb or lc:

where R 2a and R 2a are the same and are selected from:

(h)

R 7a and R 7a are the same and are selected from methoxy and benzyloxy;

R°, R L1 and R 11a are as defined above.

In some embodiments of the third aspect of the present invention, the drug linkers (D L ) are of formula Ilia, Nib or I lie:

where R 2a and R 2a are the same and are selected from: R 7a and R 7a are the same and are selected from methoxy and benzyloxy;

R°, R L1a and R 11a are as defined above.

Examples

General Information

Flash chromatography was performed using a Biotage Isolera 1™ using gradient elution with hexane/EtOAc or CFhCh/MeOH mixtures as indicated until all UV active components eluted from the column. The gradient was manually held whenever substantial elution of UV active material was observed. Fractions were checked for purity using thin-layer chromatography (TLC) using Merck Kieselgel 60 F254 silica gel, with fluorescent indicator on aluminium plates. Visualisation of TLC was achieved with UV light or iodine vapour unless otherwise stated. Extraction and chromatography solvents were bought and used without further purification from VWR U.K. All fine chemicals were purchased from Sigma-Aldrich or VWR.

The analytical LC/MS conditions (for reaction monitoring and purity determination) were as follows: Positive mode electrospray mass spectrometry was performed using a Shimadzu Nexera®/Prominence® LCMS-2020. Mobile phases used were solvent A (FhO with 0.1 % formic acid) and solvent B (CH3CN with 0.1 % formic acid).

LCMS-A: Gradient for 3-minute run: Initial composition 5% B held over 25 seconds, then increased from 5% B to 100% B over a 1 minute 35 seconds’ period. The composition was held for 50 seconds at 100% B, then returned to 5% B in 5 seconds and held there for 5 seconds. The total duration of the gradient run was 3.0 minutes at a flow rate of 0.8 mL/min. Column: Waters Acquity UPLC® BEH Shield RP18 1.7pm 2.1 x 50 mm at 50 °C fitted with Waters Acquity UPLC® BEH Shield RP18 VanGuard Pre-column, 130A, 1.7pm, 2.1 mm x 5 mm

LCMS-B: Gradient for 15-minute run: Initial composition 5% B held over 1 minute, then increased from 5% B to 100% B over a 9 minute period. The composition was held for 2 minutes at 100% B, then returned to 5% B in 10 seconds and held there for 2 minutes 50 seconds. The total duration of the gradient run was 15.0 minutes at a flow rate of 0.6 mL/minute. Detection was monitored at 254 nm, 223 nm and 214 nm. ACE Excel 2 C18-AR, 2 m, 3.0 x 100mm fitted with Waters Acquity UPLC® BEH Shield RP18 VanGuard Pre- column, 130A, 1 7pm, 2.1 mm x 5 mm. Intermediates

Synthesis of tert-butyl (11S,11aS)-11 -((tert-butyldimethylsilyl)oxy)-8-hydroxy-7- methoxy-2-methylene-5-oxo-2,3,11,11 a-tetrahydro-1 H-benzo[e]pyrrolo[1 ,2- a][1 ,4]diazepine-10(5H)-carboxylate (I5)

I6 is Compound 152 of WO 2018/182341

(i) tert-Butyl (S)-(2-(2-(((tert-butyldimethylsilyl)oxy)methyl)-4-methylene pyrrolidine-1 - carbonyl)-4-methoxy-5-((triisopropylsilyl)oxy)phenyl)carbama te (I7)

I6 (400 g, 1 eq.), CH2CI2 (4 L) and B0C2O (206.7 g, 1.3 eq.) were added to a reactor and stirred for 20 h at 70 °C. The mixture was cooled to rt and concentrated in vacuo to afford I7 (450 g, 95%) which was used without further purification.

(ii) tert-Butyl (S)-(2-(2-(hvdroxymethyl)-4-methylenepyrrolidine-1 -carbonyl)-4-methoxy-5- ((triisopropylsilyl)oxy)phenyl)carbamate (I8)

I7 (210 g, 1.0 eq.) THF (76 mL), MeOH (76 mL) AcOH (533 mL) and H 2 0 (151 mL) were added to a reactor and stirred for 20 h. The mixture was diluted with EtOAc (30 V) and H2O (20 V) and the mixture vigorously stirred. The organic phase was washed with sat. NaHCC>3 solution (20 V), then brine (20 V) and dried over Na 2 S0 4 and concentrated in vacuo. Flash column chromatography (8:1 - 2:1 petroleum ether/EtOAc) afforded I8 as a white solid (150 g, 87%).

(iii) tert-Butyl (1 1 S,1 1 aS)-1 1 -hvdroxy-7-methoxy-2-methylene-5-oxo-8-((triisopropylsilyl)o xy)-

2,3,1 1 ,1 1 a-tetrahydro-1 H-benzorelpyrroloH ,2-alH ,4ldiazepine-10(5H)-carboxylate (I9)

(COCI)2 (28.5 g, 1 .0 eq.) wass added to CH2CI2 (900 mL) in a reactor under N2 and the mixture was cooled to -80°C. A solution of I8 (120 g, 1.0 eq.) in CH2CI2 (600 mL) was added dropwise over 30 min, then EίbN (1 13.5 g, 5.0 eq.) was added dropwise over 30 min. The mixture was warmed to rt and stirred for 5 h the quenched by the addition of 5% aqueous citric acid (10 V). The organic phase was washed successively with sat. NaHCC>3 (5 V) and H2O (5 V), dried over Na 2 S0 4 and concentrated in vacuo. The crude product was stirred in heptane (10 V) at 90 °C, then cooled to 0-5°C. The resulting white precipitate was isolated by filtration and dried in a vacuum oven at 50°C to afford I9 (105 g, 88%).

(iv) tert-Butyl (1 1 S, 1 1 aS)-1 1 -((tert-butyldimethylsilyl)oxy)-7-methoxy-2-methylene-5-oxo- 8-

((triisopropylsilyl)oxy)-2,3,1 1 ,1 1 a-tetrahydro-1 H-benzorelpyrroloH ,2-alH ,4ldiazepine-10(5H)- carboxylate (110)

I9 (150 g, 1.0 eq.) and CH2CI2 (1 .5 L, 10 V) were charged to a reactor and the mixture cooled to 0-5°C. Lutidine (120.6 g, 4.0 eq.), then TBSOTf (223.2 g, 3.0 eq.) were added and the mixture was stirred for 10 min at 0-10°C, then 4 h at rt. The reaction was quenched by the addition of 5% aqueous citric acid (5 V) and the organic phase was successively washed with sat. NaHCC>3 (5 V), H2O (5 V) then dried over Na 2 S0 4 and concentrated in vacuo to afford 5 (179 g, 98%) as an orange oil.

(v) tert-Butyl (1 1 S, 1 1 aS)-1 1 -((tert-butyldimethylsilyl)oxy)-8-hvdroxy-7-methoxy-2-methyl ene-

5-oxo-2,3,1 1 ,1 1 a-tetrahydro-1 H-benzorelpyrroloH ,2-alH ,4ldiazepine-10(5H)-carboxylate (I5)

DMF (1 .83 L, 1 V), H2O (37.4 mL, 0.2 V) and 110 (187 g, 1.0 eq.) were charged to a reactor. LiOAc (19.1 g, 1.0 eq.) was added and the mixture stirred for 20 h at 10-20°C. The reaction mixture was diluted with cold water (10 V) and EtOAc (20 V). The aqueous phase was washed with EtOAc (10 V), the organic phases combined, successively washed with 5% aqueous citric acid (10 V), 5% aqueous NaHCOs (10 V) and H2O (10 V) and dried over Na 2 S0 4 and concentrated in vacuo. The resulting crude was dissolved in EtOAc (6 V) at 77°C, heptane (12 V) was added and the mixture cooled to 0-5°C, the white precipitate collected by filtration and dried at 50°C in a vacuum oven to afford 6 (101 g, 71 %). Example 1 : 3-((((11 S,11aS)-10-(((4-((S)-2-((S)-2-(3-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 - yl)propanamido)-3-methylbutanamido)propanamido)benzyl)oxy)ca rbonyl)-11 -hydroxy- 7-methoxy-2-methylene-5-oxo-2,3,5,10,11 ,11a-hexahydro-1 H-benzo[e]pyrrolo[1 ,2- a][1 ,4]diazepin-8-yl)oxy)methyl)-5-((((S)-7-methoxy-2-methylene- 5-oxo-2,3,5,11a- tetrahydro-1 H-benzo[e]pyrrolo[1 ,2-a][1 ,4]diazepin-8-yl)oxy)methyl)benzoic acid (19)

(i) (((5-iodo-1 ,3-phenylene)b/s(methylene))ib/s(oxy))bis(te/f-butyldimethyl silane) (2)

A round-bottomed flask was charged with a magnetic stirrer bar, DMF (100 ml_), diol 1 (10.0 g, 37.87 mmol, 1.0 eq.), TBSCI (17.1 g, 1 13.6 mmol, 3.0 eq.) imidazole (15.5 g, 227.2 mmol, 6.0 eq.) and the mixture stirred for 20 min, whereupon LCMS indicated the reaction was complete. The reaction mixture was filtered, diluted with EtOAc, washed with water, the organic phase dried over MgS0 4 and concentrated in vacuo. Isolera (0-8% EtOAc in hexane) afforded the desired product as a colourless oil (17.9 g, 96%). LCMS-A: 2.29 min (ES+) no ionisation.

(ii) tert- butyl 3,5-b/s(((te/f-butyldimethylsilyl)oxy)methyl)benzoate (3)

An oven-dried 3-necked round-bottomed flask was charged with a magnetic stirrer bar anhydrous toluene (7.5 ml.) and the flask purged with Ar and the flask cooled to -10 °C. BuLi (5.6 ml. of a 2.5 M solution in hexanes, 14.00 mmol, 2.0 eq.), then slowly BuMgCI (3.5 ml. of a 2.0 M solution in THF, 7.000 mmol, 1.0 eq.) were added and stirred for 30 min. 2 (8.21 g,

16.67 mmol, 2.4 eq.) in anhydrous toluene (15 ml.) was added slowly and stirred for 2 h at - 10 °C. B0C2O (4.55 g, 20.84 mmol, 3.0 eq.) in anhydrous toluene (7.5 ml.) was added slowly at -10 °C and stirred for 1 h, then poured into cold 10% citric acid. The aqueous phase was extracted with EtOAc, the organics combined, dried over MgS0 4 and concentrated in vacuo. Isolera purification (0-20% EtOAc in hexane afforded the product as a colourless oil (7.39 g, 95%). LCMS-A: 1.48 min (ES+) no ionisation.

(iii) tert- butyl 3,5-b/s(hvdroxymethyl)benzoate (4)

A round-bottomed flask was charged with a magnetic stirrer, 3 (7.39 g, 15.83 mmol, 1.0 eq.) and THF (75 mL). TBAF (31.7 mL of a 1 M solution, 31.70 mmol, 2.0 eq.) was added by syringe and stirred for 20 min whereupon LCMS indicated the reaction was complete. The reaction mixture was diluted with EtOAc, washed with H2O, dried over MgS0 4 and concentrated in vacuo to afford the product as a white crystalline solid (3,29 g, 87%).

LCMS-A: 1.24 min (ES+) no ionisation.

(iv) tert- butyl 3,5-b/s(bromomethyl)benzoate (5)

A round-bottomed flask was charged with a magnetic stirrer, 4 (3.29 g, 15.83 mmol, 1.0 eq.), CH2CI2 (200 mL) and cooled to -78 °C. PPfi 3 (17.4 g, 66.34 mmol, 4.2 eq.) was added, then CBr 4 (23.1 g, 69.65 mmol, 4.4 eq.) was added portionwise and the reaction allowed to warm to rt. The reaction was filtered through S1O2 (C^Ch/hexane) and purified by isolera (0-20% EtOAc in hexane) to afford the product as a white solid (1.43 g, 26%).

LCMS-A: 1.81 min (ES+) no ionisation.

(v) Methyl (S)-1-(5-methoxy-2-nitro-4-((tri/sopropylsilyl)oxy)benzoyl)- 4-methylenepyrrolidine- 2-carboxylate (8)

A round-bottomed flask was charged with a magnetic stirrer, THF (15 mL), 6 (1.89 g, 5.1 18 mmol, 1.0 eq.), HOBt (761 mg, 5.630 mmol, 1.1 eq.), DIC (872 pL, 5.630 mmol, 1.1 eq.) and / ' -Pr2NEt (1.96 mL, 11.26 mmol, 2.2 eq.) and the mixture was stirred for 10 min. 7 (1.00 g, 5.630 mmol, 1.1 eq.) was added portionwise and the reaction mixture was stirred for 1 h.

The reaction was diluted with CH2CI2, washed with 0.2N HCI solution, the organics dried over MgS0 4 and concentrated in vacuo. Flash column chromatography (20-30% EtOAc in hexane) afforded the desired product as a yellow oil (1.19 g, 47%). LCMS-A: 2.00 min (ES+) m/z 493 [M + H] + , 515 [M + Na] +

(vi) (S)-(2-(hvdroxymethyl)-4-methylenepyrrolidin-1-yl)(5-methoxy -2-nitro-4- ((tri/sopropylsilyl)oxy)phenyl)methanone (9)

A round-bottomed flask was charged with a magnetic stirrer, THF (15 ml_), 8 (1.17 g, 2.383 mmol, 1.0 eq.) and the stirring mixture was cooled to 0 °C. Cautiously, LiBFU (156 mg, 7.149 mmol, 3.0 eq.) was added and the mixture was stirred for 1 h at 0 °C and a further 1.5 h at rt. The reaction was quenched by the addition of ice-cold H2O and the pH adjusted to ca. 6 with 1 N HCI. The aqueous mixture was extracted with CH2CI2, the organics combined, dried over MgS0 4 and concentrated in vacuo to afford the desired product as a yellow foam (1.06 g, 96%) which was used without further purification. LCMS-A: 1.92 min (ES+) m/z 465 [M +

H] + , 487 [M + Na] +

(vii) (S)-(2-(((te/f-butyldimethylsilyl)oxy)methyl)-4-methylenepyr rolidin-1-yl)(5-methoxy-2- nitro-4-((tri/sopropylsilyl)oxy)phenyl)methanone (10)

A round-bottomed flask was charged with a magnetic stirrer, CH2CI2 (10 mL), 9 (1.06 g,

2.281 mmol, 1.0 eq.), imidazole (467 mg, 6.858 mmol, 3.0 eq.) TBSCI (517 mg, 3.429 mmol, 1.5 eq.) and stirred for 1 h, whereupon LCMS indicated the reaction was complete. The reaction mixture was filtered, the filtrate washed with H2O, brine, dried over MgS0 4 and concentrated in vacuo. Flash column chromatography (20-30% EtOAc in hexane) afforded the desired product as a yellow oil (1.27 g, 96%). LCMS-A: 2.30 min (ES+) m/z 579 [M +

H] + , 601 [M + Na] +

(viii) (S)-(2-amino-5-methoxy-4-((tri/sopropylsilyl)oxy)phenyl)(2-( ((te/f- butyldimethylsilyl)oxy)methyl)-4-methylenepyrrolidin-1-yl)me thanone (1 1 )

A three-necked round-bottomed flask was charged with a magnetic stirrer, 10 (10.0 g, 17.27 mmol, 1.0 eq.), EtOH (170 mL), Zn (23.6 g, 345.4 mmol, 20 eq.) and cooled to 0 °C. 5% HCO2H in EtOH (340 mL) was added dropwise by dropping funnel and stirred for 30 min, whereupon, LCMS indicated reaction was complete. The reaction mixture was diluted with EtOAc (200 mL) and filtered through Celite. Sat. NaHCOs solution was added to the filtrate until the pH of the aqueous phase was basic. The phases were separated, the aqueous phase was washed with EtOAc (2 x 100 mL), the organic phases combined, washed with brine, dried over MgS0 4 and concentrated in vacuo to afford a yellow oil which was used without further purification (8.99 g, 95%). LCMS-A: 2.23 min (ES+) m/z 549 [M + H] + , 571 [M + Na] +

(ix) 4-((S)-2-((S)-2-(((allyloxy)carbonyl)amino)-3-methylbutanami do)propanamido)benzyl (2- ((S)-2-(((te/f-butyldimethylsilyl)oxy)methyl)-4-methylenepyr rolidine-1-carbonyl)-4-methoxy-5- ((tri/sopropylsilyl)oxy)phenyl)carbamate (12)

A round-bottomed flask was charged with a magnetic stirrer, 11 (8.99 g, 16.38 mmol, 1.0 eq.), THF (90 ml_), EίbN (5.4 ml_, 35.90 mmol, 2.2 eq.), the flask purged with Ar and the mixture cooled to -10 °C. Triphosgene (1.75 g, 5.897 mmol, 0.36 eq.) was added in one portion over a flow of Ar. After 5 min, LCMS indicated isocyanate formation complete (by way of MeOH quench and observation of methyl carbamate). 11 (9.27 g, 24.57 mmol, 1.5 eq.) was added in one portion over a flow of Ar and the reaction mixture was warmed to room temperature and stirred for 24 h. The reaction mixture was filtered, concentrated onto Si0 2 in vacuo and purified by flash-column chromatography (0-3% MeOH in CH2CI2) to afford the product (10.9 g, 70%). LCMS-A: 2.20 min (ES+) m/z 952 [M + H] + , 974 [M + Na] +

(x) 4-((S)-2-((S)-2-(((allyloxy)carbonyl)amino)-3-methylbutanami do)propanamido)benzyl (2- ((S)-2-(((te/f-butyldimethylsilyl)oxy)methyl)-4-methylenepyr rolidine-1-carbonyl)-5-hvdroxy-4- methoxyphenvDcarbamate (13)

A round-bottomed flask was charged with a magnetic stirrer, silyl ether 12 (10.9 g, 11.47 mmol, 1.0 eq.) DMF (85 mL), H2O (1.7 mL) and LiOAc (1.17 g, 1 1.47 mmol, 1.0 eq.). The reaction mixture was stirred for 16 h, whereupon LCMS indicated the reaction was complete. The reaction mixture was diluted with EtOAc (150 mL) and washed successively with H2O (50 mL) and brine (50 mL). The organic phase was dried over MgS0 4 and purified by Isolera chromatography (0-4% MeOH in CH2CI2) to afford the product as a yellow foam (6.65 g, 73%). LCMS-A: 1.78 min (ES+) m/z 796 [M + H] + , 818 [M + Na] +

(xi) tert-butyl 3-((5-((((4-((S)-2-((S)-2-(((allyloxy)carbonyl)amino)-3- methylbutanamido)propanamido)benzyl)oxy)carbonyl)amino)-4-(( S)-2-(((te/f- butyldimethylsilyl)oxy)methyl)-4-methylenepyrrolidine-1-carb onyl)-2- methoxyphenoxy)methyl)-5-(bromomethyl)benzoate (14)

A round-bottomed flask was charged with a magnetic stirrer, phenol 13 (1.09 g, 1.364 mmol, 1 eq.), bis bromo 5 (1.43 g, 4.093 mmol, 3.0 eq.), DMF (10 mL) and K2CO 3 (207 mg, 1.500 mmol, 1.1 eq.) and the reaction stirred for 2.5 h at 35 °C, whereupon LCMS indicated the reaction was complete. The reaction mixture was diluted with H2O and washed with CH2CI2, dried over MgS0 4 and concentrated in vacuo. Isolera purification (10-50% EtOAc in hexane) afforded the product as an off-white foam (946 mg, 64%). LCMS-A: 2.06 min (ES+) m/z 1 100.20 [M + Na] +

(xiini 1 S.11 aSV8-((3-((5-((((4-((SV2-((SV2-(((allyloxy)carbonvnaminoV3- methylbutanamido)propanamido)benzyl)oxy)carbonyl)amino)-4-(( S)-2-(((te/f- butyldimethylsilyl)oxy)methyl)-4-methylenepyrrolidine-1-carb onyl)-2- methoxyphenoxy)methyl)-5-(te/f-butoxycarbonyl)benzyl)oxy)-7- methoxy-2-methylene-5-oxo-

1 1-((tetrahvdro-2H-pyran-2-yl)oxy)-2,3,11 ,11 a-tetrahvdro-1 H-benzorelpyrrolon ,2- alH ,4ldiazepine-10(5H)-carboxylate (15)

A round-bottomed flask was charged with a magnetic stirrer, 14 (946 mg, 0.8766 mmol, 1.0 eq.), I2 (444 mg, 0.9643 mmol, 1.1 eq.), DMF (12 ml.) and K2CO3 (133 mg, 0.9643 mmol,

1.1 eq.) were stirred for 3.5 h at 35 °C whereupon LCMS indicated the reaction was complete. The reaction mixture was diluted with H2O, washed with EtOAc and the organic phase was dried over MgS0 4 and concentrated in vacuo. Isolera purification (40-100% EtOAc in hexane afforded the product as a white foam (1.04 g, 82%). LCMS-A: 2.1 1 min (ES+) m/z 1458.20 [M + H] + , 1480.35 [M + Na] +

(xiii) te/f-butyl (1 1 S.11 aSV8-((3-((5-((((4-((SV2-((SV2-(((allyloxy)carbonvnamino 3- methylbutanamido)propanamido)benzyl)oxy)carbonyl)amino)-4-(( S)-2-(hvdroxymethyl)-4- methylenepyrrolidine-1-carbonyl)-2-methoxyphenoxy)methyl)-5- (te/f- butoxycarbonyl)benzyl)oxy)-7-methoxy-2-methylene-5-oxo-11-(( tetrahydro-2H-pyran-2- yl)oxy)-2,3,11 ,11 a-tetrahydro-1 H- alH ,4ldiazepine-10(5H)-carboxylate

(16)

A round-bottomed flask was charged with a magnetic stirrer, 15(1.04 g, 0.7129 mmol, 1.0 eq.), THF (40 mL) AcOH (82 pL, 1.430 mmol, 2.0 eq.) and TBAF (2.1 mL of a 1 M solution, 2.100 mmol, 3.0 eq.) and the reaction stirred for 1 h whereupon LCMS indicated the reaction was complete. The reaction mixture was diluted with CHCI 3 and washed with sat. NaHCC> 3 , the organic phase dried over MgS0 4 and concentrated in vacuo. Isolera purification (0-4% MeOH in CH 2 CI 2 ) afforded the product as a white foam (961 mg, 100%). LCMS-A: 1.86 min (ES+) m/z 1344.70 [M + H] + , 1366.85 [M + Na] +

(xiv) 4-((S)-2-((S)-2-(((allyloxy)carbonyl)amino)-3-methylbutanami do)propanamido)benzyl (1 1 S,1 1aS)-8-((3-(te/f-butoxycarbonyl)-5-((((11 S,1 1aS)-10-(te/f-butoxycarbonyl)-7-methoxy- 2-methylene-5-oxo-11-((tetrahvdro-2H-pyran-2-yl)oxy)-2,3,5,1 0,1 1 ,1 1a-hexahvdro-1 H- benzorelpyrrolor 1 ,2-aH 1 ,4ldiazepin-8-yl)oxy)methyl)benzyl)oxy)-1 1 -hydroxy-7-methoxy-2- methylene-5-oxo-2,3,1 1 ,1 1a-tetrahydro-1 H-benzorelpyrroloH ,2-alH ,4ldiazepine-10(5H)- carboxylate (17)

A round-bottomed flask was charged with a magnetic stirrer, 16 (961 mg, 0.7150 mmol, 1.0 eq.) DMSO (7 ml.) and IBX (489 mg, 0.7865, 1.1 eq.) and the reaction mixture stirred at 35 °C for 16 h. IBX (50 mg, 80.35 pmol, 0.1 1 eq.) was added and the reaction mixture stirred for 48 h, whereupon the reaction mixture was poured into water and washed with 10% MeOH in CH2CI2, the organic layers combined, dried over MgS0 4 and concentrated in vacuo. Isolera purification (0-4% MeOH in CH2CI2) afforded the product as a white foam (492 mg, 51%). Mixed fractions were purified again by isolera (0-2.5% MeOH in CH2CI2) affording the product as a white solid (302 mg, 31%). Both clean fractions were combined to afford the product as a white foam (794 mg, 83%). LCMS-A: 1.83 min (ES+) m/z 1364.55 [M + Na] +

(xy) terf-butyl (11 S.1 1aSV8-((3-(terf-butoxycarbonvn-5-((((11 S.11 aS)-10-(((4-((S)-2-((S)-2- (3-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)propanamido)-3- methylbutanamido)propanamido)benzyl)oxy)carbonyl)-1 1-hvdroxy-7-methoxy-2-methylene- 5-oxo-2,3,5,10,1 1 ,1 1a-hexahvdro-1 H-benzorelpyrroloH ,2-alH ,41diazepin-8- yl)oxy)methyl)benzyl)oxy)-7-methoxy-2-methylene-5-oxo-11-((t etrahvdro-2H-pyran-2-yl)oxy)- 2,3,11 ,11 a-tetrahvdro-1 H-benzorelpyrroloH ,2-alH ,4ldiazepine-10(5H)-carboxylate (18)

Around-bottomed flask was charged with a magnetic stirrer, 17 (794 mg, 0.5914 mmol, 1.0 eq.), CH2CI2 (50 ml_), pyrrolidine (121 pl_, 1.479 mmol, 2.5 eq.) and the flask purged with Ar. Pd(PPh3)4 (34 mg, 29.58 pmol, 0.050 eq.) was added over a flow of Ar and the reaction mixture stirred for 10 min, whereupon LCMS indicated the reaction was complete. The reaction mixture was thoroughly concentrated in vacuo overnight to remove pyrrolidine traces. The resulting residue was dissolved in CH2CI2 (10 ml.) and maleimide propionic acid (78 mg, 0.4612, 1.1 eq.), EDCI.HCI (880 mg, 0.4589 mmol, 1.1 eq.) and /-Pr 2 NEt (218 pL,

1.252 mmol, 3.0 eq.) were added and the mixture stirred for 5 h, whereupon LCMS indicated the reaction was incomplete. A further 0.1 eq of EDCI.HCI, 0.1 eq. of maleimide propionic acid and 0.3 eq. of / ' -P^NEt were added and the mixture stirred for 16 h, whereupon LCMS indicated the reaction was incomplete. A further 0.2 eq of EDCI.HCI, 0.2 eq. of maleimide propionic acid and 0.6 eq. of / ' -P^NEt were added and the mixture stirred for 24 h, diluted with CH2CI2 (20 mL), washed with H2O (10 mL) the organic phase dried over MgS0 4 and concentrated in vacuo. Isolera chromatography (2-6% MeOH in CH2CI2) afforded the desired product (205 mg, 35%). LCMS-A: 1.77 min (ES+) m/z 1410 [M + H] + ,.1431 [M + Na] +

(xyi) 3-((((11 S.11 aS)-10-(((4-((S)-2-((S ' )-2-(3-(2.5-dioxo-2.5-dihvdro-1 H-pyrrol-1 - yl)propanamido)-3-methylbutanamido)propanamido)benzyl)oxy)ca rbonyl)-1 1 -hydroxy-7- methoxy-2-methylene-5-oxo-2,3,5,10,1 1 ,1 1a-hexahydro-1 H-benzorelpyrrolon ,2- aH1 , 4ldiazepin-8-yl)oxy)methyl)-5-((((S)-7-methoxy-2-methylene-5 -oxo-2, 3,5,11 a-tetrahvdro-

1 H-benzorelpyrrolor 1 ,2-aH 1 ,4ldiazepin-8-yl)oxy)methyl)benzoic acid (19)

A round-bottomed flask was charged with a magnetic stirrer, 18 (205 mg, 0.1454 mmol, 1.0 eq.) and cooled to ca. -5 °C. 95:5 v/v TFA/H2O was cooled to ca. -20 °C and added to the reaction flask and stirred for 75 min, whereupon LCMS indicated the reaction was complete. The pH of the reaction mixture was adjusted to ca. 7 by the cautious addition of sat.

NaHCC>3, whereupon a precipitate formed. The aqueous mixture was extracted with 10% MeOH in CH2CI2, dried over Na 2 S0 4 and concentrated in vacuo. Preparative HPLC (15-70% A in B) afforded the product as a white solid (52 mg, 31%, QC=94%). A = 0.05% formic acid in acetonitrile. B = 0.05% formic acid in water

LCMS-B: 7.13 min (ES+) m/z 1 151 [M + H] +

Example 2: 4-((S)-2-((S)-2-(3-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)propanamido)-3- methylbutanamido)propanamido)benzyl (11 S,11aS)-11 -hydroxy-7-methoxy-8-((3-((((S)- 7-methoxy-2-methylene-5-oxo-2,3,5,11 a-tetrahydro-1 H-benzo[e]pyrrolo[1 ,2- a][1 ,4]diazepin-8-yl)oxy)methyl)-5-(methoxycarbonyl)benzyl)oxy)- 2-methylene-5-oxo- 2,3,11 ,11 a-tetrahydro-1 H-benzo[e]pyrrolo[1 ,2-a][1 ,4]diazepine-10(5H)-carboxylate (27)

(i) Methyl 3,5-b/s(((te/f-butyldimethylsilyl)oxy)methyl)benzoate (20)

An oven-dried 3-necked round-bottomed flask was charged with a magnetic stirrer bar anhydrous toluene (15 ml.) and the flask purged with Ar and the flask cooled to -10 °C. BuLi (9.9 ml. of a 2.5 M solution in hexanes, 24.86 mmol, 2.0 eq.), then slowly BuMgCI (6.2 ml. of a 2.0 M solution in THF, 12.40 mmol, 1.0 eq.) were added and stirred for 30 min. 2 (14.7 g, 29.84 mmol, 2.4 eq.) in anhydrous toluene (30 ml.) was added slowly and stirred for 2 h at - 10 °C. CICC>2Me (2.9 ml_, 37.29 mmol, 3.0 eq.) in anhydrous toluene (15 ml.) was added slowly at -50 °C and stirred for 2 h, then poured into cold 10% citric acid. The aqueous phase was extracted with EtOAc, the organics combined, washed with NaHCC>3, dried over MgS0 4 and concentrated in vacuo. Isolera purification (0-20% EtOAc in hexane afforded the product as a pale-yellow oil (1 1.5 g, 91%). LCMS-A: 2.18 min (ES+) m/z 425 [M + H] +

(ii) Methyl 3,5-b/s(bromomethyl)benzoate (21 )

A round-bottomed flask was charged with a magnetic stirrer, AcOH (15 ml_), 20 (1.00 g, 2.354 mmol, 1.0 eq.) and HBr (33% in AcOH) (10 ml.) and the mixture was stirred for 2 h at 50 °C. The reaction mixture was cooled to rt, poured into ice-cold water, the resulting precipitate collected by filtration, dissolved in CH2CI2, dried over MgS0 4 and concentrated in vacuo. Isolera (0-50% CH2CI2 in hexane) afforded the desired product as an off-white solid (460 mg, 61%). LCMS-A: 1.59 min (ES+) No ionisation

(iii) tert- Butyl (11 S,11 aS)-8-((3-(bromomethyl)-5-(methoxycarbonyl)benzyl)oxy)-7-met hoxy-2- methylene-5-oxo-1 1-((tetrahvdro-2H-pyran-2-yl)oxy)-2,3,11 ,11 a-tetrahvdro-1 H- benzorelpyrrolori ,2-aln ,4ldiazepine-10(5H)-carboxylate (22)

A round-bottomed flask was charged with a magnetic stirrer, DMF (20 mL), I2 (256 mg, 0.5559 mmol, 1.0 eq.) 21 (716 mg, 2.224 mmol, 4.0 eq.) and K2CO 3 (84.5 mg, 0.61 15 mmol,

1.1 eq.). The mixture was stirred for 1 h 45 min, whereupon LCMS indicated the reaction was complete. DMF was removed in vacuo, the resulting residue dissolved in CH2CI2 and washed with sat. NH 4 CI solution. The organic phase was collected, dried over MgS0 4 and purified by isolera (10-60% EtOAc in heptane) to afford the desired product as a white solid (234 mg, 60%). LCMS-A: 1.77 min (ES+) m/z 701 [M + H] + , 723 [M + Na] +

(iv) tert- Butyl (1 1 S.1 1aSV8-((3-((5-((((4-((SV2-((S)-2-(((allyloxy)carbonvnaminoV3 - methylbutanamido)propanamido)benzyl)oxy)carbonyl)amino)-4-(( S)-2-(((te/f- butyldimethylsilyl)oxy)methyl)-4-methylenepyrrolidine-1-carb onyl)-2- methoxyphenoxy)methyl)-5-(methoxycarbonyl)benzyl)oxy)-7-meth oxy-2-methylene-5-oxo- 1 1-((tetrahvdro-2H-pyran-2-yl)oxy)-2,3,11 ,11 a-tetrahvdro-1 H-benzorelpyrrolon ,2- aH1 ,4ldiazepine-10(5H)-carboxylate (23)

A round-bottomed flask was charged with a magnetic stirrer, DMF (2 mL), 22 (234 mg, 0.3335 mmol, 1.0 eq.), 13 (292 mg, 0.3669 mmol, 1.1 eq.) and K2CO 3 (51 mg, 0.3690 mmol,

1.1 eq.). The mixture was stirred for 4 h whereupon LCMS indicated the reaction was complete. DMF was removed in vacuo, the resulting residue dissolved in CH2CI2 and washed with sat. NhUCI solution. The organic phase was collected, dried over MgS0 4 and purified by isolera (0-3% MeOH in CH2CI2) to afford the desired product as a white solid (450 mg, 95%). LCMS-A: 2.00 min (ES+) m/z 1416 [M + H] + , 1438 [M + Na] +

(v) tert- Butyl (11 S.1 1aSV8-((3-((5-((((4-((SV2-((SV2-(((allyloxy)carbonvnamino 3- methylbutanamido)propanamido)benzyl)oxy)carbonyl)amino)-4-(( S)-2-(hvdroxymethyl)-4- methylenepyrrolidine-1-carbonyl)-2-methoxyphenoxy)methyl)-5-

(methoxycarbonyl)benzyl)oxy)-7-methoxy-2-methylene-5-oxo- 1 1-((tetrahydro-2H-pyran-2- yl)oxy)-2,3,11 ,11 a-tetrahydro-1 H- alH ,4ldiazepine-10(5H)-carboxylate

(24)

A round-bottomed flask was charged with a magnetic stirrer, THF (18 ml_), 23 (450 mg, 0.3176 mmol, 1.0 eq.), AcOH (36.3 mI_, 0.6352 mmol, 2.0 eq.) and TBAF (953 pl_ of 1 M solution in THF, 0.9530 mmol, 3.0 eq.). The mixture was stirred for 7 h at rt, then a few drops of TBAF were added and stirred at rt for a further 3 h, whereupon LCMS indicated reaction was complete. The reaction mixture was diluted with EtOAc and washed with H2O, then brine, dried over MgS0 4 and concentrated in vacuo. Isolera (0-4% MeOH in CH2CI2) afforded the desired product with Bu 4 N+ salts as an inseparable mixture. The resulting material was used without further purification. LCMS-A: 1.73 min (ES+) m/z 1302 [M + H] + , 1324 [M + Na] +

(vi) 4-((S)-2-((S)-2-(((allyloxy)carbonyl)amino)-3-methylbutanami do)propanamido)benzyl

(1 1 S,1 1aS)-8-((3-((((11 S,11 aS)-10-(te/f-butoxycarbonyl)-7-methoxy-2-methylene-5-oxo-1 1- ((tetrahvdro-2H-pyran-2-yl)oxy)-2,3,5,10,1 1 ,1 1a-hexahvdro-1 H-benzorelpyrrolon ,2- alH ,4ldiazepin-8-yl)oxy)methyl)-5-(methoxycarbonyl)benzyl)oxy)- 11-hydroxy-7-methoxy-2- methylene-5-oxo-2,3,1 1 ,1 1a-tetrahydro-1 H-benzorelpyrroloH ,2-alH ,4ldiazepine-10(5H)- carboxylate (25)

A round-bottomed flask was charged with a magnetic stirrer, DMSO (3 mL), 24 (ca. 0.3176 mmol, 1.0 eq.) and IBX (45% wt.) (237 mg, 0.381 1 mmol, 1.2 eq.). The mixture was sturred at rt for 16 h, then a knifepoint of IBX was added and was stirred for 35 °C for 24 h. The reaction mixture was diluted with H2O and extracted with CHCI3. The organics were combined, dried over MgS0 4 and concentrated in vacuo. Isolera (0-4% MeOH in CH2CI2) afforded the desired product as a white solid (333 mg, 81% (2 steps)). LCMS-A: 1.71 min (ES+) m/z 1300 [M + H] + , 1323 [M + Na] +

(vii) te/f-butyl (1 1 S.11 aSV8-((3-((((11 S.1 1aSV10-(((4-((SV2-((SV2-(3-(2.5-dioxo-2.5-dihvdro- 1 H-pyrrol-1 -yl)propanamido)-3-methylbutanamido)propanamido)benzyl)oxy)c arbonyl)-1 1 - hvdroxy-7-methoxy-2-methylene-5-oxo-2,3,5,10,1 1 ,1 1a-hexahvdro-1 H-benzorelpyrrolon ,2- alH ,4ldiazepin-8-yl)oxy)methyl)-5-(methoxycarbonyl)benzyl)oxy)- 7-methoxy-2-methylene-5- oxo-11-((tetrahvdro-2H-pyran-2-yl)oxy)-2,3,1 1 ,1 1a-tetrahydro-1 H-benzorelpyrroloH ,2- alH ,4ldiazepine-10(5H)-carboxylate (26)

A round-bottomed flask was charged with a magnetic stirrer, CH2CI2 (19 ml_), 25 (333 mg, 0.2561 mmol, 1.0 eq.), pyrrolidine (42.1 mI_, 0.5122 mmol, 2.0 eq.) and Pd(PPh3)4 (7.4 mg, 6.403 pmol, 0.025 eq.). The reaction mixture was stirred for 20 min, whereupon LCMS indicated the reaction was complete. The reaction mixture was thoroughly concentrated in vacuo overnight to remove pyrrolidine traces. The resulting residue was dissolved in CH2CI2 (3.5 ml.) and maleimide propionic acid NHS ester (82 mg, 0.3080 mmol, 1.2 eq.) and / ' - Pr2NEt (53.5 mI_, 0.3073 mmol, 1.2 eq.) were added and the mixture stirred for 2 h, whereupon LCMS indicated the reaction was complete. The reaction mixture was concentrated in vacuo and purified by isolera (0-4% MeOH in CH2CI2) to afford the desired product as a white solid (150 mg, 43%). LCMS-A: 1.65 min (ES+) m/z 1368 [M + H] + , 1389 [M + Na] +

(viii) 4-((S)-2-((S)-2-(3-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)propanamido)-3- methylbutanamido)propanamido)benzyl (1 1 S,11 aS)-1 1-hvdroxy-7-methoxy-8-((3-((((S)-7- methoxy-2-methylene-5-oxo-2,3,5,11 a-tetrahvdro-1 H-benzorelpyrroloH , 2-alH ,4ldiazepin-8- yl)oxy)methyl)-5-(methoxycarbonyl)benzyl)oxy)-2-methylene-5- oxo-2,3,1 1 ,1 1a-tetrahvdro- 1 H-benzorelpyrrolori ,2-aln ,4ldiazepine-10(5H)-carboxylate (27)

A round-bottomed flask was charged with a magnetic stirrer, 26 (150 mg, 0.1097 mmol, 1.0 eq.) and cooled to 0 °C. 95% TFA in H2O was cooled to -20 °C and added to the reaction flask in one portion and the mixture stirred for 30 min, whereupon LCMS indicated the reaction was complete. The reaction mixture was poured into ice cold sat. NaHCC>3 solution and solid NaHCC>3 was added until the pH was approximately neutral. The aqueous mixture was extracted with 10% MeOH in CH2CI2, the organics combined, dried over MgS0 4 and concentrated in vacuo. Purification by isolera (0-5% MeOH in CH2CI2) afforded the desired product as a white solid (52.8 mg, 41% yield, 98.1% QC). LCMS-A: 7.95 min (ES+) m/z 1 165 [M + H] + Example 3: 4-((S)-2-((S)-2-(3-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)propanamido)-3- methylbutanamido)propanamido)benzyl (11 S,11aS)-11 -hydroxy-7-methoxy-8-((3-((((S)- 7-methoxy-2-methyl-5-oxo-5,11 a-dihydro-1 H-benzo[e]pyrrolo[1 ,2-a][1 ,4]diazepin-8- yl)oxy)methyl)-5-(methoxycarbonyl)benzyl)oxy)-2 -methyl -5-oxo-11 ,11 a-dihydro-1 H- benzo[e]pyrrolo[1 ,2-a][1 ,4]diazepine-10(5H)-carboxylate (33) and 3-((((11 S,11aS)-10- (((4-((S)-2-((S)-2-(3-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)propanamido)-3- methylbutanamido)propanamido)benzyl)oxy)carbonyl)-11 -hydroxy-7-methoxy-2- methyl-5-oxo-5,10,11 ,11 a-tetrahydro-1 H-benzo[e]pyrrolo[1 ,2-a][1 ,4]diazepin-8- yl)oxy)methyl)-5-((((S)-7-methoxy-2-methyl-5-oxo-5,11 a-dihydro-1 H- benzo[e]pyrrolo[1 ,2-a][1 ,4]diazepin-8-yl)oxy)methyl)benzoic acid (34)

(i) Allyl (1 1 S,1 1 aS)-8-((3-(bromomethyl)-5-(methoxycarbonyl)benzyl)oxy)-1 1 -((te/f- butyldimethylsilyl)oxy)-7-methoxy-2-methyl-5-oxo-1 1 ,1 1 a-dihydro-1 H-benzorelpyrrolon ,2- aH1 ,4ldiazepine-10(5H)-carboxylate (28) A round-bottomed flask was charged with a magnetic stirrer, DMF (15 ml_), I3 (184 mg, 0.3883 mmol, 1.0 eq.) 21 (500 mg, 1.553 mmol, 4.0 eq), K 2 CO 3 (59 mg, 0.4271 mmol, 1.1 eq.) and TBAI (3.6 mg, 9.708 pmol, 0.025 eq.) and the mixture stirred at 65 °C for 16 h. DMF was removed in vacuo, the resulting residue dissolved in EtOAc and washed with 1 N HCI solution. The organic phase was dried over MgS0 4 and concentrated in vacuo. Flash column chromatography (0-30% EtOAc in hexane) afforded the desired product (183 mg, 69%). LCMS-A: 2.06 min (ES+) m/z 714 [M + H] + , 737 [M + Na] +

(ii) AIM (1 1 S.1 1aSV8-((3-((((11 S.1 1aSV10-(((4-((SV2-((SV2-(((allyloxy)carbonvnaminoV3- methylbutanamido)propanamido)benzyl)oxy)carbonyl)-1 1-((te/f-butyldimethylsilyl)oxy)-7- methoxy-2-methyl-5-oxo-5,10,11 ,1 1a-tetrahvdro-1 H-benzolelpyrrolori ,2-alH ,4ldiazepin-8- yl)oxy)methyl)-5-(methoxycarbonyl)benzyl)oxy)-1 1-((te/f-butyldimethylsilyl)oxy)-7-methoxy-2- methyl-5-oxo-11 ,1 1a-dihydro-1 H-benzolelpyrrolori , 2-alH ,4ldiazepine-10(5H)-carboxylate (29)

A round-bottomed flask was charged with a magnetic stirrer, DMF (1 ml.) 28 (128 mg,

0.1788 mmol, 1.0 eq.), I4 (353 mg, 0.4446 mmol, 2.5 eq.), K2CO3 (27 mg, 0.1967 mmol, 1.1 eq.) and TBAI (1.7 mg, 4.470 pmol, 0.025 eq.) and the mixture was stirred for 20 h at 65 °C. DMF was removed in vacuo, the resulting residue dissolved in EtOAc and washed with H2O, then brine. The organic phase was dried over MgS0 4 and concentrated in vacuo. Flash column chromatography (0-3% MeOH in CH2CI2) afforded the desired product (1 17 mg, 46%). LCMS - A : 2.20 min (ES+) m/z 1428 [M + H] + , 1450 [M + Na] +

(iii) 3-((((1 1 S,1 1aS)-10-((allyloxy)carbonyl)-11-((te/f-butyldimethylsilyl)ox y)-7-methoxy-2- methyl-5-oxo-5,10,11 ,1 1a-tetrahvdro-1 H-benzorelpyrroloH ,2-alH ,4ldiazepin-8- v0oxylmethyll-5-((((1 1 S.11 aSI-10-(((4-((SV2-((SV2-(((allyloxylcarbonvnaminol-3- methylbutanamido)propanamido)benzyl)oxy)carbonyl)-1 1-((te/f-butyldimethylsilyl)oxy)-7- methoxy-2-methyl-5-oxo-5,10,11 ,1 1a-tetrahvdro-1 H-benzolelpyrrolori ,2-alH ,4ldiazepin-8- yl)oxy)methyl)benzoic acid (30)

A microwave vial was charged with a magnetic stirrer, CHCI3 (1 mL) 29 (100 mg, 0.06999 mmol, 1.0 eq.) SnMesOH (127 mg, 0.6999 mmol, 10 eq.) and irradiated at 100 °C for 10 h. The reaction mixture was filtered through Celite® and the filtrate concentrated in vacuo. The resulting residue was dissolved in THF (1 mL) and H2O (1 mL), K2CO3 (20 mg, 0.1400 mmol, 2.0 eq.) and allyl chloroformate (14 pL, 0.1400 mmol, 2.0 eq.) were added. The mixture was stirred for 2 h, whereupon LCMS indicated the reaction was complete. The reaction mixture was diluted (EtOAc), washed with 1 N HCI solution, dried over MgS0 4 and concentrated in vacuo. Flash column chromatography (70-100% EtOAc in hexane, 0.1% AcOH) afforded the desired product (67 mg, 68%). LCMS-A: 2.13 min (ES+) m/z 1414 [M + H] + , 1436 [M + Na] +

(iv-a) Ally! (1 1 S.11 aSV8-((3-((((11 S.11 aSV10-(((4-((SV2-((SV2-(((allyloxy)carbonvnamino 3- methylbutanamido)propanamido)benzyl)oxy)carbonyl)-1 1-hvdroxy-7-methoxy-2-methyl-5- oxo-5,10,11 ,1 1a-tetrahvdro-1 H-benzofelpyrroloH ,2-alH ,4ldiazepin-8-yl)oxy)methyl)-5- (methoxycarbonyl)benzyl)oxy)-11 -hvdroxy-7-methoxy-2-methyl-5-oxo-1 1 ,11 a-dihvdro-1 H- benzofelpyrroloH ,2-alH ,4ldiazepine-10(5H)-carboxylate (31 )

A round-bottomed flask was charged with a magnetic stirrer, THF (10 ml_), 29 (200 mg, 0.1400 mmol, 1.0 eq.), AcOH (32 mI_, 0.5600 mmol, 4.0 eq.) and TBAF (560 mI_ of a 1 M solution, 0.5600 mmol, 4.0 eq.) were added and the reaction was stirred for 16 h with gradual warming to rt, whereupon LCMS indicated the reaction was complete. The reaction mixture was diluted with CH2CI2, washed with sat. NH 4 CI, dried over MgS0 4 and

concentrated in vacuo. Flash column chromatography (3% MeOH in CHCI3) afforded the desired product (140 mg, 83%). LCMS-A: 1.62 min (ES+) m/z 1200 [M + H] + , 1222 [M +

Na] +

(iv-b) 3-((((11 S, 11 aS)-10-((allyloxy)carbonyl)-1 1 -hvdroxy-7-methoxy-2-methyl-5-oxo- 5,10,1 1 ,11 a-tetrahydro-1 H-benzofelpyrrolof 1 ,2-alf 1 ,4ldiazepin-8-yl)oxy)methyl)-5- ((((1 1S.11 aSV10-(((4-((SV2-((SV2-(((allyloxy)carbonvnaminoV3- methylbutanamido)propanamido)benzyl)oxy)carbonyl)-1 1-hvdroxy-7-methoxy-2-methyl-5- oxo-5,10,11 ,1 1a-tetrahvdro-1 H-benzofelpyrrolof 1 ,2-alf 1 ,4ldiazepin-8-yl)oxy)methyl)benzoic acid (32)

A round-bottomed flask was charged with a magnetic stirrer, THF (10 mL), 30 (135 mg, 0.09542 mmol, 1.0 eq.), cooled to 0 °C and stirred for 10 minutes. AcOH (21.9 pL, 0.3817 mmol, 4.0 eq.) and TBAF (429 pL of a 1 M solution, 0.4290 mmol, 4.0 eq.) were added and the reaction was stirred for 16 h with gradual warming to rt, whereupon LCMS indicated the reaction was complete. The reaction mixture was diluted with CH2CI2, washed with sat. NH4CI, dried over MgS0 4 and concentrated in vacuo. Flash column chromatography (0-5% MeOH in CH2CI2) afforded the product contaminated with Bu 4 N + salts. The resulting product was used without further purification and the yield was assumed to be 100%. LCMS-A: 1.63 min (ES+) m/z 1 186 [M + H] + , 1208 [M + Na] +

(v-a) 4-((S)-2-((S)-2-(3-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)propanamido)-3- methylbutanamido)propanamido)benzyl (1 1 S,11 aS)-1 1-hydroxy-7-methoxy-8-((3-((((S)-7- methoxy-2-methyl-5-oxo-5,1 1 a-dihvdro-1 H-benzofelpyrrolof 1 , 2-alf 1 ,4ldiazepin-8- yl)oxy)methyl)-5-(methoxycarbonyl)benzyl)oxy)-2-methyl-5-oxo -11 ,1 la-dihvdro-1 H- benzorelpyrrolori ,2-aln ,4ldiazepine-10(5l-0-carboxylate (33)

A round-bottomed flask was charged with a magnetic stirrer, CH2CI2 (10 ml_), 31 (120 mg, 99.98 pmol, 1.0 eq.), pyrrolidine (21 mI_, 250 pmol, 2.5 eq.) and Pd(PPh3)4 (5.8 mg, 4.999 pmol, 0.050 eq.). The mixture was stirred under Ar for 30 min, whereupon LCMS indicated the reaction was complete. The reaction mixture was thoroughly concentrated in vacuo overnight to remove pyrrolidine traces. The resulting residue was dissolved in CH2CI2 (3.5 ml.) and maleimide propionic acid (14.9 mg, 88.31 pmol, 1.0 eq.) and EDCI.HCI (13.7 mg, 88.31 pmol, 1.0 eq.) were added and the mixture stirred for 2 h, whereupon LCMS indicated the reaction was complete. The reaction mixture was concentrated in vacuo and purified by flash column chromatography (3-5% MeOH in CH2CI2) then preparative HPLC (15-90% A in B) to afford the desired product as a yellow solid (10 mg, 9.7% yield). A = 0.05% formic acid in acetonitrile. B = 0.05% formic acid in water. LCMS-B: 6.78 min (ES+) m/z 1 165 [M + H] + , 1 187 [M + Na] +

(v-b) 3-((((11 S.1 1aSV10-(((4-((SV2-((SV2-(3-(2.5-dioxo-2.5-dihvdro-1 H-pyrrol-1- yl)propanamido)-3-methylbutanamido)propanamido)benzyl)oxy)ca rbonyl)-1 1 -hydroxy-7- methoxy-2-methyl-5-oxo-5,10,11 ,1 1a-tetrahvdro-1 H-benzolelpyrrolori ,2-alH ,4ldiazepin-8- yl)oxy)methyl)-5-((((S)-7-methoxy-2-methyl-5-oxo-5,11 a-dihvdro-1 H-benzorelpyrrolon ,2- alH ,4ldiazepin-8-yl)oxy)methyl)benzoic acid (34)

A round-bottomed flask was charged with a magnetic stirrer, CH 2 CI 2 (8 mL) 32 (crude from previous step ca. 95.42 pmol, 1.0 eq.) pyrrolidine (19.6 pL, 238.6 pmol, 2.5 eq.), Pd(PPh3) 4 (5.5 mg, 4.771 pmol, 0.050 eq.) and the mixture stirred for 30 min under Ar, whereupon LCMS indicated the reaction was complete. The reaction mixture was diluted with CHCI 3 , washed with sat. NH 4 CI, the organics combined, dried over MgS0 4 and concentrated in vacuo. The resulting residue was dissolved in THF (7 mL) and NaHCC> 3 (1 mL of a 0.1 M solution) and maleimide propionic acid NHS ester (28 mg, 105 pmol, 1.1 eq.) were added. The mixture was stirred for 20 h, diluted with CHCb, washed with sat. NH 4 CI solution and concentrated in vacuo. Preparative HPLC (15-70% A in B) afforded the desired product as a yellow solid (6.4 mg, 5.8% yield, 84.6% QC). A = 0.05% formic acid in acetonitrile. B = 0.05% formic acid in water. LCMS-B: 6.52 min (ES+) m/z 1 151 [M + H] + Example 4: 4-((S)-2-((S)-2-(3-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)propanamido)-3- methylbutanamido)propanamido)benzyl (11 S,11aS)-11 -hydroxy-7-methoxy-8-((6-((((S)- 7-methoxy-2-methylene-5-oxo-2,3,5,11 a-tetrahydro-1 H-benzo[e]pyrrolo[1 ,2- a][1 ,4]diazepin-8-yl)oxy)methyl)-4-(methoxycarbonyl)pyridin-2-yl )methoxy)-2- methylene-5-oxo-2,3,11 ,11 a-tetrahydro-1 H-benzo[e]pyrrolo[1 ,2-a][1 ,4]diazepine- 10(5H)-carboxylate (44) and 2-((((11 S,11 aS)-10-(((4-((S)-2-((S)-2-(3-(2,5-dioxo-2,5- dihydro-1 H-pyrrol-1 -yl)propanamido)-3- methylbutanamido)propanamido)benzyl)oxy)carbonyl)-11 -hydroxy-7-methoxy-2- methylene-5-oxo-2,3,5,10,11 ,11a-hexahydro-1 H-benzo[e]pyrrolo[1 ,2-a][1 ,4]diazepin-8- yl)oxy)methyl)-6-((((S)-7-methoxy-2-methylene-5-oxo-2,3,5, 11 a-tetrahydro-1 H- benzo[e]pyrrolo[1 ,2-a][1 ,4]diazepin-8-yl)oxy)methyl)isonicotinic acid (45)

(i) Methyl 2,6-bis(bromomethyl)/sonicotinate (36)

A round-bottomed flask was charged with a magnetic stirrer bar, acetonitrile (46 ml_), 35 (924 mg, 4.686 mmol, 1.0 eq.) and the mixture cooled to 0 °C. PPhi 3 (3.96 g, 14.06 mmol,

3.0 eq.) then slowly CBr 4 (4.66 g, 14.06 mmol, 3.0 eq.) were added and the mixture stirred or 30 min. The reaction mixture was concentrated in vacuo and purified by isolera (0-100% EtOAc in hexane) to afford the desired product (1.01 g, 67%). LCMS-A: 1.46 min (ES+) m/z 321 [M + H] +

(ii) Methyl 2-((5-((((4-((SV2-((SV2-(((allyloxy)carbonvnaminoV3- methylbutanamido)propanamido)benzyl)oxy)carbonyl)amino)-4-(( S)-2-(((te/f- butyldimethylsilyl)oxy)methyl)-4-methylenepyrrolidine-1-carb onyl)-2- methoxyphenoxy)methyl)-6-(bromomethyl)isonicotinate (37)

A round-bottomed flask was charged with a magnetic stirrer bar, DMF (10 ml_), 13 (814 mg, 1.022 mmol, 1.0 eq.), 36 (1 ,32 g, 4.087 mmol, 4.0 eq.) and K2CO3 (155 mg, 1.124 mmol, 1.1 eq.) and stirred for 40 min at 35 °C, whereupon LCMS indicated the reaction was complete. DMF was removed in vacuo and the resulting residue was dissolved in EtOAc, washed with H2O, brine and dried over MgS0 4 . Isolera (50-100% EtOAc in hexane) afforded the desired product as a pale yellow foam (750 mg, 71%). LCMS-A: 1.90 min (ES+) m/z 1037 [M + H] +

(iii) te/f-Butyl (11 S.11 aSV8-((6-((5-((((4-((SV2-((S)-2-(((allyloxy)carbonvnaminoV3- methylbutanamido)propanamido)benzyl)oxy)carbonyl)amino)-4-(( S)-2-(((te/f- butyldimethylsilyl)oxy)methyl)-4-methylenepyrrolidine-1-carb onyl)-2- methoxyphenoxy)methyl)-4-(methoxycarbonyl)pyridin-2-yl)metho xy)-7-methoxy-2- methylene-5-oxo-1 1-((tetrahvdro-2H-pyran-2-yl)oxy)-2,3,11 ,11 a-tetrahvdro-1 H- benzorelpyrrolori ,2-aln ,4ldiazepine-10(5H)-carboxylate (38)

A round-bottomed flask was charged with a magnetic stirrer bar, DMF (6 mL), 37 (750 mg, 0.7225 mmol, 1.0 eq.), I2 (333 mg, 0.7225 mmol, 1.0 eq.) and K 2 CO 3 (100 mg, 0.7235 mmol, 1.0 eq.) and stirred for 80 min at 35 °C, whereupon LCMS indicated the reaction was complete. DMF was removed in vacuo and the resulting residue was dissolved in EtOAc, washed with H2O and dried over MgS0 4 . In vacuo concentration afforded the desired product as a pale-yellow foam (979 mg, 96%). LCMS-A: 1.99 min (ES+) m/z 1417 [M + H] +

(iv) tert- Butyl (1 1 S.1 1aSV8-((6-((5-((((4-((SV2-((S)-2-(((allyloxy)carbonvnaminoV3 - methylbutanamido)propanamido)benzyl)oxy)carbonyl)amino)-4-(( S)-2-(hvdroxymethyl)-4- methylenepyrrolidine-1-carbonyl)-2-methoxyphenoxy)methyl)-4- (methoxycarbonyl)pyridin-2- yl)methoxy)-7-methoxy-2-methylene-5-oxo-1 1 -((tetrahvdro-2H-pyran-2-yl)oxy)-2,3,1 1 ,1 1 a- tetrahydro-1 H-benzorelpyrroloH , 2-alH ,4ldiazepine-10(5H)-carboxylate (39)

A round-bottomed flask was charged with a magnetic stirrer bar, THF (9 ml.) 38 (879 mg, 0.6200 mmol, 1.0 eq.) TBAF (1 .24 mL of a 1 M solution in THF, 1.240 mmol, 2.0 eq.) and the mixture stirred for 80 min whereupon LCMS indicated the reaction was complete. The reaction mixture was concentrated in vacuo and purified by isolera (0-4% MeOH in CH2CI2) to afford the desired product (746 mg, 83%). LCMS-A: 1 .71 min (ES+) m/z 1303 [M + H] + , 1325 [M + Na] +

(v) 4-((S)-2-((S)-2-(((allyloxy)carbonyl)amino)-3-methylbutanami do)propanamido)benzyl

(1 1 S,1 1 aS)-8-((6-((((1 1 S,1 1 aS)-10-(te/f-butoxycarbonyl)-7-methoxy-2-methylene-5-oxo-1 1 - ((tetrahvdro-2H-pyran-2-yl)oxy)-2,3,5,10,1 1 ,1 1 a-hexahvdro-1 H-benzorelpyrrolon ,2- alH ,4ldiazepin-8-yl)oxy)methyl)-4-(methoxycarbonyl)pyridin-2-yl )methoxy)-1 1 -hvdroxy-7- methoxy-2-methylene-5-oxo-2,3,1 1 ,1 1 a-tetrahvdro-1 H-benzorelpyrroloH ,2-alH ,4ldiazepine- 10(5H)-carboxylate (40)

A round-bottomed flask was charged with a magnetic stirrer bar, acetonitrile (13 mL), 39 (64 mg, 0.4956 mmol, 1 .0 eq.), Stahl aerobic oxidation TEMPO solution (248 pL of a 0.2M solution in acetonitrile, 0.04960 mmol, 0.10 eq.), Cu(MeCN) 4 OTf (18.7 mg, 0.04956 mmol, 0.10 eq.) and the mixture stirred under a balloon of air for 3 d. The reaction mixture was concentrated in vacuo and purified by isolera (0-4% MeOH in CH2CI2) to afford the desired product as a white solid (523 mg, 81 %). LCMS-A: 1 .69 min (ES+) m/z 1301 [M + H] + , 1323 [M + Na] +

(vi) 2-((((1 1 S.1 1 aSV10-(((4-((S)-2-((S)-2-(((allyloxy)carbonvnamino)-3- methylbutanamido)propanamido)benzyl)oxy)carbonyl)-1 1 -hydroxy-7-methoxy-2-methylene- 5-oxo-2,3,5,10,1 1 ,1 1 a-hexahvdro-1 H-benzorelpyrroloH ,2-alri ,4ldiazepin-8-yl)oxy)methyl)-6- ((((1 1 S, 1 1 aS)-10-(te/f-butoxycarbonyl)-7-methoxy-2-methylene-5-oxo-1 1 -((tetrahydro-2H- Pyran-2-yl)oxy)-2,3,5,10,1 1 ,1 1 a-hexahvdro-1 H-benzorelpyrroloH , 2-alH ,4ldiazepin-8- yl)oxy)methyl)/sonicotinic acid (41 )

A microwave vial was charged with a magnetic stirrer bar, (CH2CI)2 (0.5 mL) 40 (100 mg, 64.62 pmol, 1.0 eq.), SnMesOH (35.1 mg, 193.9 pmol, 3.0 eq.) and the vial sealed. The mixture was heated to 80 °C for 4 h, cooled to rt, poured into H2O, extracted with 10%

MeOH in CH2CI2, the organics combined, dried over Na 2 S0 4 and concentrated in vacuo. Isolera (5-15% MeOH in CH2CI2) afforded the desired product (53 mg, 64%). LCMS-A: min (ES+) m/z 1287 [M + H] + , 1309 [M + Na] + (vii-a) te/f-Butyl (1 1 S.1 1aSV8-((6-((((11 S.1 1aSV10-(((4-((SV2-((SV2-(3-(2.5-dioxo-2.5- dihydro-1 H-pyrrol-1 -yl)propanamido)-3- methylbutanamido)propanamido)benzyl)oxy)carbonyl)-1 1-hvdroxy-7-methoxy-2-methylene- 5-oxo-2,3,5,10,1 1 ,1 1a-hexahvdro-1 H-benzorelpyrrolori ,2-alH ,4ldiazepin-8-yl)oxy)methyl)-4- (methoxycarbonyl)pyridin-2-yl)methoxy)-7-methoxy-2-methylene -5-oxo-11-((tetrahydro-2H- pyran-2-yl)oxy)-2,3,1 1 ,1 1a-tetrahydro-1 H-benzorelpyrroloH ,2-alH ,4ldiazepine-10(5H)- carboxylate (42)

A round-bottomed flask was charged with a magnetic stirrer bar, 40 (100 mg, 76.84 pmol,

1.0 eq.) CH2CI2 (6 ml_), pyrrolidine (12,6 mI_, 153.7 pmol, 2.0 eq.) and Pd(PPh3)4 (2.2 mg, 1.921 pmol, 0.025 eq.). The mixture was stirred under Ar for 25 min, whereupon LCMS indicated the reaction was complete. The reaction mixture was thoroughly concentrated in vacuo overnight to remove pyrrolidine traces. The resulting residue was dissolved in CH2CI2 (1 ml.) and maleimide propionic acid NHS ester (24.5 mg, 92.21 pmol, 1.2 eq.) and / ' -P^NEt (16.1 mI_, 92.21 pmol, 1.2 eq.) were added and the mixture stirred for 3.5 h, whereupon LCMS indicated the reaction was complete. The reaction mixture was concentrated in vacuo and purified by isolera (0-4% MeOH in CH2CI2) to afford the desired product as a white solid (89.5 mg, 85%). LCMS-A: 1.62 min (ES+) m/z 1368 [M + H] + , 1390 [M + Na] +

(vii-b) 2-((((11 S, 11 aS)-10-(te/f-butoxycarbonyl)-7-methoxy-2-methylene-5-oxo-1 1 - ((tetrahvdro-2H-pyran-2-yl)oxy)-2,3,5,10,1 1 ,1 1a-hexahvdro-1 H-benzoielpyrroioi1 ,2- alri .4ldiazepin-8- methvn-6-((((11 S.1 1aSV10-(((4-((SV2-((SV2-(3-(2.5-dioxo-2.5-

dihydro-1 H-pyrrol-1 -yl)propanamido)-3- methylbutanamido)propanamido)benzyl)oxy)carbonyl)-1 1-hydroxy-7-methoxy-2-methylene- 5-oxo-2,3,5,10,11 ,1 1a-hexahydro-1 H-benzorelpyrroloH ,2-alH ,4ldiazepin-8- yl)oxy)methyl)/sonicotinic acid (43)

A round-bottomed flask was charged with a magnetic stirrer bar, CH 2 CI 2 (3 mL), 41 (53 mg, 41.17 pmol, 1.0 eq.), pyrrolidine (10.1 pmol, 122.8 mmol, 3.0 eq.) Pd(PPh3) 4 (1.2 mg, 1.024 pmol, 0.025 eq.) and the mixture stirred under Ar for 30 min whereupon LCMS indicated the reaction was complete. The reaction mixture was diluted with CH 2 CI 2 , washed with NH 4 CI, dried over Na 2 S0 4 and concentrated in vacuo. The reaction mixture was thoroughly concentrated in vacuo overnight to remove pyrrolidine traces. The resulting residue was dissolved in THF (3 mL) and maleimide propionic acid NHS ester (13.1 mg, 49.13 pmol, 1.2 eq.) and NaHCCh (0.43 mL of a 100 mM solution) were added and the mixture stirred for 5 h, whereupon LCMS indicated the reaction was complete. The reaction mixture was diluted with EtOAc, washed with NH 4 CI, dried over MgSCU and concentrated in vacuo. Isolera (2- 10% MeOH in CH2CI2) afforded the desired product (39.9 mg, 72% yield). LCMS-A: 1.58 min (ES+) m/z 1354 [M + H] + , 1376 [M + Na] +

(viii-a) 4-((S)-2-((S)-2-(3-(2,5-dioxo-2,5-dihydro-1 H-pyrrol-1 -yl)propanamido)-3- methylbutanamido)propanamido)benzyl (1 1S,11 aS)-1 1-hydroxy-7-methoxy-8-((6-((((S)-7- methoxy-2-methylene-5-oxo-2,3,5,11 a-tetrahvdro-1 H-benzorelpyrrolori ,2-aln ,4ldiazepin-8- yl)oxy)methyl)-4-(methoxycarbonyl)pyridin-2-yl)methoxy)-2-me thylene-5-oxo-2,3,1 1 ,1 1a- tetrahydro-1 H-benzorelpyrrolori ,2-aln ,4ldiazepine-10(5H)-carboxylate (44)

A round-bottomed flask was charged with a magnetic stirrer bar, 42 (90 mg, 65.77 pmol, 1.0 eq.) and cooled to 0 °C. 95% TFA in H2O (3.5 ml.) was cooled to -20 °C and added in one portion. The reaction mixture was stirred for 30 min, whereupon LCMS indicated the reaction was complete. The reaction mixture was poured into H2O (10 ml_), the pH adjusted to ca. 5 with NaHCCh and the aqueous mixture extracted with 10% MeOH in CH2CI2, the organics dried over Na 2 S0 4 and concentrated in vacuo. Isolera purification (0-8% MeOH in CH2CI2) then preparative HPLC (15-70% A in B) afforded the desired product as a white solid (7 mg, 9.2% yield, 98.4% QC). A = 0.05% formic acid in acetonitrile. B = 0.05% formic acid in water. LCMS-B: 7.61 min (ES+) m/z 1 1.66 [M + H] +

(viii-b) 2-((((11 S.11 aSI-10-(((4-((SV2-((SV2-(3-(2.5-dioxo-2.5-dihvdro-1 H-pyrrol-1 - yl)propanamido)-3-methylbutanamido)propanamido)benzyl)oxy)ca rbonyl)-1 1 -hydroxy-7- methoxy-2-methylene-5-oxo-2,3,5,10,1 1 ,1 1a-hexahvdro-1 H-benzorelpyrrolon ,2- aH1 , 4ldiazepin-8-yl)oxy)methyl)-6-((((S)-7-methoxy-2-methylene-5 -oxo-2, 3,5,11 a-tetrahvdro- 1 H-benzofelpyrroloH ,2-alH ,41diazepin-8-yl)oxy)methyl)/sonicotinic acid (45)

A round-bottomed flask was charged with a magnetic stirrer bar, 43 (37 mg, 27.32 pmol, 1.0 eq.) and cooled to 0 °C. 95% TFA in H2O (2 ml.) was cooled to -20 °C and added in one portion. The reaction mixture was stirred for 30 min, whereupon LCMS indicated the reaction was complete. The reaction mixture was poured into H2O (10 mL) and pH adjusted to ca. 5 with NaHCC>3. The solution was loaded onto an SPE cartridge (SCX-2, 10 g), washed with H2O (2 x 10 mL) then blown dry with air. The organics were eluted from the column with MeOH and concentrated in vacuo. Preparative HPLC (15-70% A in B) afforded the desired product as an off-white solid (5 mg, 16% yield, 92.5% QC). A = 0.05% formic acid in acetonitrile. B = 0.05% formic acid in water. LCMS-B: 6.93 min (ES+) m/z 1 152 Example 5 - Conjugations

Antibody-drug conjugate with Compound 19, ConjA

A 50 mM solution of DL-dithiothreitol (DTT) in phosphate-buffered saline pH 7.4 (PBS) was added (40 molar equivalent/antibody, 12 micromoles, 240 mI_) to a 7.5 ml. solution of antibody Trastuzumab (45 mg, 300 nanomoles) in reduction buffer containing PBS and 1 mM ethylenediaminetetraacetic acid (EDTA) and a final antibody concentration of 4.0 mg/ml_.

The reduction mixture was allowed to react at room temperature for 17 hours (or until full reduction is observed by UHPLC) in an orbital shaker with gentle (60 rpm) shaking. The reduced antibody was buffer exchanged, via spin filter centrifugation, into a reoxidation buffer containing PBS and 1 mM EDTA to remove all the excess reducing agent. A 50 mM solution of dehydroascorbic acid (DHAA, 15 molar equivalent/antibody, 4.5 micromoles, 90 pl_) in DMSO was added and the reoxidation mixture was allowed to react for 2-3 hours at room temperature with gentle (50 rpm) shaking at an antibody concentration of 1.5 mg/ml_

(or more DHAA added and reaction left for longer until full reoxidation of the cysteine thiols to reform the inter-chain cysteine disulfides is observed by UHPLC). The reoxidation mixture was then sterile-filtered and diluted in a conjugation buffer containing PBS and 1 mM EDTA for a final antibody concentration of 2.0 mg/mL. Compound 19 was added as a DMSO solution (10 molar equivalent/antibody, 0.9 micromole, in 1.0 mL DMSO) to 10.0 mL of this reoxidised antibody solution (13.5 mg, 90 nanomoles) for a 10% ( v/v ) final DMSO

concentration. The solution left to react at room temperature for 2 hours at room temperature with gentle shaking. Then the conjugation was quenched by addition of N- acetyl cysteine (4.5 micromoles, 45 pL at 100 mM), then purified by spin filtration using a 15 mL Amicon Ultracell 50 kDa MW CO spin filter, sterile-filtered and analysed.

UHPLC analysis on a Shimadzu Prominence system using a Thermo Scientific MAbPac 50 mm x 2.1 mm column eluting with a gradient of water and acetonitrile on a reduced sample of ConjA at 280 nm and 330 nm (Compound 19 specific) shows unconjugated light chains and a mixture of unconjugated heavy chains and heavy chains attached to a single molecule of Compound 19, consistent with a drug-per-antibody ratio (DAR) of 1.89 molecules of Compound 19 per antibody.

UHPLC analysis on a Shimadzu Prominence system using a Tosoh Bioscience TSKgel SuperSW mAb HTP 4 pm 4.6 x 150 mm column (with a 4 pm 3.0 x 20 mm guard column) eluting with 0.3 mL/minute sterile-filtered SEC buffer containing 200 mM potassium phosphate pH 6.95, 250 mM potassium chloride and 10% isopropanol (v/v) on a sample of ConjA at 280 nm shows a monomer purity of 96%. UHPLC SEC analysis gives a

concentration of final ConjA at 1.39 mg/ml_ in 8.0 ml_, obtained mass of ConjA is 11.15 mg (74% yield).

Antibody-drug conjugate with Compound 27, ConjB

A 50 mM solution of DL-dithiothreitol (DTT) in phosphate-buffered saline pH 7.4 (PBS) was added (40 molar equivalent/antibody, 27 pmol, 536 pl_) to a 25 ml. solution of antibody Trastuzumab (100 mg, 0.67 micromoles) in reduction buffer containing PBS and 1 mM ethylenediaminetetraacetic acid (EDTA) and a final antibody concentration of 4.0 mg/mL.

The reduction mixture was allowed to react at room temperature for 17 hours (or until full reduction is observed by UHPLC) in an orbital shaker with gentle (60 rpm) shaking. After cooling down to room temperature, the reduced antibody was buffer exchanged, via

Tangential Flow Filtration unit (TFF) using mPES, MidiKros ® 30 kDa fiber filter with 115 cm 2 surface area, into a reoxidation buffer containing PBS pH 7.4 and 1 mM EDTA to remove all the excess reducing agent. The reduced antibody was centrifuged for 3 min at 4000 rpm and then filtered using 0.22 mM membrane filter. A 50 mM solution of dehydroascorbic acid (DHAA, 20 molar equivalent/antibody, 12 micromoles, 240 pL) in DMSO was added and the reoxidation mixture was allowed to react for 2-3 hours at room temperature with gentle (50 rpm) shaking at an antibody concentration of ~ 1.7 mg/mL (or more DHAA added and reaction left for longer until full reoxidation of the cysteine thiols to reform the inter-chain cysteine disulfides is observed by UHPLC). The reoxidation mixture was then sterile-filtered and diluted in a conjugation buffer containing PBS and 1 mM EDTA for a final antibody concentration of 1.7 mg/mL. Compound 27 was added as a DMSO solution (12 molar equivalent/antibody, 1.2 micromole, in 1.5 mL DMSO) to 15.0 mL of this reoxidised antibody solution (15.3 mg, 0.1 pmoles) for a 10% (v/v) final DMSO concentration. The solution left to react at room temperature for 2 hours at room temperature with gentle shaking. Then the conjugation was quenched by addition of N- acetyl cysteine (6.0 micromoles, 60 pL at 100 mM), then purified by spin filtration using a 15 mL Amicon Ultracell 50 kDa MWCO spin filter, sterile-filtered and analysed.

UHPLC analysis on a Shimadzu Prominence system using a Thermo Scientific MAbPac 50 mm x 2.1 mm column eluting with a gradient of water and acetonitrile on a reduced sample of ConjB at 280 nm and 330 nm (Compound 27 specific) shows unconjugated light chains and a mixture of unconjugated heavy chains and heavy chains attached to a single molecule of Compound 27, consistent with a drug-per-antibody ratio (DAR) of 1.69 molecules of Compound 27 per antibody.

UHPLC analysis on a Shimadzu Prominence system using a Tosoh Bioscience TSKgel SuperSW mAb HTP 4 pm 4.6 x 150 mm column (with a 4 pm 3.0 x 20 mm guard column) eluting with 0.3 mL/minute sterile-filtered SEC buffer containing 200 mM potassium phosphate pH 6.95, 250 mM potassium chloride and 10% isopropanol (v/v) on a sample of ConjB at 280 nm shows a monomer purity of 99%. UHPLC SEC analysis gives a

concentration of final ConjB at 2.31 mg/mL in 5.8 mL, obtained mass of ConjB is 13.4 mg (67% yield).

Antibody-drug conjugate with Compound 33, ConjC

A 50 mM solution of DL-dithiothreitol (DTT) in phosphate-buffered saline pH 7.4 (PBS) was added (125 molar equivalent/antibody, 25 micromoles, 500 pL) to a 7.5 mL solution of antibody (30 mg, 200 nanomoles) in reduction buffer containing PBS and 1 mM

ethylenediaminetetraacetic acid (EDTA) and a final antibody concentration of 4.0 mg/mL.

The reduction mixture was allowed to react at room temperature for 3-4 hours (or until full reduction is observed by UHPLC) in an orbital shaker with gentle (60 rpm) shaking. The reduced antibody was buffer exchanged, via spin filter centrifugation, into a reoxidation buffer containing PBS and 1 mM EDTA to remove all the excess reducing agent. A 50 mM solution of dehydroascorbic acid (DHAA, 20 molar equivalent/antibody, 4 micromoles, 80 pL) in DMSO was added and the reoxidation mixture was allowed to react for 16 hours at room temperature with gentle (50 rpm) shaking at an antibody concentration of 2.5 mg/mL (or more DHAA added and reaction left for longer until full reoxidation of the cysteine thiols to reform the inter-chain cysteine disulfides is observed by UHPLC). The reoxidation mixture was then sterile-filtered and diluted in a conjugation buffer containing PBS and 1 mM EDTA for a final antibody concentration of 2.0 mg/mL. Compound 33 was added as a DMSO solution (10 molar equivalent/antibody, 1 micromole, in 0.75 mL DMSO) to 6.75 mL of this reoxidised antibody solution (15 mg, 100 nanomoles) for a 10% (v/v) final DMSO

concentration. The solution was mixed for 4 hours at room temperature (0.5-1.0 mL propylene glycol added after 2 hours to aid solubility), then the conjugation was quenched by addition of N- acetyl cysteine (5 micromoles, 50 pL at 100 mM), then purified by spin filtration using a 15 mL Amicon Ultracell 50 kDa MWCO spin filter, sterile-filtered and analysed. UHPLC analysis on a Shimadzu Prominence system using a Thermo Scientific MAbPac 50 mm x 2.1 mm column eluting with a gradient of water and acetonitrile on a reduced sample of ConjC at 280 nm and 330 nm (Compound 33 specific) shows unconjugated light chains and a mixture of unconjugated heavy chains and heavy chains attached to a single molecule of Compound 33, consistent with a drug-per-antibody ratio (DAR) of 1.63 molecules of Compound 33 per antibody.

UHPLC analysis on a Shimadzu Prominence system using a Tosoh Bioscience TSKgel SuperSW mAb HTP 4 pm 4.6 x 150 mm column (with a 4 pm 3.0 x 20 mm guard column) eluting with 0.3 mL/minute sterile-filtered SEC buffer containing 200 mM potassium phosphate pH 6.95, 250 mM potassium chloride and 10% isopropanol ( v/v ) on a sample of ConjC at 280 nm shows a monomer purity of 99%. UHPLC SEC analysis gives a concentration of final ConjC at 1.89 mg/mL in 7.0 mL, obtained mass of ConjC is 13.2 mg (88% yield).

Antibody-drug conjugate with Compound 34, ConjD

A 50 mM solution of DL-dithiothreitol (DTT) in phosphate-buffered saline pH 7.4 (PBS) was added (40 molar equivalent/antibody, 28 micromoles, 560 pL) to a 26.25 mL solution of antibody Trastuzumab (105 mg, 700 nanomoles) in reduction buffer containing PBS and 1 mM ethylenediaminetetraacetic acid (EDTA) and a final antibody concentration of 4.0 mg/mL.

The reduction mixture was allowed to react at room temperature for 20 hours (or until full reduction is observed by UHPLC) in an orbital shaker with gentle (60 rpm) shaking. The reduced antibody was buffer exchanged, via tangential flow filtration (TFF) using a Spectrum Labs KrosFlo Research Hi system with a 30 kDa MWCO, 115 cm 2 surface area hollow fibre filter module at 50 mL/min in PBS with a transmembrane pressure (TMP) of 0.5-1.0 bar, into a reoxidation buffer containing PBS and 1 mM EDTA to remove all the excess reducing agent, recovery 97 mg (667 nanomoles). A 50 mM solution of dehydroascorbic acid (DHAA, 20 molar equivalent/antibody, 12.9 micromoles, 259 pL) in DMSO was added and the reoxidation mixture was allowed to react for 17 hours at room temperature with gentle (50 rpm) shaking at an antibody concentration of 1.5 mg/mL (or more DHAA added and reaction left for longer until full reoxidation of the cysteine thiols to reform the inter-chain cysteine disulfides is observed by UHPLC). The reoxidation mixture was then sterile-filtered and diluted in a conjugation buffer containing PBS and 1 mM EDTA for a final antibody concentration of 1.0 mg/mL. Compound 34 was added as a DMSO solution (15 molar equivalent/antibody, 1.4 micromoles, in 1.4 mL DMSO) to 12.6 mL of this reoxidised antibody solution (14 mg, 93 nanomoles) for a 10% (v/v) final DMSO concentration. The solution was mixed for 2 hours at room temperature, stored at +4 °C for 16 hours then the conjugation was quenched by addition of N- acetyl cysteine (3.7 micromoles, 37 mI_ at 100 mM). Reaction mixture was purified by spin filtration using a 15 mL Amicon Ultracell 50 kDa MWCO spin filter, further purified by TFF (800 mL permeated), sterile-filtered and analysed. UHPLC analysis on a Shimadzu Prominence system using a Phenomenex Aeris 3.6u XB- C18 150 mm x 2.1 mm column eluting with a gradient of water and acetonitrile on a reduced sample of ConjD at 280 nm and 330 nm (Compound 34 specific) shows unconjugated light chains and a mixture of unconjugated heavy chains and heavy chains attached to a single molecule of Compound 34, consistent with a drug-per-antibody ratio (DAR) of 1.81 molecules of Compound 34 per antibody.

UHPLC analysis on a Shimadzu Prominence system using a Tosoh Bioscience TSKgel SuperSW mAb HTP 4 pm 4.6 x 150 mm column (with a 4 pm 3.0 x 20 mm guard column) eluting with 0.3 mL/minute sterile-filtered SEC buffer containing 200 mM potassium phosphate pH 6.95, 250 mM potassium chloride and 10% isopropanol (v/v) on a sample of ConjD at 280 nm shows a monomer purity of 97%. UHPLC SEC analysis gives a

concentration of final ConjD at 0.49 mg/mL in 5.5 mL, obtained mass of ConjD is 2.59 mg (19% yield).

Antibody-drug conjugate with Compound 44, ConjE

A 50 mM solution of DL-dithiothreitol (DTT) in phosphate-buffered saline pH 7.4 (PBS) was added Trastuzumab (40 molar equivalent/antibody, 27 pmol, 536 pL) to a 25 mL solution of antibody (100 mg, 0.67 micromoles) in reduction buffer containing PBS and 1 mM ethylenediaminetetraacetic acid (EDTA) and a final antibody concentration of 4.0 mg/mL.

The reduction mixture was allowed to react at room temperature for 17 hours (or until full reduction is observed by UHPLC) in an orbital shaker with gentle (60 rpm) shaking. After cooling down to room temperature, the reduced antibody was buffer exchanged, via

Tangential Flow Filtration unit (TFF) using mPES, MidiKros ® 30 kDa fiber filter with 115 cm 2 surface area, into a reoxidation buffer containing PBS pH 7.4 and 1 mM EDTA to remove all the excess reducing agent. The reduced antibody was centrifuged for 3 min at 4000 rpm and then filtered using 0.22 mM membrane filter. A 50 mM solution of dehydroascorbic acid (DHAA, 20 molar equivalent/antibody, 12 micromoles, 240 pL) in DMSO was added and the reoxidation mixture was allowed to react for 2-3 hours at room temperature with gentle (50 rpm) shaking at an antibody concentration of ~ 1.7 mg/ml_ (or more DHAA added and reaction left for longer until full reoxidation of the cysteine thiols to reform the inter-chain cysteine disulfides is observed by UHPLC). The reoxidation mixture was then sterile-filtered and diluted in a conjugation buffer containing PBS and 1 mM EDTA for a final antibody concentration of 1.7 mg/mL Compound 44 was added as a DMSO solution (10 molar equivalent/antibody, 1.0 micromole, in 1.5 mL DMSO) to 15.0 ml. of this reoxidised antibody solution (15.3 mg, 0.1 pmoles) for a 10% ( v/v ) final DMSO concentration. The solution left to react at room temperature for 2 hours at room temperature with gentle shaking. Then the conjugation was quenched by addition of N- acetyl cysteine (5.0 micromoles, 50 mI_ at 100 mM), then purified by spin filtration using a 15 mL Amicon Ultracell 50 kDa MWCO spin filter, sterile-filtered and analysed.

UHPLC analysis on a Shimadzu Prominence system using a Thermo Scientific MAbPac 50 mm x 2.1 mm column eluting with a gradient of water and acetonitrile on a reduced sample of ConjE at 280 nm and 330 nm (Compound 44 specific) shows unconjugated light chains and a mixture of unconjugated heavy chains and heavy chains attached to a single molecule of Compound 44, consistent with a drug-per-antibody ratio (DAR) of 1.85 molecules of Compound 44 per antibody.

UHPLC analysis on a Shimadzu Prominence system using a Tosoh Bioscience TSKgel SuperSW mAb HTP 4 pm 4.6 x 150 mm column (with a 4 pm 3.0 x 20 mm guard column) eluting with 0.3 mL/minute sterile-filtered SEC buffer containing 200 mM potassium phosphate pH 6.95, 250 mM potassium chloride and 10% isopropanol (v/v) on a sample of ConjE at 280 nm shows a monomer purity of 99%. UHPLC SEC analysis gives a

concentration of final ConjE at 2.06 mg/mL in 6.8 mL, obtained mass of ConjE is 14 mg (70% yield).

Antibody-drug conjugate with Compound 45, ConjF

A 50 mM solution of DL-dithiothreitol (DTT) in phosphate-buffered saline pH 7.4 (PBS) was added (40 molar equivalent/antibody, 27 pmol, 536 pL) to a 25 mL solution of antibody Trastuzumab (100 mg, 0.67 micromoles) in reduction buffer containing PBS and 1 mM ethylenediaminetetraacetic acid (EDTA) and a final antibody concentration of 4.0 mg/mL.

The reduction mixture was allowed to react at room temperature for 17 hours (or until full reduction is observed by UHPLC) in an orbital shaker with gentle (60 rpm) shaking. After cooling down to room temperature, the reduced antibody was buffer exchanged, via Tangential Flow Filtration unit (TFF) using mPES, MidiKros ® 30 kDa fiber filter with 115 cm 2 surface area, into a reoxidation buffer containing PBS pH 7.4 and 1 mM EDTA to remove all the excess reducing agent. The reduced antibody was centrifuged for 3 min at 4000 rpm and then filtered using 0.22 mM membrane filter. A 50 mM solution of dehydroascorbic acid (DHAA, 20 molar equivalent/antibody, 12 micromoles, 240 pl_) in DMSO was added and the reoxidation mixture was allowed to react for 2-3 hours at room temperature with gentle (50 rpm) shaking at an antibody concentration of ~ 1.7 mg/ml_ (or more DHAA added and reaction left for longer until full reoxidation of the cysteine thiols to reform the inter-chain cysteine disulfides is observed by UHPLC). The reoxidation mixture was then sterile-filtered and diluted in a conjugation buffer containing PBS and 1 mM EDTA for a final antibody concentration of 1.7 mg/ml_. Compound 45 was added as a DMSO solution (10 molar equivalent/antibody, 1.0 micromole, in 1.5 mL DMSO) to 15.0 ml. of this reoxidised antibody solution (15.3 mg, 0.1 pmoles) for a 10% (v/v) final DMSO concentration. The solution left to react at room temperature for 2 hours at room temperature with gentle shaking. Then the conjugation was quenched by addition of N- acetyl cysteine (5.0 micromoles, 50 pl_ at 100 mM), then purified by spin filtration using a 15 mL Amicon Ultracell 50 kDa MWCO spin filter, sterile-filtered and analysed.

UHPLC analysis on a Shimadzu Prominence system using a Thermo Scientific MAbPac 50 mm x 2.1 mm column eluting with a gradient of water and acetonitrile on a reduced sample of ConjF at 280 nm and 330 nm (Compound 45 specific) shows unconjugated light chains and a mixture of unconjugated heavy chains and heavy chains attached to a single molecule of Compound 45, consistent with a drug-per-antibody ratio (DAR) of 1.72 molecules of Compound 45 per antibody.

UHPLC analysis on a Shimadzu Prominence system using a Tosoh Bioscience TSKgel SuperSW mAb HTP 4 pm 4.6 x 150 mm column (with a 4 pm 3.0 x 20 mm guard column) eluting with 0.3 mL/minute sterile-filtered SEC buffer containing 200 mM potassium phosphate pH 6.95, 250 mM potassium chloride and 10% isopropanol ( v/v ) on a sample of ConjF at 280 nm shows a monomer purity of 99%. UHPLC SEC analysis gives a

concentration of final ConjF at 2.02 mg/mL in 6.7 mL, obtained mass of ConjF is 13.5 mg (68% yield).

Antibody-drug conjugate with Compound 19, ConjA R347 antibody (30 mg) was loaded onto solid support and reduced, reoxidised, conjugated to Compound 19, purified, released from the resin and formulated onto 25 mM Histidine, 200 mM Sucrose, Tween-20 0.02%, pH 6.0 according to patent # US 2014/0361 11 A1.

UHPLC analysis on a Shimadzu Prominence system using a Thermo Scientific MAbPac 50 mm x 2.1 mm mm column eluting with a gradient of water and acetonitrile on a reduced sample of Conjugate at 214 nm and 330 nm (Compound 19 specific) shows unconjugated light chains and a mixture of unconjugated heavy chains and heavy chains attached to a single molecule of Compound 19, consistent with a drug-per-antibody ratio (DAR) of 1.9 molecules of Compound 19 per antibody.

UHPLC analysis on a Shimadzu Prominence system using a Tosoh Bioscience TSKgel SuperSW mAb HTP 4 pm 4.6 x 150 mm column (with a 4 pm 3.0 x 20 mm guard column) eluting with 0.3 mL/minute sterile-filtered SEC buffer containing 200 mM potassium phosphate pH 6.95, 250 mM potassium chloride and 10% isopropanol ( v/v ) on a sample of ADC at 280 nm shows a monomer purity of 97%. Nanodrop UV-Vis analysis gave a concentration of final ADC at 2.0 mg/mL in 107 mL, obtained mass of ADC is 214 mg (71 % yield).

Example 6 - In vitro cytotoxicity assay

Medium from sub-confluent (80-90% confluency) cell culture in a T75 flask was aspirated and the flask rinsed with PBS (about 20 ml) and emptied. Trypsin-EDTA (5 ml) was added, the flask returned to the 37C gassed incubator for up to about 5 minutes, then rapped sharply to dislodge and dissociate cells from the plastic. The cell suspension was transferred to a sterile 50 ml screw-top centrifuge tube, diluted with growth medium to a final volume of 15 ml, then centrifuged (400 g for 5 min). The supernatant was aspirated and the pellet re- suspended in 10 ml culture medium. Repeated pipetting may be necessary to produce monodisperse cell suspensions. The cell concentration and viability are measured by trypan blue staining, and counting using the LUNA-II™ Automated Cell Counter. Cells were diluted to 2x10 5 /ml, dispensed (50 pi /well) into 96-well flat-bottom plates and incubated overnight before use.

A stock solution (1 ml) of antibody drug conjugate (ADC) (20 pg/ml) was made by dilution of filter-sterilised ADC into cell culture medium. A set of 8x 10-fold dilutions of stock ADC were made in a 24-well plate by serial transfer of 100 mI into 900 mI of cell culture medium. ADC dilution was dispensed (50 mI/ well) into 4 replicate wells of the 96-well plate, containing 50 mI cell suspension seeded the previous day. Control wells received 50 mI cell culture medium. The 96-well plate containing cells and ADCs was incubated at 37C in a C02-gassed incubator for the exposure time. At the end of the incubation period, cell viability was measured by MTS assay. MTS

(Promega) was dispensed (20 pi per well) into each well and incubated for 4 hours at 37C in the CC> 2 -gassed incubator. Well absorbance was measured at 490 nm. Percentage cell survival was calculated from the mean absorbance in the 4 ADC-treated wells compared to the mean absorbance in the 4 control untreated wells (100%). IC 50 was determined from the dose-response data using GraphPad Prism using the non-linear curve fit algorithm:

sigmoidal dose response, X is log(concentration). ADC incubation times were 4 and 7 days for MDA MB 468 and NCI N87 respectively. Cell growth medium for MDA MB 468 was RPMI 1640 with glutamax, 10% (v/v) HyClone™ Fetal Bovine Serum and for NCI N87 was RPMI 1640 with glutamax, 10% (v/v) HyClone™ Fetal Bovine Serum.

Example 7 - In vivo assay

Mice

Female severe combined immune-deficient mice (Fox Chase SCID®, C.B-17 l\cr-Prkdcscid, Charles River) were eight to twelve weeks old on Day 1 of the study. The animals were fed ad libitum water (reverse osmosis, 1 ppm Cl), and NIH 31 Modified and Irradiated Lab Diet® consisting of 18.0% crude protein, 5.0% crude fat, and 5.0% crude fibre. The mice were housed on irradiated Enricho’cobs™ Laboratory Animal Bedding in static micro-isolators on a 12-hour light cycle at 20-22°C (68-72°F) and 40-60% humidity. CR Discovery Services specifically complies with the recommendations of the Guide for Care and Use of Laboratory Animals with respect to restraint, husbandry, surgical procedures, feed and fluid regulation, and veterinary care. The animal care and use program at CR Discovery Services is accredited by the Association for Assessment and Accreditation of Laboratory Animal Care International (AAALAC), which assures compliance with accepted standards for the care and use of laboratory animals.

Tumor Cell Culture

Human NCI-N87 gastric carcinoma lymphoma cells were cultured in RPMI-1640 medium supplemented with 10% fetal bovine serum, 2 mM glutamine, 100 units/mL penicillin, 100 pg/mL streptomycin sulfate and 25 pg/mL gentamicin. The cells were grown in tissue culture flasks in a humidified incubator at 37 °C, in an atmosphere of 5% CO2 and 95% air.

In Vivo Implantation and Tumor Growth

The NCI-N87 cells used for implantation were harvested during log phase growth and resuspended in phosphate buffered saline (PBS) containing 50% Matrigel™ (BD

Biosciences). On the day of tumor implant, each test mouse was injected subcutaneously in the right flank with 1 x 10 7 cells (0.1 ml. cell suspension), and tumor growth was monitored as the average size approached the target range of 100 to 150 mm 3 . Eleven days later, designated as Day 1 of the study, mice were sorted according to calculated tumor size into groups each consisting of ten animals and equivalent group mean tumor volumes. Tumors were measured in two dimensions using calipers, and volume was calculated using the formula:

Tumour Volume (mm 3 ) = 0.5 (w 2 x I)

where w = width and / = length, in mm, of the tumour. Tumour weight may be estimated with the assumption that 1 mg is equivalent to 1 mm 3 of tumour volume

Treatment

On Day 1 of the study, female SCID mice bearing established NCI-N87 xenografts were sorted into groups (n = 10), and dosing was initiated as shown below. All agents were administered intravenously (i.v.) via tail vein injection once on Day 1 (qd x 1 ). The dosing volume was 0.2 ml. per 20 grams of body weight (10 mL/kg), and was scaled to the body weight of each individual animal. A vehicle-treated group served as the control group for efficacy analysis. Tumors were measured using calipers twice per week, and each animal was euthanized when its tumor reached the endpoint volume of 800 mm 3 or at the end of the study (Day 85), whichever came first.

Treatment outcome was determined from percent tumor growth delay (%TGD), defined as the percent increase in median TTE for treated versus control mice, with differences between groups deemed statistically significant at P < 0.05 using logrank survival analysis. Treatment may cause partial regression (PR) or complete regression (CR) of the tumor in an animal. In a PR response, the tumor volume was 50% or less of its Day 1 volume for three consecutive measurements during the course of the study, and equal to or greater than 13.5 mm 3 for one or more of these three measurements. In a CR response, the tumor volume was less than 13.5 mm 3 for three consecutive measurements during the course of the study.

Treatment tolerability was assessed by body weight measurements and frequent observation for signs of treatment-related side effects. Treatment tolerability was assessed by body weight measurements and frequent observation for signs of treatment-related side effects.

All regimens were acceptably tolerated. The median TTE for vehicle-treated controls was 33.5 days, establishing a maximum possible TGD of 51.5 days (154%) for the 85-day study. All ADC treatments produced TGD outcomes that were significantly different from the vehicle-treated controls (P < 0.001 ).

Example 8 - Synthesis of drug payloads

A:

(i) Diallyl 8,8'-(((5-(methoxycarbonyl)-1 ,3- phenylene)b/s(methylene))bis(oxy))(11 S,1 1aS,1 TS,1 1a'S)-b/s(11-((te/f- butyldimethylsilyl)oxy)-7-methoxy-2-methyl-5-oxo-1 1 ,11 a-dihydro-1 H-benzorelpyrrolon ,2- a!H ,4ldiazepine-10(5H)-carboxylate) (46)

Phenol I3 (240 mg, 0.5 mmol, 2 eq.) and dibromo-m-xylene 21 (81 mg, 0.253 mmol, 1 eq.) were solubilised in dry DMF (20 ml.) before TBAI (1 1 mg, 0.03 mmol, 0.1 eq.) and K 2 CO 3 (70 mg, 0.5 mmol, 2 eq) were added. The mixture was heated to 60°C for an hour at which point the reaction was completed. DMF was removed under vacuum and the crude product was purified by Biotage chromatography to give pure product 46 (273 mg, 97% yield). LCMS-A: r.t. = 2.24 min, [M+H] + = 1109.

(ii) Methyl 3,5-b/s((((S)-7-methoxy-2-methyl-5-oxo-5,11 a-dihydro-1 H-benzorelpyrrolori ,2- alH ,4ldiazepin-8-yl)oxy)methyl)benzoate (47)

In a glovebox, ester 46 (60mg, 0.054 mmol, 1 eq.) was solubilised in CH2CI2 (5 ml.) before PdP(Ph3)4 (3.5 mg, 0.003 mmol, 0.05 eq.) and pyrrolidine (1 1 mI_, 0.135 mmol, 2.5 eq.) were added and the mixture left to stir until complete. The organics were diluted with more CH2CI2 and washed with sat NFUCIf aq) , water and brine before being dried with MgS0 4 , filtered and the volatiles removed in vacuo. The crude material was purified by Biotage chromatography to give pure product 47 (17.8 mg, 48% yield). LCMS-A: r.t. = 1.42 min, [M+H] + = 677; LCMS 2 : r.t. = 6.35 min, [M+H] + = 677.

(iii) 3,5-b/s((((1 1 S,1 1 aS)-10-((allyloxy)carbonyl)-1 1 -((te/f-butyldimethylsilyl)oxy)-7-methoxy-2- methyl-5-oxo-5,10,1 1 ,1 1 a-tetrahvdro-1 H-benzorelpyrroloH ,2-alH ,4ldiazepin-8- yl)oxy)methyl)benzoic acid (48)

Ester 46 (185 mg, 0.17 mmol, 1 eq.) was solubilised in a 1 :1 :1 mixture of THF/H 2 0/Me0H (10 ml.) and the mixture was stirred for 15h and after work up, product 48 (59 mg, 32% yield) was isolated. LCMS-A: r.t. = 2.15 min, [M+H] + =1095 .

(iv) 3,5-b/s((((S)-7-methoxy-2-methyl-5-oxo-5,1 1 a-dihydro-1 H-benzorelpyrroloH , 2- alH ,4ldiazepin-8-yl)oxy)methyl)benzoic acid (49)

In a glovebox, ester 48 (59mg, 0.054 mmol, 1 eq.) was solubilised in CH2CI2 (5 mL) before PdP(Ph3)4 (3.5 mg, 0.003 mmol, 0.05 eq.) and pyrrolidine (1 1 pL, 0.135 mmol, 2.5 eq.) were added and the mixture left to stir until complete. The organics were diluted with more CH2CI2 and washed with sat NhUCI^q ) , water and brine before being dried with MgS0 4 , filtered and the volatiles removed in vacuo. The crude material was purified by Biotage chromatography to give pure product 49 (17.08 mg, 48% yield). LCMS-A: r.t. = 1 .35 min, [M+H] + = 663;

LCMS-B: r.t. = 5.70 min, [M+H] + = 663.

B:

(i) Di-te/f-butyl 8,8'-(((5-(methoxycarbonyl)-1 ,3- phenylene)bis(methylene))bis(oxy))(1 1 S,1 1 aS,1 TS,1 1 a'S)-bis(7-methoxy-2-methylene-5- oxo-1 1 -((tetrahvdro-2H-pyran-2-yl)oxy)-2,3,1 1 ,1 1 a-tetrahvdro-1 H-benzorelpyrroloH ,2- alH ,4ldiazepine-10(5H)-carboxylate) (50)

Phenol I2 (100 mg, 0.2171 mmol, 2.0 eq.), bromide 21 (35 mg, 0.1086 mmol, 1.0 eq.), TBAI (1 mg, 0.002715 mmol, 0.025 eq.) and K 2 CO 3 (31.5 mg, 22.81 mmol, 2.1 eq.) were dissolved in DMF (1.5 ml.) and the mixture stirred at 35°C for 16 h, whereupon LCMS indicated the reaction was complete. The reaction mixture was concentrated in vacuo and purified by isolera chromatography (50-80% EtOAc in hexane) to afford 50 as an oil (98.3 mg, 84%). LCMS-A: r.t. = 1 .93, [M+H] + = 1 103

(ii) Methyl 3,5-b/s((((S)-7-methoxy-2-methylene-5-oxo-2,3,5,1 1 a-tetrahvdro-1 H- benzorelpyrrolor 1 ,2-aH 1 ,4ldiazepin-8-yl)oxy)methyl)benzoate (51 )

50 (86.6 mg, 80.09 pmol, 1 .0 eq) was dissolved in 95% aq. TFA (0.5 ml.) at 0°C and the mixture stirred for 40 min, whereupon LCMS indicated the reaction was complete. Ice cold water was added to the reaction mixture and solid NaHCCh was added until the pH was slightly basic. The aqueous was extracted with CHCI3, the combined organics dried over MgS0 4 and concentrated in vacuo. HPLC (20-60% B in A * ) afforded 51 as a yellow solid (6.2 mg, 1 1 %). * A = 0.1 % HC0 2 H in H 2 0. B = 0.1 % HC0 2 H in MeCN. LCMS-B: r.t. = 6.37 min, [M+H] + = 677.

(iii) Di-te/f-butyl 8,8'-(((5-(te/f-butoxycarbonyl)-1 ,3- phenylene)bis(methylene))bis(oxy))(1 1 S, 1 1 aS,1 TS, 1 1 a'S)-bis(7-methoxy-2-methylene-5- oxo-1 1 -((tetrahvdro-2H-pyran-2-yl)oxy)-2, 3,1 1 ,1 1 a-tetrahvdro-1 H-benzorelpyrroloH ,2- aH1 ,4ldiazepine-10(5H)-carboxylate) (52) Phenol I2 (100 mg, 0.2171 mmol, 2.0 eq.), bromide 6 (39.5 mg, 0.1086 mmol, 1.0 eq.), TBAI (1 mg, 0.002715 mmol, 0.025 eq.) and K 2 CO 3 (31.5 mg, 22.81 mmol, 2.1 eq.) were dissolved in DMF (1.5 ml.) and the mixture stirred at 35°C for 16 h, whereupon LCMS indicated the reaction was complete. The reaction mixture was concentrated in vacuo and purified by isolera chromatography (50-80% EtOAc in hexane) to afford 52 as an oil (120 mg, 98%). LCMS-A: r.t. =2.03, [M+H] + = 1 145

(iv) 3,5-b/s((((S)-7-Methoxy-2-methylene-5-oxo-2,3,5,1 1 a-tetrahvdro-1 H-benzorelpyrrolon ,2- alH ,4ldiazepin-8-yl)oxy)methyl)benzoic acid (53)

52 (106 mg, 94.36 pmol, 1 .0 eq) was dissolved in 95% aq. TFA (0.5 ml.) at 0°C and the mixture stirred for 40 min. Ice cold water was added to the reaction mixture and solid NaHCCh was added until the pH was neutral. The aqueous was extracted with CHCI3. The pH of the aqueous phase was adjusted to ca. pH 3 and extracted with CHCI3. The combined organics were dried over MgS0 4 and concentrated in vacuo. HPLC (20-60% B in A * ) afforded 53 as a yellow solid (3.8 mg, 6.1 %). * A = 0.1 % HC0 2 H in H 2 0. B = 0.1 % HC0 2 H in MeCN. LCMS-B: r.t. = 5.68, [M+H] + = 663

C:

(i) Di-te/f-butyl 8,8'-(((4-(methoxycarbonyl)pyridine-2,6- diyl)bis(methylene))bis(oxy))(1 1 S,1 1 aS,1 TS,1 1 a'S)-bis(1 1 -((tert-butyldimethylsilyl)oxy)-7- methoxy-2-methylene-5-oxo-2,3,1 1 ,1 1 a-tetrahvdro-1 H-benzorelpyrroloH ,2-alH ,4ldiazepine- 10(5H)-carboxylate) (54) Phenol I5 (668 mg, 1 .362 mmol, 2.2 eq.), bromide 36 (200 mg, 0.0.6192 mmol, 1 .0 eq.) and K2CO3 (188 mg, 1.362 mmol, 2.2 eq.) were dissolved in DMF (3 ml.) and the mixture stirred at rt for 3 h, then 2 h at 40°C, whereupon LCMS indicated the reaction was complete. The reaction mixture was concentrated in vacuo and purified by isolera chromatography (0-5% MeOH in CH2CI2) to afford 54 as a white solid (610 mg, 86%). LCMS-A: r.t. = 2.17, [M+Na] + = 1 164

(ii) Methyl 2,6-b/s((((S)-7-methoxy-2-methylene-5-oxo-2,3,5,1 1 a-tetrahvdro-1 H- benzorelpyrroloH ,2-alH ,4ldiazepin-8-yl)oxy)methyl)isonicotinate (55)

54 (200 mg, 0.1751 mmol, 1.0 eq) was dissolved in 95% aq. TFA (14 ml.) at 0°C and the mixture stirred for 30 min. Ice cold water was added to the reaction mixture and solid NaHCCh was added until the pH was neutral to slightly basic. The aqueous was extracted with CHCI3, dried over MgS0 4 and concentrated. Isolera chromatography (0-4% MeOH in CH2CI2) afforded 55 (61.4 mg, 52%). LCMS-A: r.t. = 5.43 min, [M+H] + = 678

(iii) 2,6-5/s((((S)-7-methoxy-2-methylene-5-oxo-2,3,5,1 1 a-tetrahydro-1 H-benzorelpyrrolori ,2- aH1 ,4ldiazepin-8-yl)oxy)methyl)/sonicotinic acid (56)

55 (55 mg, 81 .16 pmol, 1.0 eq.) and SnMesOH (58.7 mg, 324.6 pmol, 4.0 eq.) were dissolved in (CH2CI)2 (0.6 mL) and the mixture warmed to 80°C and stirred for 2.5 h, whereupon LCMS indicated the reaction was complete. The reaction mixture was diluted with CH2CI2, filtered through celite and concentrated in vacuo. HPLC (5-70% B in A * ) afforded 56 (20 mg, 37%). * A = 0.1 % HC0 2 H in H 2 0. B = 0.1 % HC0 2 H in MeCN.

LCMS-B: r.t. = 5.50 min, [M+H] + = 664

D:

(i) Di-te/f-butyl 8,8'-((1 ,3-phenylenebis(methylene))ib/s(oxy))(1 1 S,11 aS,1 S,11 a'S)-ib/s(11- ((te/f-butyldimethylsilyl)oxy)-7-methoxy-2-methylene-5-oxo-2 ,3,1 1 ,1 1a-tetrahvdro-1 H- benzorelpyrrolori ,2-aln ,4ldiazepine-10(5l-0-carboxylate) (57)

Phenol I5 (400 mg, 0.8152 mmol, 2.1 eq.), bromide 56 (103 mg, 0.3902 mmol, 1.0 eq.) and K 2 CO 3 (1 15 mg, 0.8321 mmol, 2.1 eq.) were dissolved in MEK (5 ml.) and the mixture stirred at 40°C for 2 h, then 2 h at 90 °C, whereupon LCMS indicated the reaction was incomplete. Kl (knifept.) was added and the mixture stirred at 90 °C for 2 h, whereupon LCMS indicated the reaction was complete. The reaction mixture was concentrated in vacuo and purified by isolera chromatography (10-25% EtOAc in isohexane) to afford 57 as an off-white foam (391 mg, 92%). LCMS-A: r.t. = 2.47 min, [M+H] + = 1084

(ii) (1 1 aS,11 a'S)-8,8'-((1 ,3-phenylenebis(methylene))bis(oxy))b/ ' s(7-methoxy-2-methylene- 1 ,2,3,11 a-tetrahydro-5H-benzo[e]pyrrolo[1 ,2-a][1 ,4]diazepin-5-one) (58)

57 (98 mg, 0.09045 mmol, 1.0 eq) was dissolved in 95% aq. TFA (2 mL) at 0 °C and the mixture stirred for 30 min. Ice cold water was added to the reaction mixture and solid NaHCC>3 was added until the pH was neutral to slightly basic. The aqueous was extracted with CHCI3, dried over MgS0 4 and concentrated. Isolera chromatography (0-4% MeOH in CH2CI2) afforded 58 (27.9 mg, 50%) as a pale-yellow solid. LCMS-B: r.t. = 7.55min ,

[M+H] + = 619.

K562 assay

The potency of each of the PBD molecules were measured via in vitro cytotox assays in the carcinoma cell line K562.

Solid PBD material was dissolved in DMSO to a 2 mM stock solution, from which eight serial dilutions were made at a 1 :10 ratio in DMSO and stored at -20°C until use.

Adherent K562 cells were washed with D-PBS and detached with Trypsin-EDTA, cell density and viability were then determined in duplicate by Trypan blue exclusion assay using an automated cell counter (LUNA-II™). Cell suspension was diluted to 1 x 10 5 cells/ml in growth media (RPMI 1640 with Glutamax + 10% (v/v) HyClone™ Fetal Bovine Serum) and vortexed before dispensing 2mL per well into sterile 3 mL polypropylene plates. Warhead dilutions were then dispensed into the appropriate wells at 10 mI/well and mixed by repeat pipetting. For control wells 10 m I of DMSO was dispensed onto 2 mL cell suspension, and thoroughly mixed. 100mI of each sample was then aliquoted into 2 replicate wells of a sterile flat 96-well microplate and incubated in a 37°C C0 2 -gassed (5%) incubator. At the end of the incubation period time (4 days), cell viability was measured by CellTiter 96® Aqueous One (MTS) assay, which was dispensed at 20pl/well and incubated for 4 hours at 37°C, 5% CO2 . Plates were then read on an EnVision® Multi-label Plate Reader (Perkin Elmer) using absorbance at 490 nm.

Cell survival percentage was calculated from the mean absorbance of the 2 replicate wells for each sample, compared to the mean absorbance in the two control wells treated with DMSO only (100%). The I C50 was determined by fitting each data set to sigmoidal dose- response curves with a variable slope using the non-linear curve fit algorithm on the

GraphPad Prism software (San Diego, CA).

All the experiments in this report were carried out and tested in three independent experiments. Data are reported as the mean of the three independent replicates.

NCI-N87 assay

The potency of each of the PBD molecules were measured via in vitro cytotoxicity assays in the carcinoma cell line NCI-N87.

Solid PBD material was dissolved in DMSO to a 2 mM stock solution, from which eight serial dilutions were made at a 1 :10 ratio in DMSO and stored at -20°C until use.

Adherent NCI-N87 cells were washed with D-PBS and detached with Trypsin-EDTA, cell density and viability were then determined in duplicate by Trypan blue exclusion assay using an automated cell counter (LUNA-II™). Cell suspension was diluted to 1 x 10 5 cells/ml in growth media (RPMI 1640 with Glutamax + 10% (v/v) HyClone™ Fetal Bovine Serum) and vortexed before dispensing 2ml_ per well into sterile 3 ml. polypropylene plates. Warhead dilutions were then dispensed into the appropriate wells at 10 mI/well and mixed by repeat pipetting. For control wells 10 m I of DMSO was dispensed onto 2 ml. cell suspension, and thoroughly mixed. 100mI of each sample was then aliquoted into 2 replicate wells of a sterile flat 96-well microplate and incubated in a 37°C C0 2 -gassed (5%) incubator. At the end of the incubation period time (7 days), cell viability was measured by CellTiter 96® Aqueous One (MTS) assay, which was dispensed at 20pl/well and incubated for 4 hours at 37°C, 5% CO2 . Plates were then read on an EnVision® Multi-label Plate Reader (Perkin Elmer) using absorbance at 490 nm.

Cell survival percentage was calculated from the mean absorbance of the 2 replicate wells for each sample, compared to the mean absorbance in the two control wells treated with DMSO only (100%). The I C50 was determined by fitting each data set to sigmoidal dose- response curves with a variable slope using the non-linear curve fit algorithm on the

GraphPad Prism software (San Diego, CA).

All the experiments in this report were carried out and tested in three independent experiments. Data are reported as the mean of the three independent replicates.

These results show the acid payload to have a much lower activtity in vitro than the ester, and that the addition of the ester group slightly reduces the activity of the payload,

Example 9 - Toxicity of conjugates

A single dose nonGLP toxicity study was carried out on ConjA * .

Male Sprague Dawley rats (Envigo, Inc) were dosed once by slow bolus intravenous injection via the tail vein with ADC. The vehicle for dilution used was 25 mM Histidine-HCI, 7% sucrose, 0.02% Polysorbate 80, pH 6.0. Parameters evaluated during the study included mortality, physical examinations, cageside observations, body weights, body weight changes, clinical pathology (clinical chemistry, hematology, and coagulation), and gross pathology findings. All animals were terminated on Study Day (SD) 29.

ConjA *

Adminstration of either dose did not not result in any early mortalities. Test article- associated changes in the hematology data were indicative of myelotoxicity and included decreases in all cell lineages (red blood cells, reticulocytes, platelets, neutrophils, lymphocytes, monocytes and eosinophils in most dose groups. The values generally increased at subsequent intervals, indicating reversibility.

In the coagulation data, increased fibrinogen on Day 29, in association with decreased albumin and A/G ratio, and increased globulin, was consistent with an acute systemic inflammatory response.

In group 2, there was an increased mean ALT on Day 8 that correlated with increased liver weights and mild multifocal hepatocellular hypertrophy in that group. The activities of these enzymes were comparable to, or lower than, the control group at the subsequent collections on Days 15 and 29. ALP increases of a lesser magnitude were observed in group 1 on Day 8 only. Total cholesterol and triglycerides were also elevated in both groups on Days 15 and/or 29, consistent with altered lipid metabolism and likely associated with the liver enzymes and anatomic pathology findings. Microscopic changes included randomly oriented mild multifocal hepatocellular hypertrophy in all Group 2 animals. This hepatic change correlated to increased liver weights and increased liver enzymes.

No significant changes in the bone marrow were seen in either group, which contrasts with the typical profile associated with pyrrolobenzodiazepine dimers.

Abbreviations

Ac acetyl

Acm acetamidomethyl

Alloc allyloxycarbonyl

Boc di-fe/f-butyl dicarbonate

t-Bu tert-butyl

Bzl benzyl, where Bzl-OMe is methoxybenzyl and Bzl-Me is methylbenzene

Cbz or Z benzyloxy-carbonyl, where Z-CI and Z-Br are chloro- and bromobenzyloxy carbonyl respectively

DMF /V,/V-dimethylformamide

Dnp dinitrophenyl

DTT dithiothreitol

Fmoc 9/-/-fluoren-9-ylmethoxycarbonyl

imp N- 10 imine protecting group: 3-(2-methoxyethoxy)propanoate-Val-Ala-PAB

MC-OSu maleimidocaproyi-0-/V-succinimide

Moc methoxycarbonyl

MP maleimidopropanamide

Mtr 4-methoxy-2,3,6-trimethtylbenzenesulfonyl

PAB para-aminobenzyloxycarbonyl

PEG ethyleneoxy

PNZ p-nitrobenzyl carbamate

Psec 2-(phenylsulfonyl)ethoxycarbonyl

TBDMS tert-butyldimethylsilyl

TBDPS tert-butyldiphenylsilyl

Teoc 2-(trimethylsilyl)ethoxycarbonyl

Tos tosyl

Troc 2,2,2-trichlorethoxycarbonyl chloride

Trt trityl

Xan xanthyl Numbered Statements of the Invention

1 . A compound with the formula I:

and salts and solvates thereof, wherein:

R" is a group of formula II:

where each of n and m are independently selected from 1 , 2 and 3;

R° is selected from the group consisting of H, methyl, ethyl, iso-propyl and benzyl;

Q c is selected from N and CH;

Y and Y’ are selected from O, S, or NH;

when there is a double bond present between C2 and C3, R 2 is selected from the group consisting of:

(ia) C5-10 aryl group, optionally substituted by one or more substituents selected from the group comprising: halo, nitro, cyano, ether, carboxy, ester, C alkyl, C3-7 heterocyclyl and bis-oxy-Ci-3 alkylene;

(ib) C 1-5 saturated aliphatic alkyl;

(ic) C 3-6 saturated cycloalkyl;

, wherein each of R 11 , R 12 and R 13 are independently selected from H, C1-3 saturated alkyl, C2-3 alkenyl, C2-3 alkynyl and cyclopropyl, where the total number of carbon atoms in the R 2 group is no more than 5;

(ie) , wherein one of R 15a and R 15b is H and the other is selected from: phenyl, which phenyl is optionally substituted by a group selected from halo, methyl, methoxy; pyridyl; and thiophenyl; and , where R 14 is selected from: H; C1-3 saturated alkyl; C2-3 alkenyl; C2-3 alkynyl; cyclopropyl; phenyl, which phenyl is optionally substituted by a group selected from halo, methyl, methoxy; pyridyl; and thiophenyl;

when there is a single bond present between C2 and C3,

R 2 is H or , where R 16a and R 16b are independently selected from H, F, Ci -4 saturated alkyl, C2-3 alkenyl, which alkyl and alkenyl groups are optionally substituted by a group selected from C1-4 alkyl amido and Ci -4 alkyl ester; or, when one of R 16a and R 16b is H, the other is selected from nitrile and a Ci -4 alkyl ester;

when there is a double bond present between C2’ and C3’, R 2 is selected from the group consisting of:

(iia) C 5-10 aryl group, optionally substituted by one or more substituents selected from the group comprising: halo, nitro, cyano, ether, carboxy, ester, C 1-7 alkyl, C 3-7 heterocyclyl and bis-oxy-Ci-3 alkylene;

(iib) C 1-5 saturated aliphatic alkyl;

(iic) C 3-6 saturated cycloalkyl;

wherein each of R 21 , R 22 and R 23 are independently selected from H, C1-3 saturated alkyl, C2-3 alkenyl, C2-3 alkynyl and cyclopropyl, where the total number of carbon atoms in the R 2 group is no more than 5;

,25b

R

(iie) , wherein one of R 25a and R 25b is H and the other is selected from: phenyl, which phenyl is optionally substituted by a group selected from halo, methyl, methoxy;

pyridyl; and thiophenyl; and

(iif) R ' , where R 24 is selected from: H; C1-3 saturated alkyl; C2-3 alkenyl; C2-3 alkynyl; cyclopropyl; phenyl, which phenyl is optionally substituted by a group selected from halo, methyl, methoxy; pyridyl; and thiophenyl;

when there is a single bond present between C2’ and C3’,

R 2’ is H or , where R 26a and R 26b are independently selected from H, F, Ci -4 saturated alkyl, C2-3 alkenyl, which alkyl and alkenyl groups are optionally substituted by a group selected from Ci -4 alkyl amido and Ci -4 alkyl ester; or, when one of R 26a and R 26b is H, the other is selected from nitrile and a Ci -4 alkyl ester;

R 6 and R 9 are independently selected from H, R, OH, OR, SH, SR, NH2, NHR, NRR’, nitro, MesSn and halo;

where R and R’ are independently selected from optionally substituted C1-12 alkyl, C3-20 heterocyclyl and C5-20 aryl groups;

R 7 is selected from H, R, OH, OR, SH, SR, NH2, NHR, NHRR’, nitro, MesSn and halo;

R 6 , R 7 , R 9 are selected from the same groups as R 6 , R 7 and R 9 respectively;

R 11a is selected from OH, OR A , where R A is Ci -4 alkyl, and SO z M, where z is 2 or 3 and M is a monovalent pharmaceutically acceptable cation;

and either

(a) R 30 is H, and R 31 is OH, OR A , where R A is Ci -4 alkyl;

(b) R 30 and R 31 form a nitrogen-carbon double bond between the nitrogen and carbon atoms to which they are bound; or

(c) R 30 is H and R 31 is SO z M, where z is 2 or 3 and M is a monovalent pharmaceutically acceptable cation, wherein if R 11a and R 31 are SO z M, M may represent a divalent pharmaceutically acceptable cation

R L is a linker for connection to a cell binding agent, which is selected from:

(ilia):

wherein

Q is:

, where Q x is such that Q is an amino-acid residue, a dipeptide residue or a tripeptide residue;

X is:

where a = 0 to 5, b = 0 to 16, c = 0 or 1 , d = 0 to 5;

G L is a linker for connecting to a Ligand Unit; and

(iiib): where R L1 and R L2 are independently selected from H and methyl, or together with the carbon atom to which they are bound form a cyclopropylene or cyclobutylene group;

and e is 0 or 1 .

2. A compound according to statement 1 , wherein Y and Y’ are O.

3. A compound according to either statement 1 or statement 2, wherein n is 1 .

4. A compound according to either statement 1 or statement 2, wherein n is 2.

5. A compound according to either statement 1 or statement 2, wherein n is 3.

6. A compound according to any one of statements 1 to 5, wherein m is 1.

7. A compound according to any one of statements 1 to 5, wherein m is 2.

8. A compound according to any one of statements 1 to 5, wherein m is 3.

9. A compound according to any one of statements 1 to 8, wherein Q c is N. 10. A compound according to any one of statements 1 to 8, wherein Q c is CH.

1 1. A compound according to any one of statements 1 to 10, wherein R° is H.

12. A compound according to any one of statements 1 to 10, wherein R° is methyl.

13. A compound according to any one of statements 1 to 10, wherein R° is ethyl.

14. A compound according to any one of statements 1 to 10, wherein R° is iso-propyl.

15. A compound according to any one of statements 1 to 10, wherein R° is benzyl.

16. A compound according to either statement 1 or statement 2, wherein R" is of formula lla:

where R° 1 is selected from the group consisting of H and methyl.

17. A compound according to any one of statements 1 to 16, wherein R 9 and R 9 are H.

18. A compound according to any one of statements 1 to 17, wherein R 6 and R 6 are independently selected from H and halo.

19. A compound according to statement 18, wherein R 6 and R 6 are H.

20. A compound according to any one of statements 1 to 19, wherein R 7 and R 7 are independently selected from H, OH and OR.

21. A compound according to statement 20, wherein R 7 and R 7 are independently a Ci -4 alkoxy group.

22. A compound according to statement 21 , wherein R 7 and R 7 are methoxy. 23. A compound according to any one of statements 1 to 22, wherein there is a double bond between C2 and C3, and R 2 is a C5-7 aryl group.

24. A compound according to statement 23, wherein R 2 is phenyl.

25. A compound according to any one of statements 1 to 22, wherein there is a double bond between C2 and C3, and R 2 is a Ce-io aryl group.

26. A compound according to any one of statements 23 to 25, wherein R 2 bears one to three substituent groups.

27. A compound according to any one of statements 23 to 26, wherein the substituents are selected from methoxy, ethoxy, fluoro, chloro, cyano, bis-oxy-methylene, methyl- piperazinyl, morpholino and methyl-thiophenyl.

28. A compound according to any one of statements 1 to 22, wherein there is a double bond between C2 and C3, and R 2 is a C1-5 saturated aliphatic alkyl group.

29. A compound according to statement 28, wherein R 2 is methyl, ethyl or propyl.

30. A compound according to any one of statements 1 to 22, wherein there is a double bond between C2 and C3, and R 2 is a C3-6 saturated cycloalkyl group.

31 . A compound according to statement 30, wherein R 2 is cyclopropyl.

32. A compound according to any one of statements 1 to 22, wherein there is a double bond between C2 and C3, and R 2 is a group of formula:

33. A compound according to statement 32, wherein the total number of carbon atoms in the R 2 group is no more than 4. 34. A compound according to statement 32, wherein the total number of carbon atoms in the R 2 group is no more than 3.

35. A compound according to any one of statements 32 to 34, wherein one of R 11 , R 12 and R 13 is H, with the other two groups being selected from H, C1-3 saturated alkyl, C2-3 alkenyl, C2-3 alkynyl and cyclopropyl.

36. A compound according to any one of statements 32 to 34, wherein two of R 11 , R 12 and R 13 are H, with the other group being selected from H, C1-3 saturated alkyl, C2-3 alkenyl, C2-3 alkynyl and cyclopropyl.

37. A compound according to any one of statements 1 to 22, wherein there is a double bond between C2 and C3, and R 2 is a group of formula:

38. A compound according to statement 37, wherein R 2 is the group:

39. A compound according to any one of statements 1 to 22, wherein there is a double bond between C2 and C3, and R 2 is a group of formula:

40. A compound according to statement 39, wherein R 14 is selected from H, methyl, ethyl, ethenyl and ethynyl.

41 . A compound according to statement 40, wherein R 14 is selected from H and methyl.

42. A compound according to any one of statements 1 to 22, wherein there is a single bond between C2 and C3, R 2 is H. 43. A compound according to any one of statements 1 to 22, wherein there is a single

bond between and R 16a and R 16b are both H.

44. A compound according to any one of statements 1 to 22, wherein there is a single

bond between C2 and C3, R 2 , and R 16a and R 16b are both methyl.

45. A compound according to any one of statements 1 to 22, wherein there is a single

bond between C2 and C3, R 2 , one of R 16a and R 16b is H, and the other is selected from Ci -4 saturated alkyl, C2-3 alkenyl, which alkyl and alkenyl groups are optionally substituted.

46. A compound according to any one of statements 1 to 45, wherein there is a double bond between C2’ and C3’, and R 2’ is a C5-7 aryl group.

47. A compound according to statement 46, wherein R 2’ is phenyl.

48. A compound according to any one of statements 1 to 45, wherein there is a double bond between C2’ and C3’, and R 2’ is a Ce-io aryl group.

49. A compound according to any one of statements 46 to 48, wherein R 2’ bears one to three substituent groups.

50. A compound according to any one of statements 46 to 49, wherein the substituents are selected from methoxy, ethoxy, fluoro, chloro, cyano, bis-oxy-methylene, methyl- piperazinyl, morpholino and methyl-thiophenyl.

51. A compound according to any one of statements 1 to 45, wherein there is a double bond between C2’ and C3’, and R 2 is a C1-5 saturated aliphatic alkyl group. 52. A compound according to statement 51 , wherein R 2’ is methyl, ethyl or propyl.

53. A compound according to any one of statements 1 to 45, wherein there is a double bond between C2’ and C3’, and R 2’ is a C saturated cycloalkyl group.

54. A compound according to statement 53, wherein R 2’ is cyclopropyl.

55. A compound according to any one of statements 1 to 45, wherein there is a double bond between C2’ and C3’, and R 2 is a group of formula:

56. A compound according to statement 55, wherein the total number of carbon atoms in the R 2 group is no more than 4.

57. A compound according to statement 56, wherein the total number of carbon atoms in the R 2 group is no more than 3.

58. A compound according to any one of statements 55 to 57, wherein one of R 21 , R 22 and R 23 is H, with the other two groups being selected from H, C1-3 saturated alkyl, C2-3 alkenyl, C2-3 alkynyl and cyclopropyl.

59. A compound according to any one of statements 55 to 57, wherein two of R 21 , R 22 and R 23 are H, with the other group being selected from H, C1-3 saturated alkyl, C2-3 alkenyl, C2-3 alkynyl and cyclopropyl.

60. A compound according to any one of statements 1 to 45, wherein there is a double bond between C2’ and C3’, and R 2 is a group of formula:

61 . A compound according to statement 60, wherein R 2’ is the group:

62. A compound according to any one of statements 1 to 45, wherein there is a double bond between C2’ and C3’, and R 2 is a group of formula:

63. A compound according to statement 62, wherein R 24 is selected from H, methyl, ethyl, ethenyl and ethynyl. 64. A compound according to statement 63, wherein R 24 is selected from H and methyl.

65. A compound according to any one of statements 1 to 45, wherein there is a single bond between C2’ and C3’, R 2 is H. 66. A compound according to any one of statements 1 to 45, wherein there is a single

bond between and R 26a and R 26b are both H.

67. A compound according to any one of statements 1 to 45, wherein there is a single

bond between C2’ and C3’, R 2 is , and R 26a and R 26b are both methyl.

68. A compound according to any one of statements 1 to 45, wherein there is a single .R 26a

l 26b

bond between C2’ and C3’, R 2 is R , one of R 26a and R 26b is H, and the other is selected from Ci -4 saturated alkyl, C2-3 alkenyl, which alkyl and alkenyl groups are optionally substituted. 69. A compound according to any one of statements 1 to 68, wherein R 11a is OH.

70. A compound according to any one of statements 1 to 68, wherein R 11a is OR A .

71. A compound according to statement 70, wherein R A is methyl.

72. A compound according to any one of statements 1 to 68, wherein R 11a is SO z M.

73. A compound according to statement 72, wherein M is Na + and z is 3.

74. A compound according to any one of statements 1 to 73, wherein R 30 is H, and R 31 is OH.

75. A compound according to any one of statements 1 to 73, wherein R 30 is H, and R 31 is OR A .

76. A compound according to statement 75, wherein R A is methyl.

77. A compound according to any one of statements 1 to 73, wherein R 30 and R 31 form a nitrogen-carbon double bond between the nitrogen and carbon atoms to which they are bound.

78. A compound according to any one of statements 1 to 73, wherein R 30 is H and R 31 is SO z M.

79. A compound according to statement 78, wherein M is Na + and z is 3.

80. A compound according to statement 1 of formula la:

where R 2a and R 2a are the same and are selected from:

R 7a and R 7a are the same and are selected from methoxy and benzyloxy.

81 . A compound according to statement 1 of formula lb:

wherein R 7a and R 7a are the same and are selected from methoxy and benzyloxy. 82. A compound according to statement 1 of formula lc:

wherein R 7a and R 7a are the same and are selected from methoxy and benzyloxy. 83. A compound according to any one of statements 1 to 82, wherein R L is of formula

Ilia, and Q is an amino acid residue selected from Phe, Lys, Val, Ala, Cit, Leu, lie, Arg, and Trp.

84. A compound according to any one of statements 1 to 82, wherein R L is of formula Ilia, and Q is a dipeptide residue selected from:

c o -Phe-Lys- NH ,

c o -Val-Ala- NH ,

c o -Val-Lys- NH ,

c o -Ala-Lys- NH ,

c o -Val-Cit- NH ,

c o -Phe-Cit- NH ,

c o -Leu-Cit- NH ,

c o -lle-Cit- NH ,

c o -Phe-Arg- NH , and

c o -Trp-Cit- NH .

85. A compound according to statement 84, wherein Q is selected from co -Phe-Lys- NH , co -Val-Cit- NH and co -Val-Ala- NH . 86. A compound according to any one of statements 1 to 82, wherein R L is of formula

Ilia, and Q is a tripeptide residue. 87. A compound according to any one of statements 1 to 86, wherein R L is of formula Ilia and a is 0 to 3.

88. A compound according to statement 87, wherein a is 0.

89. A compound according to any one of statements 1 to 88, wherein R L is of formula Ilia and b is 0 to 12.

90. A compound according to statement 89, wherein b is 0 to 8.

91. A compound according to any one of statements 1 to 90, wherein R L is of formula Ilia and d is 0 to 3.

92. A compound according to statement 91 , wherein d is 2.

93. A compound according to any one of statements 1 to 86, wherein R L is of formula Ilia and, a is 0, c is 1 and d is 2, and b is from 0 to 8.

94. A compound according to statement 93 wherein b is 0, 4 or 8.

95. A compound according to any one of statements 1 to 94, wherein R L is of formula Ilia and G L is selected from

where Ar represents a C5-6 arylene group, e.g. phenylene, and X represents C1-4 alkyl.

96. A compound according to statement 95, wherein Ar is a phenylene group. 97. A compound according to either statement 95 or statement 96, wherein G L is selected from G L1 1 and G L1 2 . 98. A compound according to statement 97, wherein G L is G L1 1 .

99. A compound according to any one of statements 1 to 82, wherein R L is of formula 11 lb, and both R L1 and R L2 are H.

100. A compound according to any one of statements 1 to 82, wherein R L is of formula

I Mb, R L1 is H and R L2 is methyl.

101 . A compound according to any one of statements 1 to 82, wherein R L is of formula

I I lb, and both R L1 and R L2 are methyl.

102. A compound according to any one of statements 1 to 82, wherein R L is of formula 11 lb, and, R L1 and R L2 together with the carbon atom to which they are bound form a cyclopropylene group.

103. A compound according to any one of statements 1 to 82, wherein R L is of formula 11 lb, and, R L1 and R L2 together with the carbon atom to which they are bound form a cyclobutylene group.

104. A compound according to any one of statements 1 to 82 and 99 to 103, wherein R L is of formula I Mb, and e is 0.

105. A compound according to any one of statements 1 to 82 and 99 to 103, wherein R L is of formula lllb, and e is 1.

106. A compound according to statement 105, wherein the nitro group is in the para position.

107. The use of a compound according to any one of statements 1 to 106 in the manufacture of a medicament for treating a proliferative disease.

108. A compound according to any one of statements 1 to 106 for use in the treatment of a proliferative disease.

109. A conjugate comprising a compound of formula I according to any one of statements 1 to 106, or a pharmaceutically acceptable salt or solvate thereof, linked to a Ligand unit. 1 10. A conjugate of formula IV:

L - (D L ) P (IV)

or a pharmaceutically acceptable salt or solvate thereof, wherein L is a Ligand unit , D L is of formula III:

wherein R 2 , R 6 , R 7 , R 9 , R 11a , Y, R”, Y’, R 2’ , R 6’ , R 7’ , R 9’ , R 30 and R 31 are as defined in any one of statements 1 to 79;

R LL is a linker for connection to a cell binding agent, which is selected from:

(iiia):

where Q and X are as defined in any one of statements 1 and 83 to 86 and G LL is a linker connected to a Ligand Unit; and

(iiib): mb'

where R L1 and R L2 are as defined in any one of statements 1 and 99 to 103;

wherein p is an integer of from 1 to 20.

1 1 1 . A conjugate according to statement 1 10, wherein G LL is selected from:

112. A conjugate according to statement 111, wherein Ar is a phenylene group.

113. A compound according to either statement 111 or statement 112, wherein G LL is selected from G LL11 and G LL12 .

114. A compound according to statement 112, wherein G LL is G LL11 .

115. A conjugate according to statement 110, wherein D L is of formula Ilia:

where R 2a and R 2a are the same and are selected from:

R 7a and R 7a are the same and are slelected from methoxy and benzyloxy. 1 16. A conjugate according to statement 1 10, wherein D L is of formula I Mb:

where R 7a and R 7a are the same and are selected from methoxy and benzyloxy. 1 17. A conjugate according to statement 1 10, wherein D L is of formula lllc:

where R 7a and R 7a are the same and are selected from methoxy and benzyloxy. 1 18. A conjugate according to any one of statements 109 to 1 17, wherein the Ligand Unit is an antibody or an active fragment thereof.

1 19. The conjugate according to statement 1 18, wherein the antibody or antibody fragment is an antibody or antibody fragment for a tumour-associated antigen.

120. The conjugate according to statement 1 19, wherein the antibody or antibody fragment is an antibody which binds to one or more tumor-associated antigens or cell- surface receptors selected from (1 )-(88):

(1 ) BMPR1 B;

(2) E16;

(3) STEAP1 ; (4) 0772P;

(5) MPF;

(6) Napi3b;

(7) Sema 5b;

(8) PSCA hlg;

(9) ETBR;

(10) MSG783;

(11 ) STEAP2;

(12) TrpM4;

(13) CRIPTO;

(14) CD21 ;

(15) CD79b;

(16) FcRH2;

(17) HER2;

(18) NCA;

(19) MDP;

(20) IL20R-alpha;

(21 ) Brevican;

(22) EphB2R;

(23) ASLG659;

(24) PSCA;

(25) GEDA;

(26) BAFF-R;

(27) CD22;

(28) CD79a;

(29) CXCR5;

(30) HLA-DOB;

(31 ) P2X5;

(32) CD72;

(33) LY64;

(34) FcRH1 ;

(35) IRTA2;

(36) TENB2;

(37) PSMA - FOLH1 ;

(38) SST;

(38.1 ) SSTR2; (38.2) SSTR5;

(38.3) SSTR1 ;

(38.4) SSTR3;

(38.5) SSTR4;

(39) ITGAV;

(40) ITGB6;

(41) CEACAM5;

(42) MET;

(43) MUC1 ;

(44) CA9;

(45) EGFRvlll;

(46) CD33;

(47) CD19;

(48) IL2RA;

(49) AXL;

(50) CD30 - TNFRSF8;

(51) BCMA - TNFRSF17;

(52) CT Ags - CTA;

(53) CD174 (Lewis Y) - FUT3;

(54) CLEC14A;

(55) GRP78 - HSPA5;

(56) CD70;

(57) Stem Cell specific antigens;

(58) ASG-5;

(59) ENPP3;

(60) PRR4;

(61) GCC - GUCY2C;

(62) Liv-1 - SLC39A6;

(63) 5T4;

(64) CD56 - NCMA1 ;

(65) CanAg;

(66) F0LR1 ;

(67) GPNMB;

(68) TIM-1 - HAVCR1 ;

(69) RG-1/Prostate tumor target Mindin - Mindin/RG-1 ;

(70) B7-H4 - VTCN1 ; (71) PTK7;

(72) CD37;

(73) CD138 - SDC1 ;

(74) CD74;

(75) Claudins - CLs;

(76) EGFR;

(77) Her3;

(78) RON - MST1 R;

(79) EPHA2;

(80) CD20 - MS4A1 ;

(81) Tenascin C - TNC;

(82) FAP;

(83) DKK-1 ;

(84) CD52;

(85) CS1 - SLAMF7;

(86) Endoglin - ENG;

(87) Annexin A1 - ANXA1 ;

(88) V-CAM (CD106) - VCAM1.

121. The conjugate of any one of statements 1 18 to 120 wherein the antibody or antibody fragment is a cysteine-engineered antibody.

122. The conjugate according to any one of statements 1 10 to 121 wherein p is an integer from 1 to 8.

123. The conjugate according to statement 122, wherein p is 1 , 2, 3, or 4.

124. A composition comprising a mixture of conjugates according to any one of statements 1 10 to 123, wherein the average p in the mixture of conjugate compounds is about 1 to about 8.

125. The conjugate according to any one of statements 109 to 123, for use in therapy.

126. A pharmaceutical composition comprising the conjugate of any one of statements 109 to 123 and a pharmaceutically acceptable diluent, carrier or excipient. 127. The conjugate according to any one of statements 109 to 123 or the pharmaceutical composition according to statement 126, for use in the treatment of a proliferative disease in a subject.

128. The conjugate for use according to statement 127, wherein the disease treated is cancer.

129. Use of a conjugate according to any one of statements 109 to 123 or the

pharmaceutical composition according to statement 126 in a method of medical treatment.

130. A method of medical treatment comprising administering to a patient the

pharmaceutical composition of statement 126.

131 . The method of statement 130 wherein the method of medical treatment is for treating cancer.

132. The method of statement 131 , wherein the patient is administered a

chemotherapeutic agent, in combination with the conjugate.

133. Use of a conjugate according to any one of statements 109 to 123 in a method of manufacture of a medicament for the treatment of a proliferative disease.

134. A method of treating a mammal having a proliferative disease, comprising administering an effective amount of a conjugate according to any one of statements 109 to 123 or the pharmaceutical composition according to statement 126.

135. A compound with the formula REL:

and salts and solvates thereof, wherein R 2 , R 6 , R 7 , R 9 , Y, R”, Y’, R 2’ , R 6’ , R 7’ , R 9’ , R 30 and R 31 are as defined in any one of statements 1 to 79.