Pyrrolobenzodiazepine antibody conjugates

Information

  • Patent Grant
  • 11517626
  • Patent Number
    11,517,626
  • Date Filed
    Friday, February 10, 2017
    7 years ago
  • Date Issued
    Tuesday, December 6, 2022
    a year ago
Abstract
A conjugate of formula I: L-(DL)P  (1)
Description

Incorporated by reference in its entirety herein is a computer-readable nucleotide/amino acid sequence listing submitted herewith and identified as follows: One 45,384 Byte ASCII (Text) file named “36676_251_ST25.TXT,” created on Oct. 13, 2022. The present invention relates to conjugates comprising a specific pyrrolobenzodiazepine (PBD), and the precursor drug linker used to make such conjugates.


BACKGROUND TO THE INVENTION

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 al., J. Antibiotics, 37, 200-206 (1984)), DC-81 (Japanese Patent 58-180 487; Thurston, et al., Chem. Brit., 26, 767-772 (1990); Bose, et al., Tetrahedron, 48, 751-758 (1992)), mazethramycin (Kuminoto, et al., 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:




embedded image


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-C11 position which is the electrophilic centre responsible for alkylating DNA. All of the known natural products have an (S)-configuration at the chiral C11a 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. 3-11 (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.


It has been previously disclosed that the biological activity of this molecules can be potentiated by joining two PBD units together through their C8/C′-hydroxyl functionalities via a flexible alkylene linker (Bose, D. S., et al., J. Am. Chem. Soc., 114, 4939-4941 (1992); Thurston, D. E., et al., J. Org. Chem., 61, 8141-8147 (1996)). The PBD dimers are thought to form sequence-selective DNA lesions such as the palindromic 5′-Pu-GATC-Py-3′ interstrand cross-link (Smellie, M., et al., Biochemistry, 42, 8232-8239 (2003); Martin, C., et al., Biochemistry, 44, 4135-4147) which is thought to be mainly responsible for their biological activity.


One example of a PBD dimer is SG2000 (SJG-136):




embedded image


(Gregson, S., et al., J. Med. Chem., 44, 737-748 (2001); Alley, M. C., et al., Cancer Research, 64, 6700-6706 (2004); Hartley, J. A., et al., Cancer Research, 64, 6693-6699 (2004)) which has been involved in clinical trials as a standalone agent, for example, NCT02034227 investigating its use in treating Acute Myeloid Leukemia and Chronic Lymphocytic Leukemia (see: clinicaltrials.gov/ct2/show/NCT02034227).


Dimeric PBD compounds bearing C2 aryl substituents, such as SG2202 (ZC-207), are disclosed in WO 2005/085251:




embedded image


and in WO2006/111759, bisulphites of such PBD compounds, for example SG2285 (ZC-423):




embedded image


These compounds have been shown to be highly useful cytotoxic agents (Howard, P. W., et al., Bioorg. Med. Chem. (2009), doi: 10.1016/j.bmcl.2009.09.012).


In an impact study submitted to the 2014 Research Excellence Framework (REF) in the United Kingdom by University College London (available at impact.ref.ac.uk/casestudies2/refservice.svc/GetCaseStudyPDF/35393), it was commented that:


“The next generation of PBD dimers, which are more potent than SG2000, have been developed, including SG2057 and SG2202. They exhibit picomolar/sub-picomolar activity against a range of human tumour cell lines and demonstrate curative activity in human tumour xenograft models.” making reference to: Hartley J A, et al., DNA interstrand cross-linking and in vivo antitumor activity of the extended pyrrolo[2,1-c][1,4]benzodiazepine dimer SG2057. Invest New Drugs. 2012 June; 30(3):950-8.dx.doi.org/10.1007/s10637-011-9647-z (herein after “Hartley et al (2012)”) and:


“The ability to generate such cytotoxic molecules that display exquisite potency suggested a potential role in strategies aimed at targeting and releasing highly cytotoxic agents directly at a tumour site. An example is as the ‘warhead’ component of an antibody drug conjugate (ADC). The fully synthetic PBD dimers are ideally suited for the role of warhead in an ADC approach.”


The Hartley et al (2012) paper comments in its summary that “SG2057 is therefore a highly active antiumour agent, with more potent in vitro activity and superior in vivo activity to SG2000, warranting further development”.


SG2057 has the structure:




embedded image


Antibody drug conjugates using SG2057 as a warhead were first disclosed in WO 2011/130598. For example, claim 54 of this application includes the formula:




embedded image


wherein n is from 1 to 24, more preferably 4 to 8. The following drug linkers were exemplified: n=4, 15c; n=8, 15 d; n=24, 15e.


Claim 54 of this application also includes the formula:




embedded image



wherein n is from 1 to 24, more preferably 4 to 8. The following drug linkers were exemplified: n=8, 58; n=24, 61.


WO 2011/130598 also discloses antibody-drug conjugates including these drug linkers, for example 110 (antiSteap1-15 d), example 114 (tastuzumab-15d) and example 115 (tastuzumab-58).


WO 2013/055987 discloses the drug linkers 14 and 22:




embedded image


and their use in antibody-drug conjugates.


More recently, the warhead:




embedded image


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




embedded image


WO2015/052322 discloses:




embedded image


DISCLOSURE OF THE INVENTION

The present inventors have surprisingly found that although SG2000 is at least 10 times less cytotoxic than SG2057 (see Hartley et al 2012), particular antibody-drug conjugates appear to show at least comparable activity. These conjugates have been shown to have surprisingly well tolerated in toxicity studies in a variety of species. This leads to the conjugates exhibiting high therapeutic indices and thus are promising clinical candidates.


In a first aspect, the present invention provides Conjugates of formula I:

L-(DL)P  (I)


wherein L is a Ligand unit (i.e., a targeting agent), DL is a Drug Linker unit of formula II:




embedded image


wherein


either:


(a) R10 and R11 form a nitrogen-carbon double bond between the nitrogen and carbon atoms to which they are bound; or


(b) R11 is OH, and R10 is:




embedded image


p is an integer of from 1 to 20.


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. 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.


A second aspect of the present invention provides a compound of formula III:




embedded image


wherein


either:


(a) R10 and R11 form a nitrogen-carbon double bond between the nitrogen and carbon atoms to which they are bound; or


(b) R11 is OH, and R10 is:




embedded image


A third aspect of the present invention provides the use of a conjugate of the first aspect of the invention in the manufacture of a medicament for treating a proliferative disease. The third aspect also provides a conjugate of the first aspect of the invention for use in the treatment of a proliferative disease. The third aspect also provides a method of treating a proliferative disease comprising administering a therapeutically effective amount of a conjugate of the first aspect of the invention to a patient in need thereof.


One of ordinary skill in the art is readily able to determine whether or not a candidate conjugate 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 fourth aspect of the present invention provides the synthesis of a conjugate of the first aspect of the invention comprising conjugating a compound (drug linker) of the second aspect of the invention with a Ligand Unit.





BRIEF DESCRIPTION OF FIGURES


FIG. 1 shows the effect on volume of a BT474 tumour following treatment with a conjugate of the present invention;



FIG. 2 shows the effect on volume of a BT474 tumour following treatment with a different conjugate of the present invention;



FIG. 3 shows the effect on volume of a NCI-N87 tumour following treatment with a conjugate of the present invention;



FIG. 4 shows the effect on volume of a NCI-N87 tumour following treatment with a different conjugate of the present invention.





DL


In the first aspect DL is selected from DL-A and DL-B:




embedded image


In the second aspect, the compound is selected from A and B:




embedded image


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 1×107 MA, 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 αvβ6. The peptide may be selective for αVβ6 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), 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. IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) 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-Id) 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, U.S. Pat. No. 4,816,567). 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 in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (U.S. Pat. No. 4,816,567; and Morrison et al (1984) Proc. Natl. Acad. Sci. USA, 81:6851-6855). Chimeric antibodies include “primatized” antibodies comprising variable domain antigen-binding sequences derived from a non-human primate (e.g. Old World Monkey or Ape) and human constant region sequences.


An “intact antibody” herein is one comprising a VL and VH domains, as well as a light chain constant domain (CL) and heavy chain constant domains, CH1, CH2 and CH3. The constant domains may be native sequence constant domains (e.g. human native sequence constant domains) or amino acid sequence variant thereof. The intact antibody may have one or more “effector functions” which refer to those biological activities attributable to the Fc region (a native sequence Fc region or amino acid sequence variant Fc region) of an antibody. Examples of antibody effector functions include C1q binding; complement dependent cytotoxicity; Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; and down regulation of cell surface receptors such as B cell receptor and BCR.


Depending on the amino acid sequence of the constant domain of their heavy chains, intact antibodies can be assigned to different “classes.” There are five major classes of intact antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into “subclasses” (isotypes), e.g., IgG1, IgG2, IgG3, IgG4, IgA, and IgA2. The heavy-chain constant domains that correspond to the different classes of antibodies are called α, δ, ε, γ, and μ, respectively. The subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.


Humanisation


Techniques to reduce the in vivo immunogenicity of a non-human antibody or antibody fragment include those termed “humanisation”.


A “humanized antibody” refers to a polypeptide comprising at least a portion of a modified variable region of a human antibody wherein a portion of the variable region, preferably a portion substantially less than the intact human variable domain, has been substituted by the corresponding sequence from a non-human species and wherein the modified variable region is linked to at least another part of another protein, preferably the constant region of a human antibody. The expression “humanized antibodies” includes human antibodies in which one or more complementarity determining region (“CDR”) amino acid residues and/or one or more framework region (“FW” or “FR”) amino acid residues are substituted by amino acid residues from analogous sites in rodent or other non-human antibodies. The expression “humanized antibody” also includes an immunoglobulin amino acid sequence variant or fragment thereof that comprises an FR having substantially the amino acid sequence of a human immunoglobulin and a CDR having substantially the amino acid sequence of a non-human immunoglobulin.


“Humanized” forms of non-human (e.g., murine) antibodies are chimeric antibodies that contain minimal sequence derived from non-human immunoglobulin. Or, looked at another way, a humanized antibody is a human antibody that also contains selected sequences from non-human (e.g. murine) antibodies in place of the human sequences. A humanized antibody can include conservative amino acid substitutions or non-natural residues from the same or different species that do not significantly alter its binding and/or biologic activity. Such antibodies are chimeric antibodies that contain minimal sequence derived from non-human immunoglobulins.


There are a range of humanisation techniques, including ‘CDR grafting’, ‘guided selection’, ‘deimmunization’, ‘resurfacing’ (also known as ‘veneering’), ‘composite antibodies’, ‘Human String Content Optimisation’ and framework shuffling.


CDR Grafting


In this technique, the humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a complementary-determining region (CDR) of the recipient antibody are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat, camel, bovine, goat, or rabbit having the desired properties (in effect, the non-human CDRs are ‘grafted’ onto the human framework). In some instances, framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues (this may happen when, for example, a particular FR residue has significant effect on antigen binding).


Furthermore, humanized antibodies can comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences. These modifications are made to further refine and maximize antibody performance. Thus, in general, a humanized antibody will comprise all of at least one, and in one aspect two, variable domains, in which all or all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin sequence. The humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), or that of a human immunoglobulin.


Guided Selection


The method consists of combining the VH or VL domain of a given non-human antibody specific for a particular epitope with a human VH or VL library and specific human V domains are selected against the antigen of interest. This selected human VH is then combined with a VL library to generate a completely human VH×VL combination. The method is described in Nature Biotechnology (N.Y.) 12, (1994) 899-903.


Composite Antibodies


In this method, two or more segments of amino acid sequence from a human antibody are combined within the final antibody molecule. They are constructed by combining multiple human VH and VL sequence segments in combinations which limit or avoid human T cell epitopes in the final composite antibody V regions. Where required, T cell epitopes are limited or avoided by, exchanging V region segments contributing to or encoding a T cell epitope with alternative segments which avoid T cell epitopes. This method is described in US 2008/0206239 A1.


Deimmunization


This method involves the removal of human (or other second species) T-cell epitopes from the V regions of the therapeutic antibody (or other molecule). The therapeutic antibodies V-region sequence is analysed for the presence of MHC class II-binding motifs by, for example, comparison with databases of MHC-binding motifs (such as the “motifs” database hosted at www.wehi.edu.au). Alternatively, MHC class II-binding motifs may be identified using computational threading methods such as those devised by Altuvia et al. (J. Mol. Biol. 249 244-250 (1995)); in these methods, consecutive overlapping peptides from the V-region sequences are testing for their binding energies to MHC class II proteins. This data can then be combined with information on other sequence features which relate to successfully presented peptides, such as amphipathicity, Rothbard motifs, and cleavage sites for cathepsin B and other processing enzymes.


Once potential second species (e.g. human) T-cell epitopes have been identified, they are eliminated by the alteration of one or more amino acids. The modified amino acids are usually within the T-cell epitope itself, but may also be adjacent to the epitope in terms of the primary or secondary structure of the protein (and therefore, may not be adjacent in the primary structure). Most typically, the alteration is by way of substitution but, in some circumstances amino acid addition or deletion will be more appropriate.


All alterations can be accomplished by recombinant DNA technology, so that the final molecule may be prepared by expression from a recombinant host using well established methods such as Site Directed Mutagenesis. However, the use of protein chemistry or any other means of molecular alteration is also possible.


Resurfacing


This method involves:

    • (a) determining the conformational structure of the variable region of the non-human (e.g. rodent) antibody (or fragment thereof) by constructing a three-dimensional model of the non-human antibody variable region;
    • (b) generating sequence alignments using relative accessibility distributions from x-ray crystallographic structures of a sufficient number of non-human and human antibody variable region heavy and light chains to give a set of heavy and light chain framework positions wherein the alignment positions are identical in 98% of the sufficient number of non-human antibody heavy and light chains;
    • (c) defining for the non-human antibody to be humanized, a set of heavy and light chain surface exposed amino acid residues using the set of framework positions generated in step (b);
    • (d) identifying from human antibody amino acid sequences a set of heavy and light chain surface exposed amino acid residues that is most closely identical to the set of surface exposed amino acid residues defined in step (c), wherein the heavy and light chain from the human antibody are or are not naturally paired;
    • (e) substituting, in the amino acid sequence of the non-human antibody to be humanized, the set of heavy and light chain surface exposed amino acid residues defined in step (c) with the set of heavy and light chain surface exposed amino acid residues identified in step (d);
    • (f) constructing a three-dimensional model of the variable region of the non-human antibody resulting from the substituting specified in step (e);
    • (g) identifying, by comparing the three-dimensional models constructed in steps (a) and (f), any amino acid residues from the sets identified in steps (c) or (d), that are within 5 Angstroms of any atom of any residue of the complementarity determining regions of the non-human antibody to be humanized; and
    • (h) changing any residues identified in step (g) from the human to the original non-human amino acid residue to thereby define a non-human antibody humanizing set of surface exposed amino acid residues; with the proviso that step (a) need not be conducted first, but must be conducted prior to step (g).


Superhumanization


The method compares the non-human sequence with the functional human germline gene repertoire. Those human genes encoding canonical structures identical or closely related to the non-human sequences are selected. Those selected human genes with highest homology within the CDRs are chosen as FR donors. Finally, the non-human CDRs are grafted onto these human FRs. This method is described in patent WO 2005/079479 A2.


Human String Content Optimization


This method compares the non-human (e.g. mouse) sequence with the repertoire of human germline genes and the differences are scored as Human String Content (HSC) that quantifies a sequence at the level of potential MHC/T-cell epitopes. The target sequence is then humanized by maximizing its HSC rather than using a global identity measure to generate multiple diverse humanized variants (described in Molecular Immunology, 44, (2007) 1986-1998).


Framework Shuffling


The CDRs of the non-human antibody are fused in-frame to cDNA pools encompassing all known heavy and light chain human germline gene frameworks. Humanised antibodies are then selected by e.g. panning of the phage displayed antibody library. This is described in Methods 36, 43-60 (2005).


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.


Tumor-Associated Antigens and Cognate Antibodies


(1) BMPR1B (Bone Morphogenetic Protein Receptor-Type IB)


Nucleotide


Genbank accession no. NM_001203


Genbank version no. NM_001203.2 GI:169790809


Genbank record update date: Sep. 23, 2012 02:06 PM


Polypeptide


Genbank accession no. NP_001194


Genbank version no. NP_001194.1 GI:4502431


Genbank record update date: Sep. 23, 2012 02:06 PM


CROSS-REFERENCES



  • ten Dijke,P., et al Science 264 (5155): 101-104 (1994), Oncogene 14 (11):1377-1382 (1997)); WO2004/063362 (Claim 2); WO2003/042661 (Claim 12); US2003/134790-A1 (Page 38-39); WO2002/102235 (Claim 13; Page 296); WO2003/055443 (Page 91-92); WO2002/99122 (Example 2; Page 528-530); WO2003/029421 (Claim 6); WO2003/024392 (Claim 2; FIG. 112); WO2002/98358 (Claim 1; Page 183); WO2002/54940 (Page 100-101); WO2002/59377(Page 349-350); WO2002/30268 (Claim 27; Page 376); 15 WO2001/48204 (Example; FIG. 4); NP_001194 bone morphogenetic protein receptor, type IB/pid=NP_001194.1.; MIM:603248; AY065994



(2) E16 (Lat1, Slc7A5)


Nucleotide


Genbank accession no. NM_003486


Genbank version no. NM_003486.5 GI:71979931


Genbank record update date: Jun. 27, 2012 12:06 PM


Polypeptide


Genbank accession no. NP_003477


Genbank version no. NP_003477.4 GI:71979932


Genbank record update date: Jun. 27, 2012 12:06 PM


CROSS REFERENCES




  • Biochem. Biophys. Res. Commun. 255 (2), 283-288 (1999), Nature 395 (6699):288-291 (1998), Gaugitsch, H. W., et 20 al (1992) J. Biol. Chem. 267 (16):11267-11273); WO2004/048938 (Example 2); WO2004/032842 (Example IV); WO2003/042661 (Claim 12); WO2003/016475 (Claim 1); WO2002/78524 (Example 2); WO2002/99074 (Claim 19; Page 127-129); WO2002/86443 (Claim 27; Pages 222, 393); WO2003/003906 (Claim 10; Page 293); WO2002/64798 (Claim 33; Page 93-95); WO2000/14228 (Claim 5; Page 133-136); US2003/224454 (FIG. 3); 25 WO2003/025138 (Claim 12; Page 150); NP_003477 solute carrier family 7 (cationic amino acid transporter, y+system), member 5/pid=NP_003477.3-Homo sapiens; MIM:600182; NM_015923.



(3) STEAP1 (Six Transmembrane Epithelial Antigen of Prostate)


Nucleotide


Genbank accession no. NM_012449


Genbank version no. NM_012449.2 GI:22027487


Genbank record update date: Sep. 9, 2012 02:57 PM


Polypeptide


Genbank accession no. NP_036581


Genbank version no. NP_036581.1 GI:9558759


Genbank record update date: Sep. 9, 2012 02:57 PM


CROSS REFERENCES




  • Cancer Res. 61 (15), 5857-5860 (2001), Hubert, R. S., et al (1999) Proc. Natl. Acad. Sci. U.S.A. 96 (25):14523-14528); WO2004/065577 (Claim 6); WO2004/027049 (FIG. 1L); EP1394274 (Example 11); WO2004/016225 (Claim 2); WO2003/042661 (Claim 12); US2003/157089 (Example 5); US2003/185830 (Example 5); US2003/064397 (FIG. 2); WO2002/89747 (Example 5; Page 618-619); WO2003/022995 (Example 9; FIG. 13A, 35 Example 53; Page 173, Example 2; FIG. 2A); six transmembrane epithelial antigen of the prostate; MIM:604415.



(4) 0772P (CA125, MUC16)


Nucleotide


Genbank accession no. AF361486


Genbank version no. AF361486.3 GI:34501466


Genbank record update date: Mar. 11, 2010 07:56 AM


Polypeptide


Genbank accession no. AAK74120


Genbank version no. AAK74120.3 GI:34501467


Genbank record update date: Mar. 11, 2010 07:56 AM


CROSS REFERENCES




  • J. Biol. Chem. 276 (29):27371-27375 (2001)); WO2004/045553 (Claim 14); WO2002/92836 (Claim 6; FIG. 12); WO2002/83866 (Claim 15; Page 116-121); US2003/124140 (Example 16); GI:34501467;



(5) MPF (MPF, MSLN, SMR, Megakaryocyte Potentiating Factor, Mesothelin)


Nucleotide


Genbank accession no. NM_005823


Genbank version no. NM_005823.5 GI:293651528


Genbank record update date: Sep. 2, 2012 01:47 PM


Polypeptide


Genbank accession no. NP_005814


Genbank version no. NP_005814.2 GI:53988378


Genbank record update date: Sep. 2, 2012 01:47 PM


CROSS REFERENCES



  • Yamaguchi, N., et al Biol. Chem. 269 (2), 805-808 (1994), Proc. Natl. Acad. Sci. U.S.A. 96 (20):11531-11536 (1999), Proc. Natl. Acad. Sci. U.S.A. 93 (1):136-140 (1996), J. Biol. Chem. 270 (37):21984-21990 (1995)); WO2003/101283 (Claim 14); (WO2002/102235 (Claim 13; Page 287-288); WO2002/101075 (Claim 4; Page 308-309); WO2002/71928 (Page 320-321); WO94/10312 (Page 52-57); IM:601051.



(6) Napi3b (NAPI-3B, NPTIIb, SLC34A2, Solute Carrier Family 34 (Sodium Phosphate), Member 2, Type II Sodium-Dependent Phosphate Transporter 3b)


Nucleotide


Genbank accession no. NM_006424


Genbank version no. NM_006424.2 GI:110611905


Genbank record update date: Jul. 22, 2012 03:39 PM


Polypeptide


Genbank accession no. NP_006415


Genbank version no. NP_006415.2 GI:110611906


Genbank record update date: Jul. 22, 2012 03:39 PM


CROSS REFERENCES




  • J. Biol. Chem. 277 (22):19665-19672 (2002), Genomics 62 (2):281-284 (1999), Field, J. A., et al (1999) Biochem. Biophys. Res. Commun. 258 (3):578-582); WO2004/022778 (Claim 2); EP1394274 (Example 11); WO2002/102235 (Claim 13; Page 326); EP0875569 (Claim 1; Page 17-19); WO2001/57188 (Claim 20; Page 329); WO2004/032842 (Example IV); WO2001/75177 (Claim 24; Page 139-140); MIM:604217.



(7) Sema 5b (FLJ10372, KIAA1445, Mm.42015, SEMA5B, SEMAG, Semaphorin 5b Hiog, 25 Sema Domain, Seven Thrombospondin Repeats (Type 1 and Type 1-Like), Transmembrane Domain™ and Short Cytoplasmic Domain, (Semaphorin) 5B)


Nucleotide


Genbank accession no. AB040878


Genbank version no. AB040878.1 GI:7959148


Genbank record update date: Aug. 2, 2006 05:40 PM


Polypeptide


Genbank accession no. BAA95969


Genbank version no. BAA95969.1 GI:7959149


Genbank record update date: Aug. 2, 2006 05:40 PM


CROSS REFERENCES



  • Nagase T., et al (2000) DNA Res. 7 (2):143-150); WO2004/000997 (Claim 1); WO2003/003984 (Claim 1); WO2002/06339 (Claim 1; Page 50); WO2001/88133 (Claim 1; Page 41-43, 48-58); WO2003/054152 (Claim 20); WO2003/101400 (Claim 11); Accession: 30 Q9P283; Genew; HGNC:10737



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


Nucleotide


Genbank accession no. AY358628


Genbank version no. AY358628.1 GI:37182377


Genbank record update date: Dec. 1, 2009 04:15 AM


Polypeptide


Genbank accession no. AAQ88991


Genbank version no. AAQ88991.1 GI:37182378


Genbank record update date: Dec. 1, 2009 04:15 AM


CROSS REFERENCES



  • Ross et al (2002) Cancer Res. 62:2546-2553; US2003/129192 (Claim 2); US2004/044180 (Claim 12); US2004/044179 (Claim 11); US2003/096961 (Claim 11); US2003/232056 (Example 5); WO2003/105758 16 (Claim 12); US2003/206918 (Example 5); EP1347046 (Claim 1); WO2003/025148 (Claim 20); GI:37182378.



(9) ETBR (Endothelin Type B Receptor)


Nucleotide


Genbank accession no. AY275463


Genbank version no. AY275463.1 GI:30526094


Genbank record update date: Mar. 11, 2010 02:26 AM


Polypeptide


Genbank accession no. AAP32295


Genbank version no. AAP32295.1 GI:30526095


Genbank record update date: Mar. 11, 2010 02:26 AM


CROSS REFERENCES



  • Nakamuta M., et al Biochem. Biophys. Res. Commun. 177, 34-39, 1991; Ogawa Y., et al Biochem. Biophys. Res. Commun. 178, 248-255, 1991; Arai H., et al Jpn. Circ. J. 56, 1303-1307, 1992; Arai H., et al J. Biol. Chem. 268, 3463-3470, 1993; Sakamoto A., Yanagisawa M., et al Biochem. Biophys. Res. Commun. 178, 656-663, 1991; Elshourbagy N. A., et al J. Biol. Chem. 268, 3873-3879, 1993; Haendler B., et al J. Cardiovasc. Pharmacol. 20, s1-S4, 1992; Tsutsumi M., et al Gene 228, 43-49, 1999; Strausberg R. L., et al Proc. Natl. Acad. Sci. U.S.A. 99, 16899-16903, 2002; Bourgeois C., et al J. Clin. Endocrinol. Metab. 82, 3116-3123, 1997; Okamoto Y., et al Biol. Chem. 272, 21589-21596, 1997; Verheij J. B., et al Am. J. Med. Genet. 108, 223-225, 2002; Hofstra R. M. W., et al Eur. J. Hum. Genet. 5, 180-185, 1997; Puffenberger E. G., et al Cell 79, 1257-1266, 1994; Attie T., et al, Hum. Mol. Genet. 4, 2407-2409, 1995; Auricchio A., et al Hum. Mol. Genet. 5:351-354, 1996; Amiel J., et al Hum. Mol. Genet. 5, 355-357, 1996; Hofstra R. M. W., et al Nat. Genet. 12, 445-447, 1996; Svensson P. J., et al Hum. Genet. 103, 145-148, 1998; Fuchs S., et al Mol. Med. 7, 115-124, 2001; Pingault V., et al (2002) Hum. Genet. 111, 198-206; WO2004/045516 (Claim 1); WO2004/048938 (Example 2); WO2004/040000 (Claim 151); WO2003/087768 (Claim 1); 20 WO2003/016475 (Claim 1); WO2003/016475 (Claim 1); WO2002/61087 (FIG. 1); WO2003/016494 (FIG. 6); WO2003/025138 (Claim 12; Page 144); WO2001/98351 (Claim 1: Page 124-125); EP0522868 (Claim 8; FIG. 2); WO2001/77172 (Claim 1; Page 297-299); US2003/109676; U.S. Pat. No. 6,518,404 (FIG. 3); U.S. Pat. No. 5,773,223 (Claim 1a; Col 31-34); WO2004/001004.



(10) MSG783 (RNF124, Hypothetical Protein FLJ20315)

  • Nucleotide
  • Genbank accession no. NM_017763
  • Genbank version no. NM_017763.4 GI:167830482
  • Genbank record update date: Jul. 22, 2012 12:34 AM
  • Polypeptide
  • Genbank accession no. NP_060233
  • Genbank version no. NP_060233.3 GI:56711322
  • Genbank record update date: Jul. 22, 2012 12:34 AM


CROSS REFERENCES



  • WO2003/104275 (Claim 1); WO2004/046342 (Example 2); WO2003/042661 (Claim 12); WO2003/083074 (Claim 14; Page 61); WO2003/018621 (Claim 1); WO2003/024392 (Claim 2; FIG. 93); WO2001/66689 (Example 6); LocusID:54894.



(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)


Nucleotide


Genbank accession no. AF455138


Genbank version no. AF455138.1 GI:22655487


Genbank record update date: Mar. 11, 2010 01:54 AM


Polypeptide


Genbank accession no. AAN04080


Genbank version no. AAN04080.1 GI:22655488


Genbank record update date: Mar. 11, 2010 01:54 AM


CROSS REFERENCES



  • Lab. Invest. 82 (11):1573-1582 (2002)); WO2003/087306; US2003/064397 (Claim 1; FIG. 1); WO2002/72596 (Claim 13; Page 54-55); WO2001/72962 (Claim 1; FIG. 4B); WO2003/104270 (Claim 11); WO2003/104270 (Claim 16); US2004/005598 (Claim 22); WO2003/042661 (Claim 12); US2003/060612 (Claim 12; FIG. 10); WO2002/26822 (Claim 23; FIG. 2); WO2002/16429 (Claim 12; FIG. 10); GI:22655488.



(12) TrpM4 (BR22450, FLJ20041, TRPM4, TRPM4B, Transient Receptor Potential Cation 5 Channel, Subfamily M, Member 4)


Nucleotide


Genbank accession no. NM_017636


Genbank version no. NM_017636.3 GI:304766649


Genbank record update date: Jun. 29, 2012 11:27 AM


Polypeptide


Genbank accession no. NP_060106


Genbank version no. NP_060106.2 GI:21314671


Genbank record update date: Jun. 29, 2012 11:27 AM


CROSS REFERENCES



  • Xu, X. Z., et al Proc. Natl. Acad. Sci. U.S.A. 98 (19):10692-10697 (2001), Cell 109 (3):397-407 (2002), J. Biol. Chem. 278 (33):30813-30820 (2003)); US2003/143557 (Claim 4); WO2000/40614 (Claim 14; Page 100-103); WO2002/10382 (Claim 1; FIG. 9A); WO2003/042661 (Claim 12); WO2002/30268 (Claim 27; Page 391); US2003/219806 (Claim 4); WO2001/62794 (Claim 14; FIG. 1A-D); MIM:606936.



(13) CRIPTO (CR, CR1, CRGF, CRIPTO, TDGF1, Teratocarcinoma-Derived Growth Factor)


Nucleotide


Genbank accession no. NM_003212


Genbank version no. NM_003212.3 GI:292494881


Genbank record update date: Sep. 23, 2012 02:27 PM


Polypeptide


Genbank accession no. NP_003203


Genbank version no. NP_003203.1 GI:4507425


Genbank record update date: Sep. 23, 2012 02:27 PM


CROSS REFERENCES



  • Ciccodicola, A., et al EMBO J. 8 (7):1987-1991 (1989), Am. J. Hum. Genet. 49 (3):555-565 (1991)); US2003/224411 (Claim 1); WO2003/083041 (Example 1); WO2003/034984 (Claim 12); WO2002/88170 (Claim 2; Page 52-53); WO2003/024392 (Claim 2; FIG. 58); WO2002/16413 (Claim 1; Page 94-95, 105); WO2002/22808 (Claim 2; FIG. 1); U.S. Pat. No. 5,854,399 (Example 2; Col 17-18); U.S. Pat. No. 5,792,616 (FIG. 2); MIM:187395.



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


Nucleotide


Genbank accession no M26004


Genbank version no. M26004.1 GI:181939


Genbank record update date: Jun. 23, 2010 08:47 AM


Polypeptide


Genbank accession no. AAA35786


Genbank version no. AAA35786.1 GI:181940


Genbank record update date: Jun. 23, 2010 08:47 AM


CROSS REFERENCES



  • Fujisaku et al (1989) J. Biol. Chem. 264 (4):2118-2125); Weis J. J., et al J. Exp. Med. 167, 1047-1066, 1988; Moore M., et al Proc. Natl. Acad. Sci. U.S.A. 84, 9194-9198, 1987; Barel M., et al Mol. Immunol. 35, 1025-1031, 1998; Weis J. J., et al Proc. Natl. Acad. Sci. U.S.A. 83, 5639-5643, 1986; Sinha S. K., et al (1993) J. Immunol. 150, 5311-5320; WO2004/045520 (Example 4); US2004/005538 (Example 1); WO2003/062401 (Claim 9); WO2004/045520 (Example 4); WO91/02536 (FIGS. 9.1-9.9); WO2004/020595 (Claim 1); Accession: P20023; Q13866; Q14212; EMBL; M26004; AAA35786.1.



(15) CD79b (CD79B, CD79(3, IGb (Immunoglobulin-Associated Beta), B29)


Nucleotide


Genbank accession no NM_000626


Genbank version no. NM_000626.2 GI:90193589


Genbank record update date: Jun. 26, 2012 01:53 PM


Polypeptide


Genbank accession no. NP_000617


Genbank version no. NP 000617.1 GI:11038674


Genbank record update date: Jun. 26, 2012 01:53 PM


CROSS REFERENCES




  • Proc. Natl. Acad. Sci. U.S.A. (2003) 100 (7):4126-4131, Blood (2002) 100 (9):3068-3076, Muller et al (1992) Eur. J. Immunol. 22 (6):1621-1625); WO2004/016225 (claim 2, FIG. 140); WO2003/087768, US2004/101874 (claim 1, page 102); WO2003/062401 (claim 9); WO2002/78524 (Example 2); US2002/150573 (claim 35 5, page 15); U.S. Pat. No. 5,644,033; WO2003/048202 (claim 1, pages 306 and 309); WO 99/58658, U.S. Pat. No. 6,534,482 (claim 13, FIG. 17A/B); WO2000/55351 (claim 11, pages 1145-1146); MIM:147245



(16) FcRH2 (IFGP4, IRTA4, SPAP1A (SH2 domain containing phosphatase anchor protein 51a), SPAP1B, SPAP1C)


Nucleotide


Genbank accession no NM_030764


Genbank version no. NM_030764.3 GI:227430280


Genbank record update date: Jun. 30, 2012 12:30 AM


Polypeptide


Genbank accession no. NP_110391


Genbank version no. NP_110391.2 GI:19923629


Genbank record update date: Jun. 30, 2012 12:30 AM


CROSS REFERENCES



  • AY358130); Genome Res. 13 (10):2265-2270 (2003), Immunogenetics 54 (2):87-95 (2002), Blood 99 (8):2662-2669 (2002), Proc. Natl. Acad. Sci. U.S.A. 98 (17):9772-9777 (2001), Xu, M. J., et al (2001) Biochem. Biophys. Res. Commun. 280 (3):768-775; WO2004/016225 (Claim 2); WO2003/077836; WO2001/38490 (Claim 5; FIG. 18D-1-18D-2); WO2003/097803 (Claim 12); 10 WO2003/089624 (Claim 25); MIM:606509.



(17) HER2 (ErbB2)


Nucleotide


Genbank accession no M11730


Genbank version no. M11730.1 GI:183986


Genbank record update date: Jun. 23, 2010 08:47 AM


Polypeptide


Genbank accession no. AAA75493


Genbank version no. AAA75493.1 GI:306840


Genbank record update date: Jun. 23, 2010 08:47 AM


CROSS REFERENCES



  • Coussens L., et al Science (1985) 230(4730):1132-1139); Yamamoto T., et al Nature 319, 230-234, 1986; Semba K., et al Proc. Natl. Acad. Sci. U.S.A. 82, 6497-6501, 1985; Swiercz J. M., et al J. Cell Biol. 165, 869-880, 2004; Kuhns J. J., et al J. Biol. Chem. 274, 36422-36427, 1999; Cho H.-S., et al Nature 421, 756-760, 2003; Ehsani A., et al (1993) Genomics 15, 426-429; WO2004/048938 (Example 2); WO2004/027049 (FIG. 11); WO2004/009622; WO2003/081210; WO2003/089904 (Claim 9); WO2003/016475 (Claim 1); US2003/118592; WO2003/008537 (Claim 1); WO2003/055439 (Claim 29; FIG. 1A-B); WO2003/025228 (Claim 37; FIG. 5C); 20 WO2002/22636 (Example 13; Page 95-107); WO2002/12341 (Claim 68; FIG. 7); WO2002/13847 (Page 71-74); WO2002/14503 (Page 114-117); WO2001/53463 (Claim 2; Page 41-46); WO2001/41787 (Page 15); WO2000/44899 (Claim 52; FIG. 7); WO2000/20579 (Claim 3; FIG. 2); U.S. Pat. No. 5,869,445 (Claim 3; Col 31-38); WO9630514 (Claim 2; Page 56-61); EP1439393 (Claim 7); WO2004/043361 (Claim 7); WO2004/022709; WO2001/00244 25 (Example 3; FIG. 4); Accession: P04626; EMBL; M11767; AAA35808.1. EMBL; M11761; AAA35808.1



Antibodies


Abbott: US20110177095

    • For example, an antibody comprising CDRs having overall at least 80% sequence identity to CDRs having amino acid sequences of SEQ ID NO:3 (CDR-H1), SEQ ID NO:4 (CDR-H2), SEQ ID NO:5 (CDR-H3), SEQ ID NO:104 and/or SEQ ID NO:6 (CDR-L1), SEQ ID NO:7 (CDR-L2), and SEQ ID NO:8 (CDR-L3), wherein the anti-HER2 antibody or anti-HER2 binding fragment has reduced immunogenicity as compared to an antibody having a VH of SEQ ID NO:1 and a VL of SEQ ID NO:2.


Biogen: US20100119511

    • For example, ATCC accession numbers: PTA-10355, PTA-10356, PTA-10357, PTA-10358
    • For example, a purified antibody molecule that binds to HER2 comprising a all six CDR's from an antibody selected from the group consisting of BIIB71F10 (SEQ ID NOs:11, 13), BIIB69A09 (SEQ ID NOs:15, 17); BIIB67F10 (SEQ ID NOs:19, 21); BIIB67F11 (SEQ ID NOs:23, 25), BIIB66A12 (SEQ ID NOs:27, 29), BIIB66C01 (SEQ ID NOs:31, 33), BIIB65C10 (SEQ ID NOs:35, 37), BIIB65H09 (SEQ ID NOs:39, 41) and B11B65B03 (SEQ ID NOs:43, 45), or CDRs which are identical or which have no more than two alterations from said CDRs.


Herceptin (Genentech)—U.S. Pat. No. 6,054,297; ATCC accession no. CRL-10463 (Genentech)


Pertuzumab (Genentech)

    • US20110117097
      • for example, see SEQ IDs No. 15&16, SEQ IDs No. 17&18, SEQ IDs No. 23&24 & ATCC accession numbers HB-12215, HB-12216, CRL 10463, HB-12697.
    • US20090285837
    • US20090202546
      • for example, ATCC accession numbers: HB-12215, HB-12216, CRL 10463, HB-12698.
    • US20060088523
      • for example, ATCC accession numbers: HB-12215, HB-12216
      • for example, an antibody comprising the variable light and variable heavy amino acid sequences in SEQ ID Nos. 3 and 4, respectively.
      • for example, an antibody comprising a light chain amino acid sequence selected from SEQ ID No. 15 and 23, and a heavy chain amino acid sequence selected from SEQ ID No. 16 and 24
    • US20060018899
      • for example, ATCC accession numbers: (7C2) HB-12215, (7F3) HB-12216, (4D5) CRL-10463, (2C4) HB-12697.
      • for example, an antibody comprising the amino acid sequence in SEQ ID No. 23, or a deamidated and/or oxidized variant thereof.
    • US2011/0159014
      • for example, an antibody having a light chain variable domain comprising the hypervariable regions of SEQ ID NO: 1”.
      • For example, an antibody having a heavy chain variable domain comprising the hypervariable regions of SEQ ID NO: 2.
    • US20090187007


Glycotope: TrasGEX antibody glycotope.com/pipeline

    • For example, see International Joint Cancer Institute and Changhai Hospital Cancer Cent: HMTI-Fc Ab—Gao J., et al BMB Rep. 2009 Oct. 31; 42(10):636-41.


Symphogen: US20110217305


Union Stem Cell &Gene Engineering, China—Liu H Q., et al Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 2010 May; 26(5):456-8.


(18) NCA (CEACAM6)


Nucleotide


Genbank accession no M18728


Genbank version no. M18728.1 GI:189084


Genbank record update date: Jun. 23, 2010 08:48 AM


Polypeptide


Genbank accession no. AAA59907


Genbank version no. AAA59907.1 GI:189085


Genbank record update date: Jun. 23, 2010 08:48 AM


CROSS REFERENCES



  • Barnett T., et al Genomics 3, 59-66, 1988; Tawaragi Y., et al Biochem. Biophys. Res. Commun. 150, 89-96, 1988; Strausberg R. L., et al Proc. Natl. Acad. Sci. U.S.A. 99:16899-16903, 2002; WO2004/063709; EP1439393 (Claim 7); WO2004/044178 (Example 4); WO2004/031238; WO2003/042661 (Claim 12); WO2002/78524 (Example 2); WO2002/86443 (Claim 27; Page 427); WO2002/60317 (Claim 2); Accession: P40199; Q14920; EMBL; M29541; AAA59915.1.



EMBL; M18728.


(19) MDP (DPEP1)


Nucleotide


Genbank accession no BC017023


Genbank version no. BC017023.1 GI:16877538


Genbank record update date: Mar. 6, 2012 01:00 PM


Polypeptide


Genbank accession no. AAH17023


Genbank version no. AAH17023.1 GI:16877539


Genbank record update date: Mar. 6, 2012 01:00 PM


CROSS REFERENCES




  • Proc. Natl. Acad. Sci. U.S.A. 99 (26):16899-16903 (2002)); WO2003/016475 (Claim 1); WO2002/64798 (Claim 33; Page 85-87); JP05003790 (FIG. 6-8); WO99/46284 (FIG. 9); MIM:179780.



(20) IL20R-alpha (IL20Ra, ZCYTOR7)


Nucleotide


Genbank accession no AF184971


Genbank version no. AF184971.1 GI:6013324


Genbank record update date: Mar. 10, 2010 10:00 PM


Polypeptide


Genbank accession no. AAF01320


Genbank version no. AAF01320.1 GI:6013325


Genbank record update date: Mar. 10, 2010 10:00 PM


CROSS REFERENCES



  • Clark H. F., et al Genome Res. 13, 2265-2270, 2003; Mungall A. J., et al Nature 425, 805-811, 2003; Blumberg H., et al Cell 104, 9-19, 2001; Dumoutier L., et al J. Immunol. 167, 3545-3549, 2001; Parrish-Novak J., et al J. Biol. Chem. 277, 47517-47523, 2002; Pletnev S., et al (2003) 10 Biochemistry 42:12617-12624; Sheikh F., et al (2004) J. Immunol. 172, 2006-2010; EP1394274 (Example 11); US2004/005320 (Example 5); WO2003/029262 (Page 74-75); WO2003/002717 (Claim 2; Page 63); WO2002/22153 (Page 45-47); US2002/042366 (Page 20-21); WO2001/46261 (Page 57-59); WO2001/46232 (Page 63-65); WO98/37193 (Claim 1; Page 55-59); Accession: Q9UHF4; Q6UWA9; Q96SH8; EMBL; AF184971; AAF01320.1.



(21) Brevican (BCAN, BEHAB)


Nucleotide


Genbank accession no AF229053


Genbank version no. AF229053.1 GI:10798902


Genbank record update date: Mar. 11, 2010 12:58 AM


Polypeptide


Genbank accession no. AAG23135


Genbank version no. AAG23135.1 GI:10798903


Genbank record update date: Mar. 11, 2010 12:58 AM


CROSS REFERENCES



  • Gary S. C., et al Gene 256, 139-147, 2000; Clark H. F., et al Genome Res. 13, 2265-2270, 2003; Strausberg R. L., et al Proc. Natl. Acad. Sci. U.S.A. 99, 16899-16903, 2002; US2003/186372 (Claim 11); US2003/186373 (Claim 11); US2003/119131 (Claim 1; FIG. 52); US2003/119122 (Claim 1; FIG. 52); US2003/119126 (Claim 1); US2003/119121 (Claim 1; FIG. 52); US2003/119129 (Claim 1); US2003/119130 (Claim 1); US2003/119128 (Claim 1; FIG. 52); US2003/119125 (Claim 1); WO2003/016475 (Claim 1); WO2002/02634 (Claim 1)



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


Nucleotide


Genbank accession no NM_004442


Genbank version no. NM_004442.6 GI:111118979


Genbank record update date: Sep. 8, 2012 04:43 PM


Polypeptide


Genbank accession no. NP_004433


Genbank version no. NP 004433.2 GI:21396504


Genbank record update date: Sep. 8, 2012 04:43 PM


CROSS REFERENCES



  • Chan, J. and Watt, V. M., Oncogene 6 (6), 1057-1061 (1991) Oncogene 10 (5):897-905 (1995), Annu. Rev. Neurosci. 21:309-345 (1998), Int. Rev. Cytol. 196:177-244 (2000)); WO2003042661 (Claim 12); WO200053216 (Claim 1; Page 41); WO2004065576 (Claim 1); WO2004020583 (Claim 9); WO2003004529 (Page 128-132); WO200053216 (Claim 1; Page 42); MIM:600997.



(23) ASLG659 (B7 h)


Nucleotide


Genbank accession no. AX092328


Genbank version no. AX092328.1 GI:13444478


Genbank record update date: Jan. 26, 2011 07:37 AM


CROSS REFERENCES



  • US2004/0101899 (Claim 2); WO2003104399 (Claim 11); WO2004000221 (FIG. 3); US2003/165504 (Claim 1); US2003/124140 (Example 2); US2003/065143 (FIG. 60); WO2002/102235 (Claim 13; Page 299); US2003/091580 (Example 2); WO2002/10187 (Claim 6; FIG. 10); WO2001/94641 (Claim 12; FIG. 7b); WO2002/02624 (Claim 13; FIG. 1A-1B); US2002/034749 (Claim 54; Page 45-46); WO2002/06317 (Example 2; Page 320-321, Claim 34; Page 321-322); WO2002/71928 (Page 468-469); WO2002/02587 (Example 1; FIG. 1); WO2001/40269 (Example 3; Pages 190-192); WO2000/36107 (Example 2; Page 205-207); WO2004/053079 (Claim 12); WO2003/004989 (Claim 1); WO2002/71928 (Page 233-234, 452-453); WO 01/16318.



(24) PSCA (Prostate stem cell antigen precursor)


Nucleotide


Genbank accession no AJ297436


Genbank version no. AJ297436.1 GI:9367211


Genbank record update date: Feb. 1, 2011 11:25 AM


Polypeptide


Genbank accession no. CAB97347


Genbank version no. CAB97347.1 GI:9367212


Genbank record update date: Feb. 1, 2011 11:25 AM


CROSS REFERENCES



  • Reiter R. E., et al Proc. Natl. Acad. Sci. U.S.A. 95, 1735-1740, 1998; Gu Z., et al Oncogene 19, 1288-1296, 2000; Biochem. Biophys. Res. Commun. (2000) 275(3):783-788; WO2004/022709; EP1394274 (Example 11); US2004/018553 (Claim 17); WO2003/008537 (Claim 1); WO2002/81646 (Claim 1; Page 164); WO2003/003906 (Claim 10; Page 288); WO2001/40309 (Example 1; FIG. 17); US2001/055751 (Example 1; FIG. 1b); WO2000/32752 (Claim 18; FIG. 1); WO98/51805 (Claim 17; Page 97); WO98/51824 (Claim 10; Page 94); WO98/40403 (Claim 2; FIG. 1B); Accession: 043653; EMBL; AF043498; AAC39607.1



(25) GEDA


Nucleotide


Genbank accession no AY260763


Genbank version no. AY260763.1 GI:30102448


Genbank record update date: Mar. 11, 2010 02:24 AM


Polypeptide


Genbank accession no. AAP14954


Genbank version no. AAP14954.1 GI:30102449


Genbank record update date: Mar. 11, 2010 02:24 AM


CROSS REFERENCES



  • AP14954 lipoma HMGIC fusion-partnerlike protein/pid=AAP14954.1-Homo sapiens (human); WO2003/054152 (Claim 20); WO2003/000842 (Claim 1); WO2003/023013 (Example 3, Claim 20); US2003/194704 (Claim 45); GI:30102449;



(26) BAFF-R (B Cell —Activating Factor Receptor, BLyS Receptor 3, BR3)


Nucleotide


Genbank accession no AF116456


Genbank version no. AF116456.1 GI:4585274


Genbank record update date: Mar. 10, 2010 09:44 PM


Polypeptide


Genbank accession no. AAD25356


Genbank version no. AAD25356.1 GI:4585275


Genbank record update date: Mar. 10, 2010 09:44 PM


CROSS REFERENCES



  • BAFF receptor/pid=NP_443177.1-Homo sapiens: Thompson, J. S., et al Science 293 (5537), 2108-2111 (2001); WO2004/058309; WO2004/011611; WO2003/045422 (Example; Page 32-33); WO2003/014294 (Claim 35; FIG. 6B); WO2003/035846 (Claim 70; Page 615-616); WO2002/94852 (Col 136-137); WO2002/38766 (Claim 3; Page 133); WO2002/24909 (Example 3; FIG. 3); MIM:606269; NP_443177.1; NM_052945_1; AF132600



(27) CD22 (B-Cell Receptor CD22-B Isoform, BL-CAM, Lyb-8, Lyb8, SIGLEC-2, FLJ22814)


Nucleotide


Genbank accession no AK026467


Genbank version no. AK026467.1 GI:10439337


Genbank record update date: Sep. 11, 2006 11:24 PM


Polypeptide


Genbank accession no. BAB15489


Genbank version no. BAB15489.1 GI:10439338


Genbank record update date: Sep. 11, 2006 11:24 PM


CROSS REFERENCES



  • Wilson et al (1991) J. Exp. Med. 173:137-146; WO2003/072036 (Claim 1; FIG. 1); IM:107266; NP_001762.1; NM_001771_1.



(27a) CD22 (CD22 molecule)


Nucleotide


Genbank accession no X52785


Genbank version no. X52785.1 GI:29778


Genbank record update date: Feb. 2, 2011 10:09 AM


Polypeptide


Genbank accession no. CAA36988


Genbank version no. CAA36988.1 GI:29779


Genbank record update date: Feb. 2, 2011 10:09 AM


CROSS REFERENCES



  • Stamenkovic I. et al., Nature 345 (6270), 74-77 (1990)??



Other Information


Official Symbol: CD22


Other Aliases: SIGLEC-2, SIGLEC2


Other Designations: B-cell receptor CD22; B-lymphocyte cell adhesion molecule; BL-


CAM; CD22 antigen; T-cell surface antigen Leu-14; sialic acid binding Ig-like lectin 2; sialic acid-binding Ig-like lectin 2


Antibodies


G5/44 (Inotuzumab): DiJoseph J F.,et al Cancer Immunol Immunother. 2005 January; 54(1):11-24.


Epratuzumab-Goldenberg D M., et al Expert Rev Anticancer Ther. 6(10): 1341-53, 2006.


(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 35 Molecules, Transduces a Signal Involved in B-Cell Differentiation), pl: 4.84, MW: 25028 TM: 2


[P] Gene Chromosome: 19q13.2).


Nucleotide


Genbank accession no NM_001783


Genbank version no. NM_001783.3 GI:90193587


Genbank record update date: Jun. 26, 2012 01:48 PM


Polypeptide


Genbank accession no. NP_001774


Genbank version no. NP_001774.1 GI:4502685


Genbank record update date: Jun. 26, 2012 01:48 PM


CROSS REFERENCES



  • WO2003/088808, US2003/0228319; WO2003/062401 (claim 9); US2002/150573 (claim 4, pages 13-14); WO99/58658 (claim 13, FIG. 16); WO92/07574 (FIG. 1); U.S. Pat. No. 5,644,033; Ha et al (1992) J. Immunol. 148(5):1526-1531; Müller et al (1992) Eur. J. Immunol. 22:1621-1625; Hashimoto et al (1994) Immunogenetics 40(4):287-295; Preud'homme et al (1992) Clin. Exp. 5 Immunol. 90(1):141-146; Yu et al (1992) J. Immunol. 148(2) 633-637; Sakaguchi et al (1988) EMBO J. 7(11):3457-3464



(29) CXCR5 (Burkitt's Lymphoma Receptor 1, a G Protein-Coupled Receptor that is Activated by the CXCL13 Chemokine, Functions in Lymphocyte Migration and Humoral Defense, Plays a 10 Role in HIV-2 Infection and Perhaps Development of AIDS, Lymphoma, Myeloma, and Leukemia); 372 aa, pl: 8.54 MW: 41959 TM: 7 [P] Gene Chromosome: 11q23.3,


Nucleotide


Genbank accession no NM_001716


Genbank version no. NM_001716.4 GI:342307092


Genbank record update date: Sep. 30, 2012 01:49 PM


Polypeptide


Genbank accession no. NP_001707


Genbank version no. NP_001707.1 GI:4502415


Genbank record update date: Sep. 30, 2012 01:49 PM


CROSS REFERENCES



  • WO2004/040000; WO2004/015426; US2003/105292 (Example 2); U.S. Pat. No. 6,555,339 (Example 2); WO2002/61087 (FIG. 1); WO2001/57188 (Claim 20, page 269); WO2001/72830 (pages 12-13); WO2000/22129 (Example 1, pages 152-153, Example 2, pages 254-256); WO99/28468 (claim 1, page 38); U.S. Pat. No. 5,440,021 (Example 2, col 49-52); WO94/28931 (pages 56-58); WO92/17497 (claim 7, FIG. 5); Dobner et al (1992) Eur. J. Immunol. 22:2795-2799; Barella et al (1995) Biochem. J. 309:773-779



(30) HLA-DOB (Beta Subunit of MHC Class II Molecule (La Antigen) that Binds Peptides and 20 Presents them to CD4+T Lymphocytes); 273 aa, pl: 6.56, MW: 30820.TM: 1 [P] Gene Chromosome: 6p21.3)


Nucleotide


Genbank accession no NM_002120


Genbank version no. NM_002120.3 GI:118402587


Genbank record update date: Sep. 8, 2012 04:46 PM


Polypeptide


Genbank accession no. NP_002111


Genbank version no. NP_002111.1 GI:4504403


Genbank record update date: Sep. 8, 2012 04:46 PM


CROSS REFERENCES



  • Tonnelle et al (1985) EMBO J. 4(11):2839-2847; Jonsson et al (1989) Immunogenetics 29(6):411-413; Beck et al (1992) J. Mol. Biol. 228:433-441; Strausberg et al (2002) Proc. Natl. Acad. Sci USA 99:16899-16903; Servenius et al (1987) J. Biol. Chem. 262:8759-8766; Beck et al (1996) J. Mol. Biol. 255:1-13; Naruse et al (2002) Tissue Antigens 59:512-519; WO99/58658 (claim 13, FIG. 15); U.S. Pat. No. 6,153,408 (Col 35-38); U.S. Pat. No. 5,976,551 (col 168-170); U.S. Pat. No. 6,011,146 (col 145-146); Kasahara et al (1989) Immunogenetics 30(1):66-68; Larhammar et al (1985) J. Biol. Chem. 260(26):14111-14119



(31) P2X5 (Purinergic Receptor P2X Ligand-Gated Ion Channel 5, an Ion Channel Gated by Extracellular ATP, May be Involved in Synaptic Transmission and Neurogenesis, Deficiency May Contribute to the Pathophysiology of Idiopathic Detrusor Instability); 422 Aa), pl: 7.63, MW: 47206 TM: 1 [P] Gene Chromosome: 17p13.3).


Nucleotide


Genbank accession no NM_002561


Genbank version no. NM_002561.3 GI:325197202


Genbank record update date: Jun. 27, 2012 12:41 AM


Polypeptide


Genbank accession no. NP_002552


Genbank version no. NP_002552.2 GI:28416933


Genbank record update date: Jun. 27, 2012 12:41 AM


CROSS REFERENCES



  • Le et al (1997) FEBS Lett. 418(1-2):195-199; WO2004/047749; WO2003/072035 (claim 10); Touchman et al (2000) Genome Res. 10:165-173; WO2002/22660 (claim 20); WO2003/093444 (claim 1); WO2003/087768 (claim 1); WO2003/029277 (page 82)



(32) CD72 (B-Cell Differentiation Antigen CD72, Lyb-2); 359 aa, pl: 8.66, MW: 40225, TM: 1 5 [P] Gene Chromosome: 9p13.3).


Nucleotide


Genbank accession no NM_001782


Genbank version no. NM_001782.2 GI:194018444


Genbank record update date: Jun. 26, 2012 01:43 PM


Polypeptide


Genbank accession no. NP_001773


Genbank version no. NP_001773.1 GI:4502683


Genbank record update date: Jun. 26, 2012 01:43 PM


CROSS REFERENCES



  • WO2004042346 (claim 65); WO2003/026493 (pages 51-52, 57-58); WO2000/75655 (pages 105-106); Von Hoegen et al (1990) J. Immunol. 144(12):4870-4877; Strausberg et al (2002) Proc. Natl. Acad. Sci USA 99:16899-16903.



(33) LY64 (Lymphocyte Antigen 64 (RP105), Type I Membrane Protein of the Leucine Rich Repeat (LRR) Family, Regulates B-Cell Activation and Apoptosis, Loss of Function is Associated with Increased Disease Activity in Patients with Systemic Lupus Erythematosis); 661 aa, pl: 6.20, MW: 74147 TM: 1 [P] Gene Chromosome: 5q12).


Nucleotide


Genbank accession no NM_005582


Genbank version no. NM_005582.2 GI:167555126


Genbank record update date: Sep. 2, 2012 01:50 PM


Polypeptide


Genbank accession no. NP_005573


Genbank version no. NP_005573.2 GI:167555127


Genbank record update date: Sep. 2, 2012 01:50 PM


CROSS REFERENCES



  • US2002/193567; WO97/07198 (claim 11, pages 39-42); Miura et al (1996) Genomics 38(3):299-304; Miura et al (1998) Blood 92:2815-2822; WO2003/083047; WO97/44452 (claim 8, pages 57-61); WO2000/12130 (pages 24-26).



(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, pl: 5.28, MW: 46925 TM: 1 [P] Gene Chromosome: 1q21-1q22)


Nucleotide


Genbank accession no NM_052938


Genbank version no. NM_052938.4 GI:226958543


Genbank record update date: Sep. 2, 2012 01:43 PM


Polypeptide


Genbank accession no. NP_443170


Genbank version no. NP_443170.1 GI:16418419


Genbank record update date: Sep. 2, 2012 01:43 PM


CROSS REFERENCES

WO2003/077836; WO2001/38490 (claim 6, FIG. 18E-1-18-E-2); Davis et al (2001) Proc. Natl. Acad. Sci USA 98(17):9772-9777; WO2003/089624 (claim 8); EP1347046 (claim 1); WO2003/089624 (claim 7).


(35) IRTA2 (Immunoglobulin Superfamily Receptor Translocation Associated 2, a Putative Immunoreceptor with Possible Roles in B Cell Development and Lymphomagenesis; Deregulation of the Gene by Translocation Occurs in Some B Cell Malignancies); 977 aa, pl: 6.88, MW: 106468, TM: 1 [P] Gene Chromosome: 1q21)


Nucleotide


Genbank accession no AF343662


Genbank version no. AF343662.1 GI:13591709


Genbank record update date: Mar. 11, 2010 01:16 AM


Polypeptide


Genbank accession no. AAK31325


Genbank version no. AAK31325.1 GI:13591710


Genbank record update date: Mar. 11, 2010 01:16 AM


CROSS REFERENCES



  • AF343663, AF343664, AF343665, AF369794, AF397453, AK090423, AK090475, AL834187, AY358085; Mouse:AK089756, AY158090, AY506558: NP_112571.1; WO2003/024392 (claim 2, FIG. 97); Nakayama et al (2000) Biochem. Biophys. Res. Commun. 277(1):124-127; WO2003/077836; WO2001/38490 (claim 3, FIG. 18B-1-18B-2).



(36) TENB2 (TMEFF2, Tomoregulin, TPEF, HPP1, TR, Putative Transmembrane 35 Proteoglycan, Related to the EGF/Heregulin Family of Growth Factors and Follistatin); 374 aa)


Nucleotide


Genbank accession no AF179274


Genbank version no. AF179274.2 GI:12280939


Genbank record update date: Mar. 11, 2010 01:05 AM


Polypeptide


Genbank accession no. AAD55776


Genbank version no. AAD55776.2 GI:12280940


Genbank record update date: Mar. 11, 2010 01:05 AM


CROSS REFERENCES



  • NCBI Accession: AAD55776, AAF91397, AAG49451, NCBI RefSeq: NP_057276; NCBI Gene: 23671; OMIM: 605734; SwissProt Q9UIK5; AY358907, CAF85723, CQ782436; WO2004/074320; JP2004113151; WO2003/042661; WO2003/009814; EP1295944 (pages 69-70); WO2002/30268 (page 329); WO2001/90304; US2004/249130; US2004/022727; WO2004/063355; US2004/197325; US2003/232350; US2004/005563; US2003/124579; Horie et al (2000) Genomics 67:146-152; Uchida et al (1999) Biochem. Biophys. Res. Commun. 266:593-602; Liang et al (2000) Cancer Res. 60:4907-12; Glynne-Jones et al (2001) Int J Cancer. October 15; 94(2):178-84.



(37) PSMA—FOLH1 (Folate Hydrolase (Prostate-Specific Membrane Antigen) 1)


Nucleotide


Genbank accession no M99487


Genbank version no. M99487.1 GI:190663


Genbank record update date: Jun. 23, 2010 08:48 AM


Polypeptide


Genbank accession no. AAA60209


Genbank version no. AAA60209.1 GI:190664


Genbank record update date: Jun. 23, 2010 08:48 AM


CROSS REFERENCES



  • Israeli R. S., et al Cancer Res. 53 (2), 227-230 (1993)



Other Information


Official Symbol: FOLH1


Other Aliases: GIG27, FGCP, FOLH, GCP2, GCPII, NAALAD1, NAALAdase, PSM, PSMA, mGCP


Other Designations: N-acetylated alpha-linked acidic dipeptidase 1; N-acetylated-alpha-linked acidic dipeptidase I; NAALADase I; cell growth-inhibiting gene 27 protein; folylpoly-gamma-glutamate carboxypeptidase; glutamate carboxylase II; glutamate carboxypeptidase 2; glutamate carboxypeptidase II; membrane glutamate carboxypeptidase; prostate specific membrane antigen variant F; pteroylpoly-gamma-glutamate carboxypeptidase


Antibodies


U.S. Pat. No. 7,666,425:


Antibodies produces by Hybridomas having the following ATCC references:ATCC accession No. HB-12101, ATCC accession No. HB-12109, ATCC accession No. HB-12127 and ATCC accession No. HB-12126.


Proscan: a monoclonal antibody selected from the group consisting of 8H12, 3E11, 17G1, 29B4, 30C1 and 20F2 (U.S. Pat. No. 7,811,564; Moffett S., et al Hybridoma (Larchmt). 2007 December; 26(6):363-72).


Cytogen: monoclonal antibodies 7E11-C5 (ATCC accession No. HB 10494) and 9H10-A4 (ATCC accession No. HB11430)—U.S. Pat. No. 5,763,202


GlycoMimetics: NUH2-ATCC accession No. HB 9762 (U.S. Pat. No. 7,135,301) Human Genome Science: HPRAJ70-ATCC accession No. 97131 (U.S. Pat. No. 6,824,993); Amino acid sequence encoded by the cDNA clone (HPRAJ70) deposited as American Type Culture Collection (“ATCC”) Deposit No. 97131


Medarex: Anti-PSMA antibodies that lack fucosyl residues—U.S. Pat. No. 7,875,278


Mouse anti-PSMA antibodies include the 3F5.4G6, 3D7.1.1, 4E10-1.14, 3E11, 4D8, 3E6, 3C9, 2C7, 1G3, 3C4, 3C6, 4D4, 1G9, 5C8B9, 3G6, 4C8B9, and monoclonal antibodies. Hybridomas secreting 3F5.4G6, 3D7.1.1, 4E10-1.14, 3E11, 4D8, 3E6, 3C9, 2C7, 1G3, 3C4, 3C6, 4D4, 1G9, 5C8B9, 3G6 or 4C8B9 have been publicly deposited and are described in U.S. Pat. No. 6,159,508. Relevant hybridomas have been publicly deposited and are described in U.S. Pat. No. 6,107,090. Moreover, humanized anti-PSMA antibodies, including a humanized version of J591, are described in further detail in PCT Publication WO 02/098897.


Other mouse anti-human PSMA antibodies have been described in the art, such as mAb 107-1A4 (Wang, S. et al. (2001) Int. J. Cancer 92:871-876) and mAb 2C9 (Kato, K. et al. (2003) Int. J. Urol. 10:439-444).


Examples of human anti-PSMA monoclonal antibodies include the 4A3, 7F12, 8C12, 8A11, 16F9, 2A10, 2C6, 2F5 and 1C3 antibodies, isolated and structurally characterized as originally described in PCT Publications WO 01/09192 and WO 03/064606 and in U.S. Provisional Application Ser. No. 60/654,125, entitled “Human Monoclonal Antibodies to Prostate Specific Membrane Antigen (PSMA)”, filed on Feb. 18, 2005. The V.sub.H amino acid sequences of 4A3, 7F12, 8C12, 8A11, 16F9, 2A10, 2C6, 2F5 and 1C3 are shown in SEQ ID NOs: 1-9, respectively. The V.sub.L amino acid sequences of 4A3, 7F12, 8C12, 8A11, 16F9, 2A10, 2C6, 2F5 and 1C3 are shown in SEQ ID NOs: 10-18, respectively.


Other human anti-PSMA antibodies include the antibodies disclosed in PCT Publication WO 03/034903 and US Application No. 2004/0033229.


NW Biotherapeutics: A hybridoma cell line selected from the group consisting of 3F5.4G6 having ATCC accession number HB12060, 3D7-1.1. having ATCC accession number HB12309, 4E10-1.14 having ATCC accession number HB12310, 3E11 (ATCC HB12488), 4D8 (ATCC HB12487), 3E6 (ATCC HB12486), 3C9 (ATCC HB12484), 2C7 (ATCC HB12490), 1G3 (ATCC HB12489), 3C4 (ATCC HB12494), 3C6 (ATCC HB12491), 4D4 (ATCC HB12493), 1G9 (ATCC HB12495), 5C8B9 (ATCC HB12492) and 3G6 (ATCC HB12485)—see U.S. Pat. No. 6,150,508


PSMA Development Company/Progenics/Cytogen—Seattle Genetics: mAb 3.9, produced by the hybridoma deposited under ATCC Accession No. PTA-3258 or mAb 10.3, produced by the hybridoma deposited under ATCC Accession No. PTA-3347—U.S. Pat. No. 7,850,971


PSMA Development Company—Compositions of PSMA antibodies (US 20080286284, Table 1)

    • This application is a divisional of U.S. patent application Ser. No. 10/395,894, filed on Mar. 21, 2003 (U.S. Pat. No. 7,850,971)


University Hospital Freiburg, Germany—mAbs 3/A12, 3/E7, and 3/F11 (Wolf P., et al Prostate. 2010 Apr. 1; 70(5):562-9).


(38) SST (Somatostatin Receptor; Note that there Are5 Subtypes)


(38.1) SSTR2 (Somatostatin Receptor 2)


Nucleotide


Genbank accession no NM_001050


Genbank version no. NM_001050.2 GI:44890054


Genbank record update date: Aug. 19, 2012 01:37 PM


Polypeptide


Genbank accession no. NP_001041


Genbank version no. NP_001041.1 GI:4557859


Genbank record update date: Aug. 19, 2012 01:37 PM


CROSS REFERENCES



  • Yamada Y., et al Proc. Natl. Acad. Sci. U.S.A. 89 (1), 251-255 (1992); Susini C., et al Ann Oncol. 2006 December; 17(12):1733-42



Other Information


Official Symbol: SSTR2


Other Designations: SRIF-1; SS2R; somatostatin receptor type 2


(38.2) SSTR5 (Somatostatin Receptor 5)


Nucleotide


Genbank accession no D16827


Genbank version no. D16827.1 GI:487683


Genbank record update date: Aug. 1, 2006 12:45 PM


Polypeptide


Genbank accession no. BAA04107


Genbank version no. BAA04107.1 GI:487684


Genbank record update date: Aug. 1, 2006 12:45 PM


CROSS REFERENCES



  • Yamada,Y., et al Biochem. Biophys. Res. Commun. 195 (2), 844-852 (1993)



Other Information


Official Symbol: SSTR5


Other Aliases: SS-5-R


Other Designations: Somatostatin receptor subtype 5; somatostatin receptor type 5


(38.3) SSTR1


(38.4) SSTR3


(38.5) SSTR4


AvB6—Both subunits (39+40)


(39) ITGAV (Integrin, Alpha V;


Nucleotide


Genbank accession no M14648 J02826 M18365


Genbank version no. M14648.1 GI:340306


Genbank record update date: Jun. 23, 2010 08:56 AM


Polypeptide


Genbank accession no. AAA36808


Genbank version no. AAA36808.1 GI:340307


Genbank record update date: Jun. 23, 2010 08:56 AM


CROSS REFERENCES



  • Suzuki S., et al Proc. Natl. Acad. Sci. U.S.A. 83 (22), 8614-8618 (1986)



Other Information


Official Symbol: ITGAV


Other Aliases: CD51, MSK8, VNRA, VTNR


Other Designations: antigen identified by monoclonal antibody L230; integrin alpha-V; integrin alphaVbeta3; integrin, alpha V (vitronectin receptor, alpha polypeptide, antigen CD51); vitronectin receptor subunit alpha


(40) ITGB6 (Integrin, Beta 6)


Nucleotide


Genbank accession no NM_000888


Genbank version no. NM_000888.3 GI:9966771


Genbank record update date: Jun. 27, 2012 12:46 AM


Polypeptide


Genbank accession no. NP_000879


Genbank version no. NP_000879.2 GI:9625002


Genbank record update date: Jun. 27, 2012 12:46 AM


CROSS REFERENCES



  • Sheppard D. J., et al Biol. Chem. 265 (20), 11502-11507 (1990)



Other Information


Official Symbol: ITGB6


Other Designations: integrin beta-6


Antibodies


Biogen: U.S. Pat. No. 7,943,742—Hybridoma clones 6.3G9 and 6.8G6 were deposited with the ATCC, accession numbers ATCC PTA-3649 and -3645, respectively.


Biogen: U.S. Pat. No. 7,465,449—In some embodiments, the antibody comprises the same heavy and light chain polypeptide sequences as an antibody produced by hybridoma 6.1A8, 6.3G9, 6.8G6, 6.2B1, 6.2B10, 6.2A1, 6.2E5, 7.1G10, 7.7G5, or 7.1C5.


Centocor (J&J): U.S. Pat. Nos. 7,550,142; 7,163,681

    • For example in U.S. Pat. No. 7,550,142— an antibody having human heavy chain and human light chain variable regions comprising the amino acid sequences shown in SEQ ID NO: 7 and SEQ ID NO: 8.


Seattle Genetics: 15H3 (Ryan M C., et al Cancer Res Apr. 15, 2012; 72(8 Supplement): 4630)


(41) CEACAM5 (Carcinoembryonic Antigen-Related Cell Adhesion Molecule 5)


Nucleotide


Genbank accession no M17303


Genbank version no. M17303.1 GI:178676


Genbank record update date: Jun. 23, 2010 08:47 AM


Polypeptide


Genbank accession no. AAB59513


Genbank version no. AAB59513.1 GI:178677


Genbank record update date: Jun. 23, 2010 08:47 AM


CROSS REFERENCES



  • Beauchemin N., et al Mol. Cell. Biol. 7 (9), 3221-3230 (1987)



Other Information


Official Symbol: CEACAM5


Other Aliases: CD66e, CEA


Other Designations: meconium antigen 100


Antibodies


AstraZeneca-MedImmune:US 20100330103; US20080057063;

    • US20020142359
      • for example an antibody having complementarity determining regions (CDRs) with the following sequences: heavy chain; CDR1-DNYMH, CDR2-WIDPENGDTE YAPKFRG, CDR3-LIYAGYLAMD Y; and light chain CDR1-SASSSVTYMH, CDR2-STSNLAS, CDR3-QQRSTYPLT.
      • Hybridoma 806.077 deposited as European Collection of Cell Cultures (ECACC) deposit no. 96022936.


Research Corporation Technologies, Inc.: U.S. Pat. No. 5,047,507


Bayer Corporation: U.S. Pat. No. 6,013,772


BioAlliance: U.S. Pat. Nos. 7,982,017; 7,674,605

    • U.S. Pat. No. 7,674,605
      • an antibody comprising the heavy chain variable region sequence from the amino acid sequence of SEQ ID NO: 1, and the light chain variable region sequence from the amino acid sequence of SEQ ID NO:2.
    • an antibody comprising the heavy chain variable region sequence from the amino acid sequence of SEQ ID NO:5, and the light chain variable region sequence from the amino acid sequence of SEQ ID NO:6.


Celltech Therapeutics Limited: U.S. Pat. No. 5,877,293


The Dow Chemical Company: U.S. Pat. Nos. 5,472,693; 6,417,337; 6,333,405

    • U.S. Pat. No. 5,472,693-for example, ATCC No. CRL-11215
    • U.S. Pat. No. 6,417,337-for example, ATCC CRL-12208
    • U.S. Pat. No. 6,333,405-for example, ATCC CRL-12208


Immunomedics, Inc: U.S. Pat. Nos. 7,534,431; 7,230,084; 7,300,644; 6,730,300;

    • US20110189085
      • an antibody having CDRs of the light chain variable region comprise: CDR1 comprises KASQDVGTSVA (SEQ ID NO: 20); CDR2 comprises WTSTRHT (SEQ ID NO: 21); and CDR3 comprises QQYSLYRS (SEQ ID NO: 22);
      • and the CDRs of the heavy chain variable region of said anti-CEA antibody comprise: CDR1 comprises TYWMS (SEQ ID NO: 23); CDR2 comprises EIHPDSSTINYAPSLKD (SEQ ID NO: 24); and CDR3 comprises LYFGFPWFAY (SEQ ID NO: 25).
    • US20100221175; US20090092598; US20070202044; US20110064653; US20090185974; US20080069775.


(42) MET (Met Proto-Oncogene; Hepatocyte Growth Factor Receptor)


Nucleotide


Genbank accession no M35073


Genbank version no. M35073.1 GI:187553


Genbank record update date: Mar. 6, 2012 11:12 AM


Polypeptide


Genbank accession no. AAA59589


Genbank version no. AAA59589.1 GI:553531


Genbank record update date: Mar. 6, 2012 11:12 AM


CROSS REFERENCES



  • Dean M., et al Nature 318 (6044), 385-388 (1985)



Other Information


Official Symbol: MET


Other Aliases: AUTS9, HGFR, RCCP2, c-Met


Other Designations: HGF receptor; HGF/SF receptor; SF receptor; hepatocyte growth factor receptor; met proto-oncogene tyrosine kinase; proto-oncogene c-Met; scatter factor receptor; tyrosine-protein kinase Met


Antibodies


Abgenix/Pfizer: US20100040629

    • for example, the antibody produced by hybridoma 13.3.2 having American Type Culture Collection (ATCC) accession number PTA-5026; the antibody produced by hybridoma 9.1.2 having ATCC accession number PTA-5027; the antibody produced by hybridoma 8.70.2 having ATCC accession number PTA-5028; or the antibody produced by hybridoma 6.90.3 having ATCC accession number PTA-5029.


Amgen/Pfizer: US20050054019

    • for example, an antibody comprising a heavy chain having the amino acid sequences set forth in SEQ ID NO: 2 where X2 is glutamate and X4 is serine and a light chain having the amino acid sequence set forth in SEQ ID NO: 4 where X8 is alanine, without the signal sequences; an antibody comprising a heavy chain having the amino acid sequences set forth in SEQ ID NO: 6 and a light chain having the amino acid sequence set forth in SEQ ID NO: 8, without the signal sequences; an antibody comprising a heavy chain having the amino acid sequences set forth in SEQ ID NO: 10 and a light chain having the amino acid sequence set forth in SEQ ID NO: 12, without the signal sequences; or an antibody comprising a heavy chain having the amino acid sequences set forth in SEQ ID NO: 14 and a light chain having the amino acid sequence set forth in SEQ ID NO: 16, without the signal sequences.


Agouron Pharmaceuticals (Now Pfizer): US20060035907


Eli Lilly: US20100129369


Genentech: U.S. Pat. No. 5,686,292; US20100028337; US20100016241; US20070129301; US20070098707; US20070092520, US20060270594; US20060134104; US20060035278; US20050233960; US20050037431

    • U.S. Pat. No. 5,686,292—for example, ATCC HB-11894 and ATCC HB-11895
    • US 20100016241—for example, ATCC HB-11894 (hybridoma 1A3.3.13) or HB-11895 (hybridoma 5D5.11.6)


National Defense Medical Center, Taiwan: Lu R M., et al Biomaterials. 2011 April; 32(12):3265-74.


Novartis: US20090175860

    • for example, an antibody comprising the sequences of CDR1, CDR2 and CDR3 of heavy chain 4687, wherein the sequences of CDR1, CDR2, and CDR3 of heavy chain 4687 are residues 26-35, 50-65, and 98-102, respectively, of SEQ ID NO: 58; and the sequences of CDR1, CDR2, and CDR3 of light chain 5097, wherein the sequences of CDR1, CDR2, and CDR3 oflight chain 5097 are residues 24-39,55-61, and 94-100 of SEQ ID NO: 37.


Pharmacia Corporation: US20040166544


Pierre Fabre: US20110239316, US20110097262, US20100115639


Sumsung: US 20110129481—for example a monoclonal antibody produced from a hybridoma cell having accession number KCLRF-BP-00219 or accession number of KCLRF-BP-00223.


Samsung: US 20110104176—for example an antibody produced by a hybridoma cell having Accession Number: KCLRF-BP-00220.


University of Turin Medical School: DN-30 Pacchiana G., et al J Biol Chem. 2010 Nov. 12; 285(46):36149-57


Van Andel Research Institute: Jiao Y., et al Mol Biotechnol. 2005 September; 31(1):41-54.


(43) MUC1 (Mucin 1, Cell Surface Associated)


Nucleotide


Genbank accession no J05581


Genbank version no. J05581.1 GI:188869


Genbank record update date: Jun. 23, 2010 08:48 AM


Polypeptide


Genbank accession no. AAA59876


Genbank version no. AAA59876.1 GI:188870


Genbank record update date: Jun. 23, 2010 08:48 AM


CROSS REFERENCES



  • Gendler S. J., et al J. Biol. Chem. 265 (25), 15286-15293 (1990)



Other Information


Official Symbol: MUC1


Other Aliases: RP11-263K19.2, CD227, EMA, H23AG, KL-6, MAM6, MUC-1, MUC-1/SEC, MUC-1/X, MUC1/ZD, PEM, PEMT, PUM


Other Designations: DF3 antigen; H23 antigen; breast carcinoma-associated antigen DF3; carcinoma-associated mucin; episialin; krebs von den Lungen-6; mucin 1, transmembrane; mucin-1; peanut-reactive urinary mucin; polymorphic epithelial mucin; tumor associated epithelial mucin; tumor-associated epithelial membrane antigen; tumor-associated mucin


Antibodies


AltaRex—Quest Pharma Tech: U.S. Pat. No. 6,716,966—for example an Alt-1 antibody produced by the hybridoma ATCC No PTA-975.


AltaRex- Quest Pharma Tech: U.S. Pat. No. 7,147,850


CRT: 5E5—Sørensen A L., et al Glycobiology vol. 16 no. 2 pp. 96-107, 2006; HMFG2-Burchell J., et al Cancer Res., 47, 5476-5482 (1987); see WO2015/159076


Glycotope GT-MAB: GT-MAB 2.5-GEX (Website: glycotope.com/pipeline/pankomab-gex)


Immunogen: U.S. Pat. No. 7,202,346

    • for example, antibody MJ-170: hybridoma cell line MJ-170 ATCC accession no. PTA-5286Monoclonal antibody MJ-171: hybridoma cell line MJ-171 ATCC accession no. PTA-5287; monoclonal antibody MJ-172: hybridoma cell line MJ-172 ATCC accession no. PTA-5288; or monoclonal antibody MJ-173: hybridoma cell line MJ-173 ATCC accession no. PTA-5302


Immunomedics: U.S. Pat. No. 6,653,104


Ramot Tel Aviv Uni: U.S. Pat. No. 7,897,351


Regents Uni. CA: U.S. Pat. No. 7,183,388; US20040005647; US20030077676.


Roche GlycArt: U.S. Pat. No. 8,021,856


Russian National Cancer Research Center: Imuteran—Ivanov P K., et al Biotechnol J. 2007 July; 2(7):863-70


Technische Univ Braunschweig: (IIB6, HT186-B7, HT186-D11, HT186-G2, HT200-3A-C1, HT220-M-D1, HT220-M-G8)—Thie H., et al PLoS One. 2011 Jan. 14;6(1):e15921


(44) CA9 (Carbonic Anhydrase IX)


Nucleotide


Genbank accession no. X66839


Genbank version no. X66839.1 GI:1000701


Genbank record update date: Feb. 2, 2011 10:15 AM


Polypeptide


Genbank accession no. CAA47315


Genbank version no. CAA47315.1 GI:1000702


Genbank record update date: Feb. 2, 2011 10:15 AM


CROSS REFERENCES



  • Pastorek J., et al Oncogene 9 (10), 2877-2888 (1994)



Other Information


Official Symbol: CA9


Other Aliases: CAIX, MN


Other Designations: CA-IX; P54/58N; RCC-associated antigen G250; RCC-associated protein G250; carbonate dehydratase IX; carbonic anhydrase 9; carbonic dehydratase; membrane antigen MN; pMW1; renal cell carcinoma-associated antigen G250


Antibodies


Abgenix/Amgen: US20040018198


Affibody: Anti-CAIX Affibody molecules

    • (affibody.com/en/Product-Portfolio/Pipeline/)


Bayer: U.S. Pat. No. 7,462,696


Bayer/Morphosys: 3ee9 mAb—Petrul H M., et al Mol Cancer Ther. 2012 February; 11(2):340-9


Harvard Medical School: Antibodies G10, G36, G37, G39, G45, G57, G106, G119, G6, G27, G40 and G125. Xu C., et al PLoS One. 2010 Mar. 10; 5(3):e9625


Institute of Virology, Slovak Academy of Sciences (Bayer)—U.S. Pat. No. 5,955,075

    • for example, M75—ATCC Accession No. HB 11128 or MN12—ATCC Accession No. HB 11647


Institute of Virology, Slovak Academy of Sciences: U.S. Pat. No. 7,816,493

    • for example the M75 monoclonal antibody that is secreted from the hybridoma VU-M75, which was deposited at the American Type Culture Collection under ATCC No. HB 11128; or the V/10 monoclonal antibody secreted from the hybridoma V/10-VU, which was deposited at the International Depository Authority of the Belgian Coordinated Collection of Microorganisms (BCCM) at the Laboratorium voor Moleculaire Bioloqie-Plasmidencollectie (LMBP) at the Universeit Gent in Gent, Belgium, under Accession No. LMBP 6009CB.


Institute of Virology, Slovak Academy of Sciences US20080177046; US20080176310; US20080176258; US20050031623


Novartis: US20090252738


Wilex: U.S. Pat. No. 7,691,375—for example the antibody produced by the hybridoma cell line DSM ASC 2526.


Wilex: US20110123537; Rencarex: Kennett R H., et al Curr Opin Mol Ther. 2003 February; 5(1):70-5


Xencor: US20090162382


(45) EGFRvIII (Epidermal Growth Factor Receptor (EGFR), Transcript Variant 3,


Nucleotide


Genbank accession no. NM_201283


Genbank version no. NM_201283.1 GI:41327733


Genbank record update date: Sep. 30, 2012 01:47 PM


Polypeptide


Genbank accession no. NP_958440


Genbank version no. NP_958440.1 GI:41327734


Genbank record update date: Sep. 30, 2012 01:47 PM


CROSS-REFERENCES



  • Batra S K., et al Cell Growth Differ 1995; 6:1251-1259.



Antibodies:


U.S. Pat. Nos. 7,628,986 and 7,736,644 (Amgen)

    • For example, a heavy chain variable region amino acid sequence selected from the group consisting of SEQ ID NO: 142 and variants & a light chain variable region amino acid sequence selected from the group consisting of: SEQ ID NO: 144 and variants.


US20100111979 (Amgen)

    • For example, an antibody comprising a heavy chain amino acid sequence comprising:
    • CDR1 consisting of a sequence selected from the group consisting of the amino acid sequences for the CDR1 region of antibodies 13.1.2 (SEQ ID NO: 138), 131 (SEQ ID NO: 2), 170 (SEQ ID NO: 4), 150 (SEQ ID NO: 5), 095 (SEQ ID NO: 7), 250 (SEQ ID NO: 9), 139 (SEQ ID NO: 10), 211 (SEQ ID NO: 12), 124 (SEQ ID NO: 13), 318 (SEQ ID NO: 15), 342 (SEQ ID NO: 16), and 333 (SEQ ID NO: 17); CDR2 consisting of a sequence selected from the group consisting of the amino acid sequences for the CDR2 region of antibodies 13.1.2 (SEQ ID NO: 138), 131 (SEQ ID NO: 2), 170 (SEQ ID NO: 4), 150 (SEQ ID NO: 5), 095 (SEQ ID NO: 7), 250 (SEQ ID NO: 9), 139 (SEQ ID NO: 10), 211 (SEQ ID NO: 12), 124 (SEQ ID NO: 13), 318 (SEQ ID NO: 15), 342 (SEQ ID NO: 16), and 333 (SEQ ID NO: 17); and
    • CDR3 consisting of a sequence selected from the group consisting of the amino acid sequences for the CDR3 region of antibodies 13.1.2 (SEQ ID NO: 138), 131 (SEQ ID NO: 2), 170 (SEQ ID NO: 4), 150 (SEQ ID NO: 5), 095 (SEQ ID NO: 7), 250 (SEQ ID NO: 9), 139 (SEQ ID NO: 10), 211 (SEQ ID NO: 12), 124 (SEQ ID NO: 13), 318 (SEQ ID NO: 15), 342 (SEQ ID NO: 16), and 333 (SEQ ID NO: 17).


US20090240038 (Amgen)

    • For example, an antibody having at least one of the heavy or light chain polypeptides comprises an amino acid sequence that is at least 90% identical to the amino acid sequence selected from the group consisting of: SEQ ID NO: 2, SEQ ID NO: 19, SEQ ID NO: 142, SEQ ID NO: 144, and any combination thereof.


US20090175887 (Amgen)

    • For example, an antibody having a heavy chain amino acid sequence selected from the group consisting of the heavy chain amino acid sequence of antibody 13.1.2 (SEQ ID NO: 138), 131 (SEQ ID NO: 2), 170 (SEQ ID NO: 4), 150 (SEQ ID NO: 5), 095 (SEQ ID NO: 7), 250 (SEQ ID NO: 9), 139 (SEQ ID NO: 10), 211 (SEQ ID NO: 12), 124 (SEQ ID NO: 13), 318 (SEQ ID NO: 15), 342 (SEQ ID NO: 16), and 333 (SEQ ID NO: 17).


US20090156790 (Amgen)

    • For example, antibody having heavy chain polypeptide and a light chain polypeptide, wherein at least one of the heavy or light chain polypeptides comprises an amino acid sequence that is at least 90% identical to the amino acid sequence selected from the group consisting of: SEQ ID NO: 2, SEQ ID NO: 19, SEQ ID NO: 142, SEQ ID NO: 144, and any combination thereof.


US20090155282, US20050059087 and US20050053608 (Amgen)

    • For example, an antibody heavy chain amino acid sequence selected from the group consisting of the heavy chain amino acid sequence of antibody 13.1.2 (SEQ ID NO: 138), 131 (SEQ ID NO: 2), 170 (SEQ ID NO: 4), 150 (SEQ ID NO: 5), 095 (SEQ ID NO: 7), 250 (SEQ ID NO: 9), 139 (SEQ ID NO: 10), 211 (SEQ ID NO: 12), 124 (SEQ ID NO: 13), 318 (SEQ ID NO: 15), 342 (SEQ ID NO: 16), and 333 (SEQ ID NO: 17).


MR1-1 (U.S. Pat. No. 7,129,332; Duke)

    • For example, a variant antibody having the sequence of SEQ ID NO.18 with the substitutions S98P-T99Y in the CDR3 VH, and F92W in CDR3 VL.


L8A4, H10, Y10 (Wikstrand C J., et al Cancer Res. 1995 Jul. 15; 55(14):3140-8; Duke)


US20090311803 (Harvard University)

    • For example, SEQ ID NO:9 for antibody heavy chain variable region, and SEQ ID NO: 3 for light chain variable region amino acid sequences


US20070274991 (EMD72000, also known as matuzumab; Harvard University)

    • For example, SEQ ID NOs: 3 & 9 for light chain and heavy chain respectively


U.S. Pat. No. 6,129,915 (Schering)

    • For example, SEQ. ID NOs: 1, 2, 3, 4, 5 and 6.


mAb CH12—Wang H., et al FASEB J. 2012 January; 26(1):73-80 (Shanghai Cancer Institute).


RAbDMvIII—Gupta P., et al BMC Biotechnol. 2010 Oct. 7; 10:72 (Stanford University Medical Center).


mAb Ua30—Ohman L., et al Tumour Biol. 2002 March-April; 23(2):61-9 (Uppsala University).


Han D G., et al Nan Fang Yi Ke Da Xue Xue Bao. 2010 January; 30(1):25-9 (Xi'an Jiaotong University).


(46) Cd33 (Cd33 Molecule)


Nucleotide


Genbank accession no. M_23197


Genbank version no. NM_23197.1 GI:180097


Genbank record update date: Jun. 23, 2010 08:47 AM


Polypeptide


Genbank accession no. AAA51948


Genbank version no. AAA51948.1 GI:188098


Genbank record update date: Jun. 23, 2010 08:47 AM


CROSS-REFERENCES



  • Simmons D., et al J. Immunol. 141 (8), 2797-2800 (1988)



Other Information


Official Symbol: CD33


Other Aliases: SIGLEC-3, SIGLEC3, p67


Other Designations: CD33 antigen (gp67); gp67; myeloid cell surface antigen CD33; sialic acid binding Ig-like lectin 3; sialic acid-binding Ig-like lectin


Antibodies


H195 (Lintuzumab)—Raza A., et al Leuk Lymphoma. 2009 August; 50(8):1336-44; U.S. Pat. No. 6,759,045 (Seattle Genetics/Immunomedics)


mAb OKT9: Sutherland, D. R. et al. Proc Natl Acad Sci USA 78(7): 4515-4519 1981, Schneider,C., et al J Biol Chem 257, 8516-8522 (1982)


mAb E6: Hoogenboom, H. R., et al J Immunol 144, 3211-3217 (1990)


U.S. Pat. No. 6,590,088 (Human Genome Sciences)

    • For example, SEQ ID NOs: 1 and 2 and ATCC accession no. 97521


U.S. Pat. No. 7,557,189 (Immunogen)

    • For example, an antibody or fragment thereof comprising a heavy chain variable region which comprises three CDRs having the amino acid sequences of SEQ ID NOs:1-3 and a light chain variable region comprising three CDRs having the amino acid sequences of SEQ ID NOs:4-6.


(47) Cd19 (Cd19 Molecule)


Nucleotide


Genbank accession no. NM_001178098


Genbank version no. NM_001178098.1 GI:296010920


Genbank record update date: Sep. 10, 2012 12:43 AM


Polypeptide


Genbank accession no. NP_001171569


Genbank version no. NP_001171569.1 GI:296010921


Genbank record update date: Sep. 10, 2012 12:43 AM


CROSS-REFERENCES



  • Tedder T F., et al J. Immunol. 143 (2): 712-7 (1989)



Other Information


Official Symbol: CD19


Other Aliases: B4, CVID3


Other Designations: B-lymphocyte antigen CD19; B-lymphocyte surface antigen B4; T-cell surface antigen Leu-12; differentiation antigen CD19


Antibodies


Immunogen: HuB4—Al-Katib A M., et al Clin Cancer Res. 2009 Jun. 15; 15(12):4038-45.


4G7: Kügler M., et al Protein Eng Des Sel. 2009 March; 22(3):135-47

    • For example, sequences in FIG. 3 of Knappik, A. et al. J Mol Biol 2000 February; 296(1):57-86


AstraZeneca/MedImmune: MEDI-551—Herbst R., et al J Pharmacol Exp Ther. 2010 October; 335(1):213-22


Glenmark Pharmaceuticals: GBR-401—Hou S., et al Mol Cancer Ther November 2011 (Meeting Abstract Supplement) C164


U.S. Pat. No. 7,109,304 (Immunomedics)

    • For example, an antibody comprising the sequence of hA19Vk (SEQ ID NO:7) and the sequence of hA19VH (SEQ ID NO:10)


U.S. Pat. No. 7,902,338 (Immunomedics)

    • For example, an antibody or antigen-binding fragment thereof that comprises the light chain complementarity determining region CDR sequences CDR1 of SEQ ID NO: 16 (KASQSVDYDGDSYLN); CDR2 of SEQ ID NO: 17 (DASNLVS); and CDR3 of SEQ ID NO: 18 (QQSTEDPWT) and the heavy chain CDR sequences CDR1 of SEQ ID NO: 19 (SYWMN); CDR2 of SEQ ID NO: 20 (QIWPGDGDTNYNGKFKG) and CDR3 of SEQ ID NO: 21 (RETTTVGRYYYAMDY) and also comprises human antibody framework (FR) and constant region sequences with one or more framework region amino acid residues substituted from the corresponding framework region sequences of the parent murine antibody, and wherein said substituted FR residues comprise the substitution of serine for phenylalanine at Kabat residue 91 of the heavy chain variable region.


Medarex: MDX-1342—Cardarelli P M., et al Cancer Immunol Immunother. 2010 February; 59(2):257-65.


MorphoSys/Xencor: MOR-208/XmAb-5574—Zalevsky J., et al Blood. 2009 Apr. 16; 113(16):3735-43


U.S. Pat. No. 7,968,687 (Seattle Genetics)

    • An antibody or antigen-binding fragment comprising a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:9 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO: 24.


4G7 chim—Lang P., et al Blood. 2004 May 15; 103(10):3982-5 (University of Tübingen)


For example, FIG. 6 and SEQ ID No: 80 of US20120082664


Zhejiang University School of Medicine: 2E8—Zhang J., et al J Drug Target. 2010 November; 18(9):675-8


(48) IL2RA (Interleukin 2 Receptor, Alpha); NCBI Reference Sequence: NM_000417.2);


Nucleotide


Genbank accession no. NM_000417


Genbank version no. NM_000417.2 GI:269973860


Genbank record update date: Sep. 9, 2012 04:59 PM


Polypeptide


Genbank accession no. NP_000408


Genbank version no. NP_000408.1 GI:4557667


Genbank record update date: Sep. 9, 2012 04:59 PM


CROSS-REFERENCES

Kuziel W. A., et al J. Invest. Dermatol. 94 (6 SUPPL), 27S-32S (1990)


Other Information


Official Symbol: IL2RA


Other Aliases: RP11-536K7.1, CD25, IDDM10, IL2R, TCGFR


Other Designations: FIL-2 receptor subunit alpha; IL-2-RA; IL-2R subunit alpha; IL2-RA;


TAC antigen; interleukin-2 receptor subunit alpha; p55


Antibodies


U.S. Pat. No. 6,383,487 (Novartis/UCL: Baxilisimab [Simulect])


U.S. Pat. No. 6,521,230 (Novartis/UCL: Baxilisimab [Simulect])

    • For example, an antibody having an antigen binding site comprises at least one domain which comprises CDR1 having the amino acid sequence in SEQ. ID. NO: 7, CDR2 having the amino acid sequence in SEQ. ID. NO: 8, and CDR3 chaving the amino acid sequence in SEQ. ID. NO: 9; or said CDR1, CDR2 and CDR3 taken in sequence as a whole comprise an amino acid sequence which is at least 90% identical to SEQ. ID. NOs: 7, 8 and 9 taken in sequence as a whole.


Daclizumab—Rech A J., et al Ann N Y Acad Sci. 2009 September; 1174:99-106 (Roche)


(49) AXL (AXL Receptor Tyrosine Kinase)


Nucleotide


Genbank accession no. M76125


Genbank version no. M76125.1 GI:292869


Genbank record update date: Jun. 23, 2010 08:53 AM


Polypeptide


Genbank accession no. AAA61243


Genbank version no. AAA61243.1 GI:29870


Genbank record update date: Jun. 23, 2010 08:53 AM


CROSS-REFERENCES

O'Bryan J. P., et al Mol. Cell. Biol. 11 (10), 5016-5031 (1991); Bergsagel P. L., et al J. Immunol. 148 (2), 590-596 (1992)


Other Information


Official Symbol: AXL


Other Aliases: JTK11, UFO


Other Designations: AXL oncogene; AXL transforming sequence/gene; oncogene AXL; tyrosine-protein kinase receptor UFO


Antibodies


YW327.652—Ye X., et al Oncogene. 2010 Sep. 23; 29(38):5254-64. (Genentech)


BergenBio: BGB324 (bergenbio.com/BGB324)


(50) CD30—TNFRSF8 (Tumor Necrosis Factor Receptor Superfamily, Member 8)


Nucleotide


Genbank accession no. M83554


Genbank version no. M83554.1 GI:180095


Genbank record update date: Jun. 23, 2010 08:53 AM Polypeptide


Genbank accession no. AAA51947


Genbank version no. AAA51947.1 GI:180096


Genbank record update date: Jun. 23, 2010 08:53 AM


CROSS-REFERENCES



  • Durkop H., et al Cell 68 (3), 421-427 (1992)



Other Information


Official Symbol: TNFRSF8


Other Aliases: CD30, D1S166E, Ki-1


Other Designations: CD30L receptor; Ki-1 antigen; cytokine receptor CD30; lymphocyte activation antigen CD30; tumor necrosis factor receptor superfamily member 8


(51) BCMA (B-Cell Maturation Antigen)—TNFRSF17 (Tumor Necrosis Factor Receptor Superfamily, Member 17)


Nucleotide


Genbank accession no. Z29574


Genbank version no. Z29574.1 GI:471244


Genbank record update date: Feb. 2, 2011 10:40 AM


Polypeptide


Genbank accession no. CAA82690


Genbank version no. CAA82690.1 GI:471245


Genbank record update date: Feb. 2, 2011 10:40 AM


CROSS-REFERENCES



  • Laabi Y., et al Nucleic Acids Res. 22 (7), 1147-1154 (1994)



Other Information


Official Symbol: TNFRSF17


Other Aliases: BCM, BCMA, CD269


Other Designations: B cell maturation antigen; B-cell maturation factor; B-cell maturation protein; tumor necrosis factor receptor superfamily member 17


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


CROSS-REFERENCES



  • Fratta E., et al. Mol Oncol. 2011 April; 5(2):164-82; Lim S H., at al Am J Blood Res. 2012; 2(1):29-35.



(53) CD174 (Lewis Y) —FUT3 (Fucosyltransferase 3 (Galactoside 3(4)-L-Fucosyltransferase, Lewis Blood Group)


Nucleotide


Genbank accession no. NM000149


Genbank version no. NM000149.3 GI:148277008


Genbank record update date: Jun. 26, 2012 04:49 PM


Polypeptide


Genbank accession no. NP_000140


Genbank version no. NP_000140.1 GI:4503809


Genbank record update date: Jun. 26, 2012 04:49 PM


CROSS-REFERENCES



  • Kukowska-Latallo, J. F., et al Genes Dev. 4 (8), 1288-1303 (1990)



Other Information


Official Symbol: FUT3


Other Aliases: CD174, FT3B, FucT-III, LE, Les


Other Designations: Lewis F T; alpha-(1,3/1,4)-fucosyltransferase; blood group Lewis alpha-4-fucosyltransferase; fucosyltransferase III; galactoside 3(4)-L-fucosyltransferase


(54) CLEC14A (C-Type Lectin Domain Family 14, Member a; Genbank Accession No. NM175060)


Nucleotide


Genbank accession no. NM175060


Genbank version no. NM175060.2 GI:371123930


Genbank record update date: Apr. 1, 2012 03:34 PM


Polypeptide


Genbank accession no. NP_778230


Genbank version no. NP_778230.1 GI:28269707


Genbank record update date: Apr. 1, 2012 03:34 PM


Other Information


Official Symbol: CLEC14A


Other Aliases: UNQ236/PRO269, C14orf27, CEG1, EGFR-5


Other Designations: C-type lectin domain family 14 member A; CIECT and EGF-like domain containing protein; epidermal growth factor receptor 5


(55) GRP78—HSPA5 (Heat Shock 70 kDa Protein 5 (Glucose-Regulated Protein, 78 kDa)


Nucleotide


Genbank accession no. NM005347


Genbank version no. NM005347.4 GI:305855105


Genbank record update date: Sep. 30, 2012 01:42 PM


Polypeptide


Genbank accession no. NP_005338


Genbank version no. NP_005338.1 GI:16507237


Genbank record update date: Sep. 30, 2012 01:42 PM


CROSS-REFERENCES

Ting J., et al DNA 7 (4), 275-286 (1988)


Other Information


Official Symbol: HSPA5


Other Aliases: BIP, GRP78, MIF2


Other Designations: 78 kDa glucose-regulated protein; endoplasmic reticulum lumenal Ca(2+)-binding protein grp78; immunoglobulin heavy chain-binding protein


(56) Cd70 (Cd70 Molecule) L08096


Nucleotide


Genbank accession no. L08096


Genbank version no. L08096.1 GI:307127


Genbank record update date: Jun. 23, 2012 08:54 AM


Polypeptide


Genbank accession no. AAA36175


Genbank version no. AAA36175.1 GI:307128


Genbank record update date: Jun. 23, 2012 08:54 AM


CROSS-REFERENCES



  • Goodwin R. G., et al Cell 73 (3), 447-456 (1993)



Other Information


Official Symbol: CD70


Other Aliases: CD27L, CD27LG, TNFSF7


Other Designations: CD27 ligand; CD27-L; CD70 antigen; Ki-24 antigen; surface antigen CD70; tumor necrosis factor (ligand) superfamily, member 7; tumor necrosis factor ligand superfamily member 7


Antibodies


MDX-1411 against CD70 (Medarex) hlF6 (Oflazoglu, E., et al, Clin Cancer Res. 2008 Oct. 1; 14(19):6171-80; Seattle Genetics)

    • For example, see US20060083736 SEQ ID NOs: 1, 2, 11 and 12 and FIG. 1.


(57) Stem Cell Specific Antigens. For Example:

    • 5T4 (see entry (63) below)
    • CD25 (see entry (48) above)
    • CD32
      • Polypeptide
        • Genbank accession no. ABK42161
        • Genbank version no. ABK42161.1 GI:117616286
        • Genbank record update date: Jul. 25, 2007 03:00 PM
    • LGR5/GPR49
      • Nucleotide
        • Genbank accession no. NM_003667
        • Genbank version no. NM_003667.2 GI:24475886
        • Genbank record update date: Jul. 22, 2012 03:38 PM
      • Polypeptide
        • Genbank accession no. NP_003658
        • Genbank version no. NP_003658.1 GI:4504379
        • Genbank record update date: Jul. 22, 2012 03:38 PM
    • Prominin/CD133
      • Nucleotide
        • Genbank accession no. NM_006017
        • Genbank version no. NM_006017.2 GI:224994187
        • Genbank record update date: Sep. 30, 2012 01:47 PM
      • Polypeptide
        • Genbank accession no. NP_006008
        • Genbank version no. NP_006008.1 GI:5174387
        • Genbank record update date: Sep. 30, 2012 01:47 PM
      • (58) ASG-5


CROSS-REFERENCES



  • (Smith L. M., et. al AACR 2010 Annual Meeting (abstract #2590); Gudas J. M., et. al. AACR 2010 Annual Meeting (abstract #4393)



Antibodies


Anti-AGS-5 Antibody: M6.131 (Smith, L. M., et. al AACR 2010 Annual Meeting (abstract #2590)


(59) ENPP3 (Ectonucleotide Pyrophosphatase/Phosphodiesterase 3)


Nucleotide


Genbank accession no. AF005632


Genbank version no. AF005632.2 GI:4432589


Genbank record update date: Mar. 10, 2010 09:41 PM


Polypeptide


Genbank accession no. AAC51813


Genbank version no. AAC51813.1 GI:2465540


Genbank record update date: Mar. 10, 2010 09:41 PM


CROSS-REFERENCES

Jin-Hua P., et al Genomics 45 (2), 412-415 (1997)


Other Information


Official Symbol: ENPP3


Other Aliases: RP5-988G15.3, B10, CD203c, NPP3, PD-IBETA, PDNP3


Other Designations: E-NPP 3; dJ1005H11.3 (phosphodiesterase I/nucleotide pyrophosphatase 3); dJ914N13.3 (phosphodiesterase I/nucleotide pyrophosphatase 3); ectonucleotide pyrophosphatase/phosphodiesterase family member 3; gpl30RB13-6; phosphodiesterase I beta; phosphodiesterase I/nucleotide pyrophosphatase 3; phosphodiesterase-I beta


(60) PRR4 (Proline Rich 4 (Lacrimal))


Nucleotide


Genbank accession no. NM_007244


Genbank version no. NM_007244.2 GI:154448885


Genbank record update date: Jun. 28, 2012 12:39 PM


Polypeptide


Genbank accession no. NP_009175


Genbank version no. NP_009175.2 GI:154448886


Genbank record update date: Jun. 28, 2012 12:39 PM


CROSS-REFERENCES



  • Dickinson D. P., et al Invest. Ophthalmol. Vis. Sci. 36 (10), 2020-2031 (1995)



Other Information


Official Symbol: PRR4


Other Aliases: LPRP, PROL4


Other Designations: lacrimal proline-rich protein; nasopharyngeal carcinoma-associated proline-rich protein 4; proline-rich polypeptide 4; proline-rich protein 4


(61) GCC—GUCY2C (Guanylate Cyclase 2C (Heat Stable Enterotoxin Receptor)


Nucleotide


Genbank accession no. NM_004963


Genbank version no. NM_004963.3 GI:222080082


Genbank record update date: Sep. 2, 2012 01:50 PM


Polypeptide


Genbank accession no. NP_004954


Genbank version no. NP_004954.2 GI:222080083


Genbank record update date: Sep. 2, 2012 01:50 PM


CROSS-REFERENCES



  • De Sauvage F. J., et al J. Biol. Chem. 266 (27), 17912-17918 (1991); Singh S., et al Biochem. Biophys. Res. Commun. 179 (3), 1455-1463 (1991)



Other Information


Official Symbol: GUCY2C


Other Aliases: DIAR6, GUC2C, MUCIL, STAR


Other Designations: GC-C; STA receptor; guanylyl cyclase C; hSTAR; heat-stable enterotoxin receptor; intestinal guanylate cyclase


(62) Liv-1—SLC39A6 (Solute Carrier Family 39 (Zinc Transporter), Member 6)


Nucleotide


Genbank accession no. U41060


Genbank version no. U41060.2 GI:12711792


Genbank record update date: Nov. 30, 2009 04:35 PM


Polypeptide


Genbank accession no. AAA96258


Genbank version no. AAA96258.2 GI:12711793


Genbank record update date: Nov. 30, 2009 04:35 PM


CROSS-REFERENCES



  • Taylor K M., et al Biochim Biophys Acta. 2003 Apr. 1; 1611(1-2):16-30



Other Information


Official Symbol: SLC39A6


Other Aliases: LIV-1


Other Designations: LIV-1 protein, estrogen regulated; ZIP-6; estrogen-regulated protein LIV-1; solute carrier family 39 (metal ion transporter), member 6; solute carrier family 39 member 6; zinc transporter ZIP6; zrt- and Irt-like protein 6


(63) 5T4, Trophoblast Glycoprotein, TPBG—TPBG (Trophoblast Glycoprotein)


Nucleotide


Genbank accession no. AJ012159


Genbank version no. AJ012159.1 GI:3805946


Genbank record update date: Feb. 1, 2011 10:27 AM


Polypeptide


Genbank accession no. CAA09930


Genbank version no. CAA09930.1 GI:3805947


Genbank record update date: Feb. 1, 2011 10:27 AM


CROSS-REFERENCES



  • King K. W., et al Biochim. Biophys. Acta 1445 (3), 257-270 (1999)



Other Information

    • Official Symbol: TPBG
    • Other Aliases: 5T4, 5T4AG, M6P1
    • Other Designations: 5T4 oncofetal antigen; 5T4 oncofetal trophoblast glycoprotein; 5T4 oncotrophoblast glycoprotein
    • See WO2015/155345


(64) CD56—NCMA1 (Neural Cell Adhesion Molecule 1)


Nucleotide


Genbank accession no. NM_000615


Genbank version no. NM_000615.6 GI:336285433


Genbank record update date: Sep. 23, 2012 02:32 PM


Polypeptide


Genbank accession no. NP_000606


Genbank version no. NP_000606.3 GI:94420689


Genbank record update date: Sep. 23, 2012 02:32 PM


CROSS-REFERENCES



  • Dickson, G., et al, Cell 50 (7), 1119-1130 (1987)



Other Information


Official Symbol: NCAM1


Other Aliases: CD56, MSK39, NCAM


Other Designations: antigen recognized by monoclonal antibody 5.1H11; neural cell adhesion molecule, NCAM


Antibodies


Immunogen: HuN901 (Smith S V., et al Curr Opin Mol Ther. 2005 August; 7(4):394-401)

    • For example, see humanized from murine N901 antibody. See FIGS. 1b and 1e of Roguska, M. A., et al. Proc Natl Acad Sci USA February 1994; 91:969-973.


(65) CanAg (Tumor Associated Antigen CA242)


CROSS-REFERENCES



  • Haglund C., et al Br J Cancer 60:845-851, 1989; Baeckstrom D., et al J Biol Chem 266:21537-21547, 1991



Antibodies


huC242 (Tolcher A W et al., J Clin Oncol. 2003 Jan. 15; 21(2):211-22; Immunogen)

    • For example, see US20080138898A1 SEQ ID NO: 1 and 2


(66) FOLR1 (Folate Receptor 1)


Nucleotide


Genbank accession no. J05013


Genbank version no. J05013.1 GI:182417


Genbank record update date: Jun. 23, 2010 08:47 AM


Polypeptide


Genbank accession no. AAA35823


Genbank version no. AAA35823.1 GI:182418


Genbank record update date: Jun. 23, 2010 08:47 AM


CROSS-REFERENCES

Elwood P. C., et al J. Biol. Chem. 264 (25), 14893-14901 (1989)


Other Information


Official Symbol: FOLR1


Other Aliases: FBP, FOLR


Other Designations: FR-alpha; KB cells FBP; adult folate-binding protein; folate binding protein; folate receptor alpha; folate receptor, adult; ovarian tumor-associated antigen MOv18


Antibodies


M9346A—Whiteman K R., et al Cancer Res Apr. 15, 2012; 72(8 Supplement): 4628 (Immunogen)


(67) GPNMB (Glycoprotein (Transmembrane) nmb)


Nucleotide


Genbank accession no. X76534


Genbank version no. X76534.1 GI:666042


Genbank record update date: Feb. 2, 2011 10:10 AM


Polypeptide


Genbank accession no. CAA54044


Genbank version no. CAA54044.1 GI:666043


Genbank record update date: Feb. 2, 2011 10:10 AM


CROSS-REFERENCES



  • Weterman M. A., et al Int. J. Cancer 60 (1), 73-81 (1995)



Other Information


Official Symbol: GPNMB


Other Aliases: UNQ1725/PRO9925, HGFIN, NMB


Other Designations: glycoprotein NMB; glycoprotein nmb-like protein; osteoactivin; transmembrane glycoprotein HGFIN; transmembrane glycoprotein NMB


Antibodies


Celldex Therapeutics: CR011 (Tse K F., et al Clin Cancer Res. 2006 Feb. 15; 12(4):1373-82)

    • For example, see EP1827492B1 SEQ ID NO: 22, 24, 26, 31, 33 and 35


(68) TIM-1—HAVCR1 (Hepatitis a Virus Cellular Receptor 1)


Nucleotide


Genbank accession no. AF043724


Genbank version no. AF043724.1 GI:2827453


Genbank record update date: Mar. 10, 2010 06:24 PM


Polypeptide


Genbank accession no. AAC39862


Genbank version no. AAC39862.1 GI:2827454


Genbank record update date: Mar. 10, 2010 06:24 PM


CROSS-REFERENCES



  • Feigelstock D., et al J. Virol. 72 (8), 6621-6628 (1998)



Other Information


Official Symbol: HAVCR1


Other Aliases: HAVCR, HAVCR-1, KIM-1, KIM1, TIM, TIM-1, TIM1, TIMD-1, TIMD1


Other Designations: T cell immunoglobin domain and mucin domain protein 1; T-cell membrane protein 1; kidney injury molecule 1


(69) RG-1/Prostate Tumor Target Mindin—Mindin/RG-1


CROSS-REFERENCES



  • Parry R., et al Cancer Res. 2005 Sep. 15; 65(18):8397-405



(70) B7-H4—VTCN1 (V-Set Domain Containing T Cell Activation Inhibitor 1


Nucleotide


Genbank accession no. BX648021


Genbank version no. BX648021.1 GI:34367180


Genbank record update date: Feb. 2, 2011 08:40 AM


CROSS-REFERENCES



  • Sica G L., et al Immunity. 2003 June; 18(6):849-61



Other Information


Official Symbol: VTCN1


Other Aliases: RP11-229A19.4, B7-H4, B7H4, B7S1, B7X, B7h.5, PRO1291, VCTN1


Other Designations: B7 family member, H4; B7 superfamily member 1; T cell costimulatory molecule B7x; T-cell costimulatory molecule B7x; V-set domain-containing T-cell activation inhibitor 1; immune costimulatory protein B7-H4 (71) PTK7 (PTK7 protein tyrosine kinase 7)


Nucleotide


Genbank accession no. AF447176


Genbank version no. AF447176.1 GI:17432420


Genbank record update date: Nov. 28, 2008 01:51 PM


Polypeptide


Genbank accession no. AAL39062


Genbank version no. AAL39062.1 GI:17432421


Genbank record update date: Nov. 28, 2008 01:51 PM


CROSS-REFERENCES



  • Park S. K., et al J. Biochem. 119 (2), 235-239 (1996)



Other Information


Official Symbol: PTK7


Other Aliases: CCK-4, CCK4


Other Designations: colon carcinoma kinase 4; inactive tyrosine-protein kinase 7; pseudo tyrosine kinase receptor 7; tyrosine-protein kinase-like 7


(72) Cd37 (Cd37 Molecule)


Nucleotide


Genbank accession no. NM_001040031


Genbank version no. NM_001040031.1 GI:91807109


Genbank record update date: Jul. 29, 2012 02:08 PM


Polypeptide


Genbank accession no. NP_001035120


Genbank version no. NP_001035120.1 GI:91807110


Genbank record update date: Jul. 29, 2012 02:08 PM


CROSS-REFERENCES

Schwartz-Albiez R., et al J. Immunol. 140 (3), 905-914 (1988)


Other Information


Official Symbol: CD37


Other Aliases: GP52-40, TSPAN26


Other Designations: CD37 antigen; cell differentiation antigen 37; leukocyte antigen CD37; leukocyte surface antigen CD37; tetraspanin-26; tspan-26


Antibodies


Boehringer Ingelheim: mAb 37.1 (Heider K H., et al Blood. 2011 Oct. 13; 118(15):4159-68)


Trubion: CD37-SMIP (G28-1 scFv-Ig) ((Zhao X., et al Blood. 2007; 110: 2569-2577)

    • For example, see US20110171208A1 SEQ ID NO: 253


Immunogen: K7153A (Deckert J., et al Cancer Res Apr. 15, 2012; 72(8 Supplement): 4625)


(73) CD138—SDC1 (Syndecan 1)


Nucleotide


Genbank accession no. AJ551176


Genbank version no. AJ551176.1 GI:29243141


Genbank record update date: Feb. 1, 2011 12:09 PM


Polypeptide


Genbank accession no. CAD80245


Genbank version no. CAD80245.1 GI:29243142


Genbank record update date: Feb. 1, 2011 12:09 PM


CROSS-REFERENCES



  • O'Connell F P., et al Am J Clin Pathol. 2004 February; 121(2):254-63



Other Information


Official Symbol: SDC1


Other Aliases: CD138, SDC, SYND1, syndecan


Other Designations: CD138 antigen; heparan sulfate proteoglycan fibroblast growth factor receptor; syndecan proteoglycan 1; syndecan-1


Antibodies


Biotest: chimerized MAb (nBT062)—(Jagannath S., et al Poster ASH #3060, 2010; WIPO Patent Application WO/2010/128087)

    • For example, see US20090232810 SEQ ID NO: 1 and 2


Immunogen: B-B4 (Tassone P., et al Blood 104_3688-3696)

    • For example, see US20090175863A1 SEQ ID NO: 1 and 2


(74) CD74 (CD74 Molecule, Major Histocompatibility Complex, Class II Invariant Chain)


Nucleotide


Genbank accession no. NM_004355


Genbank version no. NM_004355.1 GI:343403784


Genbank record update date: Sep. 23, 2012 02:30 PM


Polypeptide


Genbank accession no. NP_004346


Genbank version no. NP_004346.1 GI:10835071


Genbank record update date: Sep. 23, 2012 02:30 PM


CROSS-REFERENCES



  • Kudo, J., et al Nucleic Acids Res. 13 (24), 8827-8841 (1985)



Other Information


Official Symbol: CD74


Other Aliases: DHLAG, HLADG, II, la-GAMMA


Other Designations: CD74 antigen (invariant polypeptide of major histocompatibility complex, class II antigen-associated); HLA class II histocompatibility antigen gamma chain; HLA-DR antigens-associated invariant chain; HLA-DR-gamma; la-associated invariant chain; MHC HLA-DR gamma chain; gamma chain of class II antigens; p33


Antibodies


Immunomedics: hLL1 (Milatuzumab,)—Berkova Z., et al Expert Opin Investig Drugs. 2010 January; 19(1):141-9)

    • For example, see US20040115193 SEQ ID NOs: 19, 20, 21, 22, 23 and 24


Genmab: HuMax-CD74 (see website)


(75) Claudins—CLs (Claudins)


CROSS-REFERENCES



  • Offner S., et al Cancer Immunol Immunother. 2005 May; 54(5):431-45, Suzuki H., et al Ann N Y Acad Sci. 2012 July; 1258:65-70)



In humans, 24 members of the family have been described—see literature reference.


(76) EGFR (Epidermal Growth Factor Receptor)


Nucleotide


Genbank accession no. NM_005228


Genbank version no. NM_005228.3 GI:41927737


Genbank record update date: Sep. 30, 2012 01:47 PM


Polypeptide


Genbank accession no. NP_005219


Genbank version no. NP_005219.2 GI:29725609


Genbank record update date: Sep. 30, 2012 01:47 PM


CROSS-REFERENCES



  • Dhomen N S., et al Crit Rev Oncog. 2012; 17(1):31-50



Other Information


Official Symbol: EGFR


Other Aliases: ERBB, ERBB1, HER1, PIG61, mENA


Other Designations: avian erythroblastic leukemia viral (v-erb-b) oncogene homolog; cell growth inhibiting protein 40; cell proliferation-inducing protein 61; proto-oncogene c-ErbB-1; receptor tyrosine-protein kinase erbB-1


Antibodies


BMS: Cetuximab (Erbitux)—Broadbridge Vt., et al Expert Rev Anticancer Ther. 2012 May; 12(5):555-65.

    • For example, see U.S. Pat. No. 6,217,866—ATTC deposit No. 9764.


Amgen: Panitumumab (Vectibix)—Argiles G., et al Future Oncol. 2012 April; 8(4):373-89

    • For example, see U.S. Pat. No. 6,235,883 SEQ ID NOs: 23-38.


Genmab: Zalutumumab—Rivera F., et al Expert Opin Biol Ther. 2009 May; 9(5):667-74.


YM Biosciences: Nimotuzumab—Ramakrishnan M S., et al MAbs. 2009 January-February; 1(1):41-8.

    • For example, see U.S. Pat. No. 5,891,996 SEQ ID NOs: 27-34.


(77) Her3 (ErbB3)—ERBB3 (v-Erb-b2 Erythroblastic Leukemia Viral Oncogene Homolog 3 (Avian))


Nucleotide


Genbank accession no. M34309


Genbank version no. M34309.1 GI:183990


Genbank record update date: Jun. 23, 2010 08:47 PM


Polypeptide


Genbank accession no. AAA35979


Genbank version no. AAA35979.1 GI:306841


Genbank record update date: Jun. 23, 2010 08:47 PM


CROSS-REFERENCES



  • Plowman, G. D., et al., Proc. Natl. Acad. Sci. U.S.A. 87 (13), 4905-4909 (1990)



Other Information


Official Symbol: ERBB3


Other Aliases: ErbB-3, HER3, LCCS2, MDA-BF-1, c-erbB-3, c-erbB3, erbB3-S, p180-ErbB3, p45-sErbB3, p85-sErbB3


Other Designations: proto-oncogene-like protein c-ErbB-3; receptor tyrosine-protein kinase erbB-3; tyrosine kinase-type cell surface receptor HER3


Antibodies


Merimack Pharma: MM-121 (Schoeberl B., et al Cancer Res. 2010 Mar. 15; 70(6):2485-2494)

    • For example, see US2011028129 SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7 and 8.


(78) RON—MST1R (macrophage stimulating 1 receptor (c-met-related tyrosine kinase))


Nucleotide


Genbank accession no. X70040


Genbank version no. X70040.1 GI:36109


Genbank record update date: Feb. 2, 2011 10:17 PM


Polypeptide


Genbank accession no. CCA49634


Genbank version no. CCA49634.1 GI:36110


Genbank record update date: Feb. 2, 2011 10:17 PM


CROSS-REFERENCES



  • Ronsin C., et al Oncogene 8 (5), 1195-1202 (1993)



Other Information


Official Symbol: MST1R


Other Aliases: CD136, CDw136, PTK8, RON


Other Designations: MSP receptor; MST1R variant RON30; MST1R variant RON62; PTK8 protein tyrosine kinase 8; RON variant E2E3; c-met-related tyrosine kinase; macrophage-stimulating protein receptor; p185-Ron; soluble RON variant 1; soluble RON variant 2; soluble RON variant 3; soluble RONvariant 4


(79) EPHA2 (EPH Receptor A2)


Nucleotide


Genbank accession no. BC037166


Genbank version no. BC037166.2 GI:33879863


Genbank record update date: Mar. 6, 2012 01:59 PM


Polypeptide


Genbank accession no. AAH37166


Genbank version no. AAH37166.1 GI:22713539


Genbank record update date: Mar. 6, 2012 01:59 PM


CROSS-REFERENCES



  • Strausberg R. L., et al Proc. Natl. Acad. Sci. U.S.A. 99 (26), 16899-16903 (2002)



Other Information


Official Symbol: EPHA2


Other Aliases: ARCC2, CTPA, CTPP1, ECK


Other Designations: ephrin type-A receptor 2; epithelial cell receptor protein tyrosine kinase; soluble EPHA2 variant 1; tyrosine-protein kinase receptor ECK


Antibodies


Medimmune: 1C1 (Lee J W., et al Clin Cancer Res. 2010 May 1; 16(9):2562-2570)

    • For example, see US20090304721A1 FIGS. 7 and 8.


(80) CD20-MS4A1 (Membrane-Spanning 4-Domains, Subfamily A, Member 1)


Nucleotide


Genbank accession no. M27394


Genbank version no. M27394.1 GI:179307


Genbank record update date: Nov. 30, 2009 11:16 AM


Polypeptide


Genbank accession no. AAA35581


Genbank version no. AAA35581.1 GI:179308


Genbank record update date: Nov. 30, 2009 11:16 AM


CROSS-REFERENCES



  • Tedder T. F., et al Proc. Natl. Acad. Sci. U.S.A. 85 (1), 208-212 (1988)



Other Information


Official Symbol: MS4A1


Other Aliases: B1, Bp35, CD20, CVID5, LEU-16, MS4A2, S7


Other Designations: B-lymphocyte antigen CD20; B-lymphocyte cell-surface antigen B1; CD20 antigen; CD20 receptor; leukocyte surface antigen Leu-16


Antibodies


Genentech/Roche: Rituximab—Abdulla N E., et al BioDrugs. 2012 Apr. 1; 26(2):71-82.

    • For example, see U.S. Pat. No. 5,736,137, ATCC deposit No. HB-69119.


GSK/Genmab: Ofatumumab—Nightingale G., et al Ann Pharmacother. 2011 October; 45(10):1248-55.

    • For example, see US20090169550A1 SEQ ID NOs: 2, 4 and 5.
    • Immunomedics: Veltuzumab—Goldenberg D M., et al Leuk Lymphoma. 2010 May; 51(5):747-55.
    • For example, see U.S. Pat. No. 7,919,273B2 SEQ ID NOs: 1, 2, 3, 4, 5 and 6.


(81) Tenascin C—TNC (Tenascin C)


Nucleotide


Genbank accession no. NM 002160


Genbank version no. NM_002160.3 GI:340745336


Genbank record update date: Sep. 23, 2012 02:33 PM


Polypeptide


Genbank accession no. NP_002151


Genbank version no. NP_002151.2 GI:153946395


Genbank record update date: Sep. 23, 2012 02:33 PM


CROSS-REFERENCES



  • Nies D. E., et al J. Biol. Chem. 266 (5), 2818-2823 (1991); Siri A., et al Nucleic Acids Res. 19 (3), 525-531 (1991)



Other Information


Official Symbol: TNC


Other Aliases: 150-225, GMEM, GP, HXB, JI, TN, TN-C


Other Designations: GP 150-225; cytotactin; glioma-associated-extracellular matrix antigen; hexabrachion (tenascin); myotendinous antigen; neuronectin; tenascin; tenascin-C isoform 14/AD1/16


Antibodies


Philogen: G11 (von Lukowicz T., et al J Nuci Med. 2007 April; 48(4):582-7) and F16 (Pedretti M., et al Lung Cancer. 2009 April; 64(1):28-33)

    • For example, see U.S. Pat. No. 7,968,685 SEQ ID NOs: 29, 35, 45 and 47.


(82) FAP (Fibroblast activation protein, alpha)


Nucleotide


Genbank accession no. U09278


Genbank version no. U09278.1 GI:1888315


Genbank record update date: Jun. 23, 2010 09:22 AM


Polypeptide


Genbank accession no. AAB49652


Genbank version no. AAB49652.1 GI:1888316


Genbank record update date: Jun. 23, 2010 09:22 AM


CROSS-REFERENCES



  • Scanlan, M. J., et al Proc. Natl. Acad. Sci. U.S.A. 91 (12), 5657-5661 (1994)



Other Information


Official Symbol: FAP


Other Aliases: DPPIV, FAPA


Other Designations: 170 kDa melanoma membrane-bound gelatinase; integral membrane serine protease; seprase


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


Nucleotide


Genbank accession no. NM_012242


Genbank version no. NM_012242.2 GI:61676924


Genbank record update date: Sep. 30, 2012 01:48 PM


Polypeptide


Genbank accession no. NP_036374


Genbank version no. NP_036374.1 GI:7110719


Genbank record update date: Sep. 30, 2012 01:48 PM


CROSS-REFERENCES

Fedi P. et al J. Biol. Chem. 274 (27), 19465-19472 (1999)


Other Information


Official Symbol: DKK1


Other Aliases: UNQ492/PRO1008, DKK-1, SK


Other Designations: dickkopf related protein-1; dickkopf-1 like; dickkopf-like protein 1; dickkopf-related protein 1; hDkk-1


Antibodies


Novartis: BHQ880 (Fulciniti M., et al Blood. 2009 Jul. 9; 114(2):371-379)

    • For example, see US20120052070A1 SEQ ID NOs: 100 and 108.


(84) Cd52 (Cd52 Molecule)


Nucleotide


Genbank accession no. NM_001803


Genbank version no. NM_001803.2 GI:68342029


Genbank record update date: Sep. 30, 2012 01:48 PM


Polypeptide


Genbank accession no. NP_001794


Genbank version no. NP_001794.2 GI:68342030


Genbank record update date: Sep. 30, 2012 01:48 PM


CROSS-REFERENCES



  • Xia M. Q., et al Eur. J. Immunol. 21 (7), 1677-1684 (1991)



Other Information


Official Symbol: CD52


Other Aliases: CDW52


Other Designations: CAMPATH-1 antigen; CD52 antigen (CAMPATH-1 antigen); CDW52 antigen (CAMPATH-1 antigen); cambridge pathology 1 antigen; epididymal secretory protein E5; he5; human epididymis-specific protein 5


Antibodies


Alemtuzumab (Campath)—Skoetz N., et al Cochrane Database Syst Rev. 2012 Feb. 15; 2:CD008078.

    • For example, see Drugbank Acc. No. DB00087 (BIOD00109, BTD00109)


(85) CS1-SLAMF7 (SLAM Family Member 7)


Nucleotide


Genbank accession no. NM_021181


Genbank version no. NM_021181.3 GI:1993571


Genbank record update date: Jun. 29, 2012 11:24 AM


Polypeptide


Genbank accession no. NP_067004


Genbank version no. NP_067004.3 GI:19923572


Genbank record update date: Jun. 29, 2012 11:24 AM


CROSS-REFERENCES



  • Boles K. S., et al Immunogenetics 52 (3-4), 302-307 (2001)



Other Information


Official Symbol: SLAMF7


Other Aliases: UNQ576/PRO1138, 19A, CD319, CRACC, CS1


Other Designations: 19A24 protein; CD2 subset 1; CD2-like receptor activating cytotoxic cells; CD2-like receptor-activating cytotoxic cells; membrane protein FOAP-12; novel LY9 (lymphocyte antigen 9) like protein; protein 19A


Antibodies


BMS: elotuzumab/HuLuc63 (Benson D M., et al J Clin Oncol. 2012 Jun. 1; 30(16):2013-2015)

    • For example, see US20110206701 SEQ ID NOs: 9, 10, 11, 12, 13, 14, 15 and 16.


(86) Endoglin—ENG (Endoglin)


Nucleotide


Genbank accession no. AF035753


Genbank version no. AF035753.1 GI:3452260


Genbank record update date: Mar. 10, 2010 06:36 PM


Polypeptide


Genbank accession no. AAC32802


Genbank version no. AAC32802.1 GI:3452261


Genbank record update date: Mar. 10, 2010 06:36 PM


CROSS-REFERENCES



  • Rius C., et al Blood 92 (12), 4677-4690 (1998)



Official Symbol: ENG


Other Information


Other Aliases: RP11-228B15.2, CD105, END, HHT1, ORW, ORW1


Other Designations: CD105 antigen


(87) Annexin A1-ANXA1 (Annexin A1)


Nucleotide


Genbank accession no. X05908


Genbank version no. X05908.1 GI:34387


Genbank record update date: Feb. 2, 2011 10:02 AM


Polypeptide


Genbank accession no. CCA29338


Genbank version no. CCA29338.1 GI:34388


Genbank record update date: Feb. 2, 2011 10:02 AM


CROSS-REFERENCES



  • Wallner B. P., et al Nature 320 (6057), 77-81 (1986)



Other Information


Official Symbol: ANXA1


Other Aliases: RP11-71A24.1, ANX1, LPC1


Other Designations: annexin I (lipocortin I); annexin-1; calpactin II; calpactin-2; chromobindin-9; lipocortin I; p35; phospholipase A2 inhibitory protein


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


Nucleotide


Genbank accession no. M60335


Genbank version no. M60335.1 GI:340193


Genbank record update date: Jun. 23, 2010 08:56 AM


Polypeptide


Genbank accession no. AAA61269


Genbank version no. AAA61269.1 GI:340194


Genbank record update date: Jun. 23, 2010 08:56 AM


CROSS-REFERENCES

Hession C., et al J. Biol. Chem. 266 (11), 6682-6685 (1991)


Other Information


Official Symbol VCAM1


Other Aliases: CD106, INCAM-100


Other Designations: CD106 antigen; vascular cell adhesion protein 1


Antibody Sequences


Anti-Integrin αvβ6









RHAB6.2


SEQ ID NO: 1:


QVQLVQSGSELKKPGASVKISCKASGFAFTDSYMHWVRQAPGQGLEWMGW





IDPENGDTEYAPKFQGRFVFSLDTSVSTAYLQISSLKAEDTAVYYCTRGT





PTAVPNLRGDLQVLAQKVAGPYPFDYWGQGTLVTVSS





RHCB6.2


SEQ ID NO: 2:


QVQLVQSGAEVKKPGASVKVSCKASGYTFIDSYMHWVRQAPGQRLEWMGW





IDPENGDTEYAPKFQGRVTITTDTSASTAYMELSSLRSEDTAVYYCARGT





PTAVPNLRGDLQVLAQKVAGPYPFDYWGQGTLVTVSS





RHF


SEQ ID NO: 3:


QVQLVQSGAEVKKPGASVKVSCKASGFNFIDSYMHWVRQAPGQRLEWMGW





IDPENGDTEYAPKFQGRVTFTTDTSASTAYMELSSLRSEDTAVYYCNEGT





PTGPYYFDYWGQGTLVTVSS





RHFB6


SEQ ID NO: 4:


QVQLVQSGAEVKKPGASVKVSCKASGFNFIDSYMHWVRQAPGQRLEWMGW





IDPENGDTEYAPKFQGRVTFTTDTSASTAYMELSSLRSEDTAVYYCNEGT





PTAVPNLRGDLQVLAQKVAGPYYFDYWGQGTLVTVSS





RHAY100bP


SEQ ID NO: 5:


QVQLVQSGSELKKPGASVKISCKASGFAFTDSYMHWVRQAPGQGLEWMGW





IDPENGDTEYAPKFQGRFVFSLDTSVSTAYLQISSLKAEDTAVYYCTRGT





PTGPYPFDYWGQGTLVTVSS





RKF


SEQ ID NO: 6:


ENVLTQSPGTLSLSPGERATLSCSASSSVSYMHWFQQKPGQAPRLLIYST





SNLASGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQRSSYPLTFGGG





TKVEIK





RKFL36L50


SEQ ID NO: 7:


ENVLTQSPGTLSLSPGERATLSCSASSSVSYMHWLQQKPGQAPRLLIYLT





SNLASGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQRSSYPLTFGGG





TKVEIK





RKC


SEQ ID NO: 8:


EIVLTQSPGTLSLSPGERATLSCSASSSVSYMHWFQQKPGQAPRLLIYST





SNLASGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQRSSYPLTFGGG





TKVEIK






Anti-CD33









CD33 Hum195 VH


SEQ ID NO: 9:


QVQLVQSGAEVKKPGSSVKVSCKASGYTFTDYNMHWVRQAPGQGLEWIGY





IYPYNGGTGYNQKFKSKATITADESTNTAYMELSSLRSEDTAVYYCARGR





PAMDYWGQGTLVTVSS





CD33 Hum195 VK


SEQ ID NO: 10:


DIQMTQSPSSLSASVGDRVTITCRASESVDNYGISFMNWFQQKPGKAPKL





LIYAASNQGSGVPSRFSGSGSGTDFTLTISSLQPDDFATYYCQQSKEVPW





TFGQGTKVEIK






Anti-CD19


CD19 B4 resurfaced VH I









CD19 B4 resurfaced VH


SEQ ID NO: 11:


QVQLVQPGAEVVKPGASVKLSCKTSGYTFTSNWMHWVKQRPGQGLEWIGE





IDPSDSYTNYNQNFKGKAKLTVDKSTSTAYMEVSSLRSDDTAVYYCARGS





NPYYYAMDYWGQGTSVTVSS





CD19 B4 resurfaced VK


SEQ ID NO: 12:


EIVLTQSPAIMSASPGERVTMTCSASSGVNYMHWYQQKPGTSPRRWIYDT





SKLASGVPARFSGSGSGTSYSLTISSMEPEDAATYYCHQRGSYTFGGGTK





LEIK






Anti-Her2









Herceptin VH chain


SEQ ID NO: 13:


EVQLVESGGGLVQPGGSLRLSCAASGFNIKDTYIHWVRQAPGKGLEWVAR





IYPTNGYTRYADSVKGRFTISADTSKNTAYLQMNSLRAEDTAVYYCSRWG





GDGFYAMDYWGQGTLVTVSS





Herceptin VL chain


SEQ ID NO: 14:


DIQMTQSPSSLSASVGDRVTITCRASQDVNTAVAWYQQKPGKAPKLLIYS





ASFLYSGVPSRFSGSRSGTDFTLTISSLQPEDFATYYCQQHYTTPPTFGQ





GTKVEIK






Anti-CD25









Simulect VK (also known as Basiliximab)


SEQ ID NO: 15:


QIVSTQSPAIMSASPGEKVTMTCSASSSRSYMQWYQQKPGTSPKRWIYDT





SKLASGVPARFSGSGSGTSYSLTISSMEAEDAATYYCHQRSSYTFGGGTK





LEIK





Simulect VH


SEQ ID NO: 16:


QLQQSGTVLARPGASVKMSCKASGYSFTRYWMHWIKQRPGQGLEWIGAIY





PGNSDTSYNQKFEGKAKLTAVTSASTAYMELSSLTHEDSAVYYCSRDYGY





YFDFWGQGTTLTVSS






Anti-PSMA










Deimmunised VH ′1



SEQ ID NO: 17:


EVQLVQSGPEVKKPGATVKISCKTSGYTFTEYTIHWVKQAPGKGLEWIGNINPNNGGTTY





NQKFEDKATLTVDKSTDTAYMELSSLRSEDTAVYYCAAGWNFDYWGQGTLLTVSS





Deimmunised VK ′1


SEQ ID NO: 18:


DIQMTQSPSSLSTSVGDRVTLTCKASQDVGTAVDWYQQKPGPSPKLLIYWASTRHTGIPS





RFSGSGSGTDFTLTISSLQPEDFADYYCQQYNSYPLTFGPGTKVDIK





Deimmunised VH1 ′5


SEQ ID NO: 19:


EVKLVESGGGLVQPGGSMKLSCVASGFTFSNYWMNWVRQAPGKGLEWVAEIRSQSNN





FATHYAESVKGRVTISRDDSKSIVYLQMNNLRAEDTGVYYCTRRWNNFWGQGTTVTVSS





Deimmunised VH2 ′5


SEQ ID NO: 20:


EVKLVESGGGLVQPGGSLKLSCVASGFTFSNYWMNWVRQAPGKGLEWVAEIRSQSNNF





ATHYAESVKGRVTISRDDSKSIVYLQMNNLRAEDTAVYYCTRRWNNFWGQGTTVTVSS





Deimmunised VH3 ′5


SEQ ID NO: 21:


EVQLVESGGGLVQPGGSLKLSCVASGFTFSNYWMNWVRQAPGKGLEWVAEIRSQSNNF





ATHYAESVKGRVTISRDDSKSIVYLQMNNLRAEDTAVYYCTRRWNNFWGQGTTVTVSS





Deimmunised VH4 ′5


SEQ ID NO: 22:


EVQLVESGGGLVQPGGSLKLSCVASGFTFSNYWMNWVRQAPGKGLEWVAEIRSQSNNF





ATHYAESVKGRFTISRDDSKSIVYLQMNNLRAEDTAVYYCTRRWNNFWGQGTTVTVSS





Deimmunised VK1 ′5


SEQ ID NO: 23:


NIVMTQFPSSMSASVGDRVTITCKASENVGTYVSWYQQKPDQSPKMLIYGASNRFTGVP





DRFTGSGSATDFTLTISSLQTEDLADYYCGQSYTFPYTFGQGTKLEMK





Deimmunised VK2 ′5


SEQ ID NO: 24:


NIVMTQFPSSMSASVGDRVTITCKASENVGTYVSWYQQKPDQSPKMLIYGASNRFTGVP





DRFSGSGSGTDFTLTISSLQAEDLADYYCGQSYTFPYTFGQGTKLEIK





Deimmunised VK3 ′5


SEQ ID NO: 25:


NIQMTQFPSAMSASVGDRVTITCKASENVGTYVSWYQQKPDQSPKMLIYGASNRFTGVP





DRFSGSGSGTDFTLTISSLQAEDLADYYCGQSYTFPYTFGQGTKLEIK





Deimmunised VK4 ′5


SEQ ID NO: 26:


NIQMTQFPSAMSASVGDRVTITCKASENVGTYVSWYQQKPDQSPKMLIYGASNRFTGVP





DRFSGSGSGTDFTLTISSLQAEDEADYYCGQSYTFPYTFGQGTKLEIK





Deimmunised VK DI 5


SEQ ID NO: 27:


NIVMTQFPKSMSASAGERMTLTCKASENVGTYVSWYQQKPTQSPKMLIYGASNRFTGVP





DRFSGSGSGTDFILTISSVQAEDLVDYYCGQSYTFPYTFGGGTKLEMK





Deimmunised VH DI ′5


SEQ ID NO: 28:


EVKLEESGGGLVQPGGSMKISCVASGFTFSNYWMNWVRQSPEKGLEWVAEIRSQSNNF





ATHYAESVKGRVIISRDDSKSSVYLQMNSLRAEDTAVYYCTRRWNNFWGQGTTVTVSS





Humanised RHA ′5


SEQ ID NO: 29:


EVQLVESGGGLVQPGGSLKLSCAASGFTFSNYWMNWVRQASGKGLEWVGEIRSQSNNF





ATHYAESVKGRFTISRDDSKNTAYLQMNSLKTEDTAVYYCTRRWNNFWGQGTTVTVSS





Humanised RHB ′5


SEQ ID NO: 30:


EVKLVESGGGLVQPGGSLKLSCAASGFTFSNYWMNWVRQASGKGLEWVAEIRSQSNNF





ATHYAESVKGRVIISRDDSKNTVYLQMNSLRTEDTAVYYCTRRWNNFWGQGTTVTVSS





Humanised RHC ′5


SEQ ID NO: 31:


EVQLVESGGGLVQPGGSLKLSCAASGFTFSNYWMNWVRQASGKGLEWVAEIRSQSNNF





ATHYAESVKGRVIISRDDSKNTVYLQMNSLRTEDTAVYYCTRRWNNFWGQGTTVTVSS





Humanised RHD ′5


SEQ ID NO: 32:


EVKLVESGGGLVQPGGSLKLSCAASGFTFSNYWMNWVRQASGKGLEWVGEIRSQSNNF





ATHYAESVKGRVIISRDDSKNTVYLQMNSLRTEDTAVYYCTRRWNNFWGQGTTVTVSS





Humanised RHE ′5


SEQ ID NO: 33:


EVKLVESGGGLVQPGGSLKLSCAASGFTFSNYWMNWVRQASGKGLEWVAEIRSQSNNF





ATHYAESVKGRFTISRDDSKNTVYLQMNSLRTEDTAVYYCTRRWNNFWGQGTTVTVSS





Humanised RHF ′5


SEQ ID NO: 34:


EVKLVESGGGLVQPGGSLKLSCAASGFTFSNYWMNWVRQASGKGLEWVAEIRSQSNNF





ATHYAESVKGRVIISRDDSKNTAYLQMNSLRTEDTAVYYCTRRWNNFWGQGTTVTVSS





Humanised RHG ′5


SEQ ID NO: 35:


EVKLVESGGGLVQPGGSLKLSCAASGFTFSNYWMNWVRQASGKGLEWVAEIRSQSNNF





ATHYAESVKGRVIISRDDSKNTAYLQMNSLRTEDTAVYYCTRRWNNFWGQGTTVTVSS





Humanised RKA ′5


SEQ ID NO: 36:


DIQMTQSPSSVSASVGDRVTITCKASENVGTYVSWYQQKPGTAPKLLIYGASNRFTGVPS





RFSGSGSATDFTLTINNLQPEDFATYYCGQSYTFPYTFGQGTKVEIK





Humanised RKB ′5


SEQ ID NO: 37:


DIQMTQSPSSVSASVGDRVTITCKASENVGTYVSWYQQKPGTAPKLLIYGASNRFTGVPS





RFSGSGSATDFTLTINNLQPEDFATYYCGQSYTFPYTFGQGTKVEIK





Humanised RKC ′5


SEQ ID NO: 38:


DIQMTQSPSSVSASVGDRVTITCKASENVGTYVSWYQQKPGTAPKMLIYGASNRFTGVPS





RFSGSGSATDFTLTINNLQPEDFATYYCGQSYTFPYTFGQGTKVEIK





Humanised RKD ′5


SEQ ID NO: 39:


DIQMTQSPSSVSASVGDRVTITCKASENVGTYVSWYQQKPGTAPKMLIYGASNRFTGVPS





RFSGSGSATDFTLTINNLQPEDFATYYCGQSYTFPYTFGQGTKVEIK





Humanised RKE ′5


SEQ ID NO: 40:


NIVMTQSPSSVSASVGDRVTITCKASENVGTYVSWYQQKPGTAPKLLIYGASNRFTGVPD





RFTGSGSATDFILTINNLQPEDFATYYCGQSYTFPYTFGQGTKVEIK





Humanised RKF ′5


SEQ ID NO: 41:


NIVMTQSPSSVSASVGDRVTITCKASENVGTYVSWYQQKPGTAPKMLIYGASNRFTGVPS





RFSGSGSATDFILTINNLQPEDFATYYCGQSYTFPYTFGQGTKVEIK





Humanised RKG ′5


SEQ ID NO: 42:


NIVMTQSPSSVSASVGDRVTITCKASENVGTYVSWYQQKPGTAPKMLIYGASNRFTGVPD





RFTGSGSATDFTLTINNLQPEDFATYYCGQSYTFPYTFGQGTKVEIK






The parent antibody may also be a fusion protein comprising an albumin-binding peptide (ABP) sequence (Dennis et al. (2002) “Albumin Binding As A General Strategy For Improving The Pharmacokinetics Of Proteins” J Biol Chem. 277:35035-35043; WO 01/45746). Antibodies of the invention include fusion proteins with ABP sequences taught by: (i) Dennis et al (2002) J Biol Chem. 277:35035-35043 at Tables III and IV, page 35038; (ii) US 2004/0001827 at [0076]; and (iii) WO 01/45746 at pages 12-13, and all of which are incorporated herein by reference.


In one embodiment, the antibody has been raised to target specific the tumour related antigen αvβ6.


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 is 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 WO2008070593, 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 I. 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.


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, Sjögren'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 arcata, 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, Sjögren'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.


In some embodiments, the amount of the Conjugate administered ranges from about 0.01 to about 10 mg/kg per dose. In some embodiments, the amount of the Conjugate administered ranges from about 0.01 to about 5 mg/kg per dose. In some embodiments, the amount of the Conjugate administered ranges from about 0.05 to about 5 mg/kg per dose. In some embodiments, the amount of the Conjugate administered ranges from about 0.1 to about 5 mg/kg per dose. In some embodiments, the amount of the Conjugate administered ranges from about 0.1 to about 4 mg/kg per dose. In some embodiments, the amount of the Conjugate administered ranges from about 0.05 to about 3 mg/kg per dose.


In some embodiments, the amount of the Conjugate administered ranges from about 0.1 to about 3 mg/kg per dose. In some embodiments, the amount of the Conjugate administered ranges from about 0.1 to about 2 mg/kg per dose.


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 conjugates 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 conjugates 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. 11:843-852). 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 (A or B). 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 (A or B) 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). U.S. Pat. No. 7,521,541 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; U.S. Pat. Nos. 7,521,541; 7,723,485; WO2009/052249). 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


The Drug Linker compounds of the present invention (A and B) may be synthesised according to the Examples.


Synthesis of Drug Conjugates


Conjugates can be prepared as previously described. Antibodies can be conjugated to the Drug Linker compounds (A or B) 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 μm 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.


In some embodiments, the C11 substituent may be in the following stereochemical arrangement relative to neighbouring groups:




embedded image


In other embodiments, the C11 substituent may be in the following stereochemical arrangement relative to neighbouring groups:




embedded image


In one embodiment of the present invention, the compound of formula III is A.


In one embodiment of the present invention, the compound of formula III is B.


In one embodiment of the present invention, the Drug Linker unit of formula III is DL-A.


In one embodiment of the present invention, the Drug Linker unit of formula III is DL-B.


EXAMPLES

Reaction progress was monitored by 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. Flash chromatography was performed using Merck Kieselgel 60 F254 silica gel. Extraction and chromatography solvents were bought and used without further purification from VWR, U.K. All chemicals were purchased from Aldrich.


Proton NMR chemical shift values were measured on the delta scale at 400 MHz using a Bruker AV400. The following abbreviations have been used: s, singlet; d, doublet; t, triplet; q, quartet; quin, quintet; m, multiplet; br, broad. Coupling constants are reported in Hz. Column chromatography was performed on an Isolera (Biotage) automated system using normal phase SNAP cartridges.


The LC/MS conditions were as follow:


LCMS data were obtained using a Shimadzu Nexera series LC/MS with a Shimadzu LCMS-2020 quadrupole MS, with Electrospray ionisation. Mobile phase A—0.1% formic acid in water. Mobile phase B—0.1% formic acid in acetonitrile.


Short run gradient: initial composition was 5% B held over 0.25 min, then increase from 5% B to 100% B over a 2 min period. The composition was held for 0.50 min at 100% B, then returned to 5% B in 0.05 minutes and hold there for 0.05 min. Total gradient run time equals 3 min. Flow rate 0.8 mL/min. Wavelength detection range: 190 to 800 nm. Oven temperature: 50° C. Column: Waters Acquity UPLC BEH Shield RP18 1.7 μm 2.1×50 mm. Long run gradient: initial composition 5% B held over 1 min, then increase from 5% B to 100% B over a 9 min period. The composition was held for 2 min at 100% B, then returned to 5% B in 0.10 minutes and hold there for 3 min. Total gradient run time equals 15 min.


Flow rate 0.6 mL/min. Wavelength detection range: 190 to 800 nm. Oven temperature: 50° C. Column: ACE Excel 2 C18-AR, 2μ, 3.0×100 mm.


Example 1
(a) (S)-2-(methoxycarbonyl)-4-methylenepyrrolidinium chloride (3) IDC-/I C3



embedded image


Commercially available proline derivative (1) was obtained from Omegachem


(i) (S)-1-tert-butyl 2-methyl 4-methylenepyrrolidine-1,2-dicarboxylate (2)

Potassium carbonate (19.92 g, 14 mmol, 3.0 eq.) was added to a stirred solution of the carboxylic acid 1 (10.92 g, 48 mmol, 1.0 eq.) in DMF (270 mL). The resulting white suspension was stirred at room temperature for 30 mins, at which point iodomethane (21.48 g, 9.5 mL, 151 mmol, 3.15 eq.) was added. The reaction mixture was allowed to stir at room temperature for 3 days. The DMF was removed by rotary evaporation under reduced pressure to afford a yellow residue which was partitioned between ethylacetate and water. The organic layer was separated and the aqueous phase was extracted with ethylacetate. The combined organic layers were washed with water brined and dried over magnesium sulphate. The ethylacetate was removed by rotary evaporation under reduced pressure to give the crude product as a yellow oil. The crude product was purified by flash chromatography [85% n-hexane/15% ethylacetate] to afford the product as a colourless oil (10.74 g, 93%).


(ii) (S)-2-(methoxycarbonyl)-4-methylenepyrrolidinium chloride (3)

A solution of 4 M hydrochloric acid in dioxane (63 mL, 254.4 mmol, 4.5 eq.) was added to the Boc protected C-ring fragment 2 (13.67 g, 56.6 mmol, 1.0 eq.) at room temperature. Effervescence was observed indicating liberation of CO2 and removal of the Boc group. The product precipitated as a white solid and additional dioxane was added to facilitate stirring the reaction mixture was allowed to stir for an hour and then diluted with diethyl ether. The precipitated product was collected by vacuum filtration and washed with additional diethyl ether. Air drying afforded the desired product as a white powder (9.42 g, 94%).


(b) tert-Butyl (5-(3-(5-amino-4-((S)-2-(((tert-butyldimethylsilyl)oxy)methyl)-4-methylenepyrrolidine-1-carbonyl)-2-methoxyphenoxy)propoxy)-2-((S)-2-(((tert-butyldimethylsilyl)oxy)methyl)-4-methvlenepvrrolidine-1-carbonyl)-4-methoxyphenyl)carbamate (12)



embedded image


(i) 1′,3′-Bis[2-methoxy-4-(methoxycarbonyl)phenoxy]propane (5)

Diisopropyl azodicarboxylate (71.3 mL, 73.2 g, 362 mmol) was added drop-wise over a period of 60 min to an overhead stirred solution of methyl vanillate 4 (60 g, 329 mmol) and Ph3P (129.4 g, 494 mmol) in anhydrous THF (800 mL) at 0-5° C. (ice/acetone) under a nitrogen atmosphere. The reaction mixture was allowed to stir at 0-5° C. for an additional 1 h after which time a solution of 1,3-propanediol (11.4 mL, 12.0 g, 158 mmol) in THF (12 mL) was added drop-wise over a period of 20 min. The reaction mixture was allowed to warm to room temperature and stirred for 5 days. The resulting white precipitate 3 was collected by vacuum filtration, washed with THF and dried in a vacuum desiccator to constant weight. Yield=54.68 g (84% based on 1,3-propanediol). Analytical Data: Purity satisfactory by LC/MS 3.20 min (ES+) m/z (relative intensity) 427 ([M+Na]+, 10); 1H NMR (400 MHz, CDCl3) δ δ7.64 (dd, 2H, J=1.8, 8.3 Hz), 7.54 (d, 2H, J=1.8 Hz), 6.93 (d, 2H, J=8.5 Hz), 4.30 (t, 4H, J=6.1 Hz), 3.90 (s, 6H), 3.89 (s, 6H), 2.40 (p, 2H, J=6.0 Hz).


(ii) 1′,3′-Bis[2-methoxy-4-(methoxycarbonyl)-5-nitrophenoxy]propane (6)

Solid Cu(NO3)2.3H2O(81.54 g, 337.5 mmol) was added slowly to an overhead stirred slurry of the bis-ester 5 (54.68 g, 135 mmol) in acetic anhydride (650 mL) at 0-5° C. (ice/acetone). The reaction mixture was allowed to stir for 1 h at 0-5° C. and then allowed to warm to room temperature. A mild exotherm (c. 40-50° C.), accompanied by thickening of the mixture and evolution of NO2 was observed at this stage. Additional acetic anhydride (300 mL) was added and the reaction mixture was allowed to stir for 16 h at room temperature. The reaction mixture was poured onto ice (˜1.5 L), stirred and allowed to return to room temperature. The resulting yellow precipitate was collected by vacuum filtration and dried in a desiccator to afford the desired bis-nitro compound 6 as a yellow solid. Yield=66.7 g (100%). Analytical Data: Purity satisfactory by LC/MS 3.25 min (ES+) m/z (relative intensity) 517 ([M+Na]+, 40); 1H NMR (400 MHz, CDCl3) δ 7.49 (s, 2H), 7.06 (s, 2H), 4.32 (t, 4H, J=6.0 Hz), 3.95 (s, 6H), 3.90 (s, 6H), 2.45-2.40 (m, 2H). See ref Thurston 1996.


(iii) 1′,3′-Bis(4-carboxy-2-methoxy-5-nitrophenoxy) propane (7)

A slurry of the methyl ester 6 (66.7 g, 135 mmol) in THF (700 mL) was treated with 1N NaOH (700 mL) and the reaction mixture was allowed to stir vigorously at room temperature. After 4 days stirring, the slurry became a dark coloured solution which was subjected to rotary evaporation under reduced pressure to remove THF. The resulting aqueous residue was acidified to pH 1 with concentrated HCl and the colourless precipitate 7 was collected and dried thoroughly in a vacuum oven (50° C.). Yield=54.5 g (87%). Analytical Data: Purity satisfactory by LC/MS 2.65 min (ES+) m/z (relative intensity) 489 ([M+Na]+, 30); 1H NMR (400 MHz, DMSO-d6) δ 7.62 (s, 2H), 7.30 (s, 2H), 4.29 (t, 4H, J=6.0 Hz), 3.85 (s, 6H), 2.30-2.26 (m, 2H).


(iv) Dimethyl 1,1′-(4,4′-(propane-1,3-diylbis(oxy))bis(5-methoxy-2-nitrobenzoyl))(2S,2'S)-bis(4-methylenepyrrolidine-2-carboxylate) (8)

A catalytic amount of anhydrous DMF (2.4 mL) was added to a stirred suspension of oxalyl chloride (14.7 g, 9.8 mL, 115.8 mmol, 3 eq.) and dimer core 7 (18 g, 38.6 mmol, 1 eq.) in anhydrous DCM (500 mL) at room temperature. Vigorous effervescence was observed after the addition of DMF and the reaction mixture was allowed to stir for 18 h in a round bottom flask fitted with a calcium chloride drying tube. The resulting clear solution was evaporated under reduced pressure and the solid triturated with ether. The solid product was collected by vacuum filtration, washed with additional ether and dried in vacuo at 40° C. for 1.5 h. This solid was then added portion wise to a suspension of the C-ring 3 (15.1 g, 84.9 mmol, 2.2 eq.) and TEA (19.5 g, 27 ml, 119.6 mmol, 5 eq.) in dry DCM (375 mL), maintaining the temperature between −40 and −50° C. with the aid of a dry ice/acetonitrile bath. The reaction mixture was allowed to stir at −40° C. for 1 h and then allowed to warm to room temperature at which point LCMS indicated the complete consumption of the starting material. The reaction mixture was diluted with additional DCM and washed sequentially with aqueous hydrochloric acid (1M, 2×200 mL), saturated aqueous sodium bicarbonate (2×250 mL), water (250 mL), brine (250 mL), dried (MgSO4). DCM was removed by rotary evaporation under reduced pressure to afford the product as a yellow foam (25.72 g, 94%). Analytical Data: RT 1.59 min; MS (ES+) m/z (relative intensity) 713 ([M+H]+, 100)


(v) ((Propane-1,3-diylbis(oxy))bis(5-methoxy-2-nitro-4,1-phenylene))bis(((S)-2-(hydroxymethyl)-4-methylenepyrrolidin-1-yl)methanone) (9)

Solid lithium borohydride (3.18 g, 146 mmol, 3 eq.) was added in one portion to a solution of the ester 8 (34.72 g, 48.7 mmol, 1 eq.) in dry THF (350 mL) under a nitrogen atmosphere at 0° C. (ice bath). The reaction mixture was allowed to stir at 0° C. for 30 mins and then allowed to warm to room temperature at which point precipitation of an orange gum was observed. The reaction mixture was allowed to stir at room temperature for a futher 2 hours and then cooled in an ice bath and treated with water to give a yellow suspension. Hydrochloric acid (1M) was carefully added until effervescence ceased. The reaction mixture was extracted with ethylacetate (×4) and the combined organic layers were washed with water (×1), brine (×1) and dried (MgSO4). Ethylacetate was removed by rotary evaporation under reduced pressure to give a yellow foam. Purification by flash column chromatography [gradient elution DCM/MeOH 0% to 5% in 1% increments] gave the product as a pale yellow foam (23.1 g, 72%). Analytical Data: RT 1.23 min; MS (ES+) m/z (relative intensity) 657 ([M+H]+, 100)


(vi) ((Propane-1,3-diylbis(oxy))bis(5-methoxy-2-nitro-4,1-phenylene))bis(((S)-2-(((tert-butyldimethylsilyl)oxy)methyl)-4-methylenepyrrolidin-1-yl)methanone) (10)

A solution of the bis-alcohol 9 (10 g, 15.2 mmol, 1 eq.), t-butyldimethylsilylchloride (5.97 g, 39.6 mmol, 2.6 eq.) and imidazole (5.38 g, 79 mmol, 5.2 eq.) in dry DMF (80 ml) was stirred at room temperature for 3h. The reaction mixture was poured into water (500 mL) to give a yellow precipitate. The mixture was extracted with DCM (4×100 mL) and the combined extracts were washed with water and brine, dried (MgSO4) and evaporated under reduced pressure to give a viscous yellow oil. Purification by column chromatography [biotage isolera, gradient elution hexane 60%/EtOAc 40% to EtOAc 100%, 8 column volumes 100 g snap ultra® cartridge] gave the product as a yellow foam (11.8 g, 88%). Analytical Data: RT 2.20 min; MS (ES+) m/z (relative intensity) 885 ([M+H]+, 100), 907 ([M+Na]+, 50)


(vii) ((Propane-1,3-diylbis(oxy))bis(2-amino-5-methoxy-4,1-phenylene))bis(((S)-2-(((tert-butyldimethylsilyl)oxy)methyl)-4-methylenepyrrolidin-1-yl)methanone) (11)

Zinc powder (31.9 g, 488 mmol, 40 eq.) was activated by stirring/sonication with 1M HCl for 10 min. The Zinc was filtered washing with 1M HCl, water (×3) and MeOH (×2). The activated Zinc was added to a solution of the nitro-TBS compound 10 (10.8 g, 12.2 mmol, 1 eq.) in MeOH (88 mL) and 5% formic acid/MeOH solution (440 mL). The temperature rose to 37° C. and the reaction mixture changed from a yellow to a colourless solution. Once the exotherm had subsided (20 min.) the reaction was shown to be complete by LCMS. The reaction mixture was filtered through celite washing with EtOAc. The EtOAc portion was washed with saturated bicarbonate solution (×4) [caution effervescence!], water (×1), brine (×1), dried (MgSO4) and evaporated under reduced pressure to give a yellow solid.


Purification by flash column chromatography [n-hexane/EtOAc 50/50 v/v to EtOAc 100% in 10% increments] gave the product as a yellow foam (9.5 g, 86%). Analytical Data: RT 2.12 min; MS (ES+) m/z (relative intensity) 825 ([M+H]+, 60), 847 ([M+Na]+, 30)


(viii) tert-Butyl (5-(3-(5-amino-4-((S)-2-(((tert-butyldimethylsilyl)oxy)methyl)-4-methylenepyrrolidine-1-carbonyl)-2-methoxyphenoxy)propoxy)-2-((S)-2-(((tert-butyldimethylsilyl)oxy)methyl)-4-methylenepyrrolidine-1-carbonyl)-4-methoxyphenyl)carbamate (12)

A solution of the bis-aniline 11 (3.27 g, 3.96 mmol) and di-t-butyldicarbonate (0.85 g, 3.96 mmol) in dry THF (125 mL) were heated under reflux for 24 h. The reaction mixture was cooled and the solvent evaporated under reduced pressure. The residue was purified by flash column chromatography [n-hexane/EtOAc 50/50 v/v to EtOAc 100% in 10% increments then EtOAc/MeOH 98/2 v/v] to give the desired product as a yellow foam (1.63 g, 44%). Analytical Data: RT 2.28 min; MS (ES+) m/z (relative intensity) 925 ([M+H]+, 70), 947 ([M+Na]+, 100)


(c) Alloc-Val-Ala-PABOH (17)



embedded image


(i) Alloc-Val-OH (14)

Allyl chloroformate (41 g, 36.2 mL, 0.34 mol, 1.2 eq.) was added dropwise to a stirred solution of L-valine 13 (33.25 g, 0.28 mol, 1 eq.) and potassium carbonate (58.9 g, 0.426 mol, 1.5 eq.) in water (650 mL) and THF (650 mL). The reaction mixture was stirred at room temperature for 18 h. The THF was evaporated under reduced pressure and the remaining solution was extracted with diethyl ether (or MTBE) (×2). The aqueous portion was acidified to pH 2 with conc. HCl and extracted with DCM (×3). The combined organic extracts were washed with brine (×1), dried (MgSO4) and evaporated under reduced pressure to give a colourless oil (57.1 g). This was used in the next step without further purification.


(ii) Alloc-Val-OSu (15)

To a stirred solution of compound 14 (57.1 g, 0.28 mol, 1 eq.) and N-hydroxysuccinimide (32.68 g, 0.28 mol, 1 eq.) in dry THF (800 mL) was added dicyclohexylcarbodiimide (58.6 g, 0.28 mol, 1 eq.). The reaction mixture was stirred at room temperature for 18h. The reaction mixture was filtered. The solid was washed with THF and the combined filtrate was concentrated under reduced pressure. The oil/solid residue was re-dissolved in DCM and left to stand at 0° C. for 30 min. The suspension was filtered washing with cold DCM. Evaporation of the filtrate under reduced pressure gave the succinimide ester as a white solid which was used in the next step without further purification.


(iii) Alloc-Val-Ala-OH (16)

A solution of Alloc-Val-OSu 15 (11.67 g, 39.0 mmol, 1 eq.) in THF (50 mL) was added to a solution of H-Ala-OH (3.66 g, 41.08 mmoL, 1.05 eq.) and NaHCO3 (3.61 g, 43.03 mmol, 1.1 eq.) in THF (100 mL) and H2O (100 mL). The mixture was stirred at room temperature for 72 h and the THF was evaporated under reduced pressure. The pH was adjusted to 3-4 with citric acid to precipitate a white gum. This was extracted with ethylacetate (6×150 mL) and the combined extracts were washed with H2O (200 mL), brine (200 mL), dried (MgSO4) and evaporated under reduced pressure to give a white solid. Trituration with diethyl ether (xs) afforded the pure product as a white powder (7.93 g, 74%). Analytical Data: RT 2.17 min; MS (ES+) m/z (relative intensity) 295 ([M+Na]+, 63), 273 ([M+1]+, 60).


(iv) Alloc-Val-Ala-PABOH (17)

EEDQ (4.79 g, 19.3 mmol, 1.05 eq.) was added to a solution of p-aminobenzyl alcohol (2.38 g, 19.3 mmol, 1.05 eq.) and Alloc-Val-Ala-OH 16 (5.02 g, 18.4 mmol, 1.0 eq) in dry THF (100 mL). The mixture was stirred at room temperature for 72 h. The solvent was evaporated under reduced pressure to give a pale brown solid. The solid was triturated with diethyl ether and filtered washing with an excess of diethyl ether. This afforded the product as a white solid (6.2 g, 89%). Analytical Data: RT 2.50 min; MS (ES+) m/z (relative intensity) 400.6 ([M+Na]+, 50), 378.6 ([M+1]+, 60).


(d) 4-((2S,5S)-37-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)-5-isopropyl-2-methyl-4,7,35-trioxo-10,13,16,19,22,25,28,31-octaoxa-3,6,34-triazaheptatriacontanamido)benzyl (11S,11aS)-11-hydroxy-7-methoxy-8-(3-(((S)-7-methoxy-2-methylene-5-oxo-2,3,5,11a-tetrahydro-1H-benzo[e]pyrrolo[1,2-a][1,4]diazepin-8-yl)oxy)propoxy)-2-methylene-5-oxo-2,3,11,11a-tetrahydro-1H-benzo[e]pyrrolo[1,2-a][1,4]diazepine-10(5H)-carboxylate (23)



embedded image


embedded image


embedded image


(i) tert-butyl (5-(3-(5-((((4-((S)-2-((S)-2-(((allyloxy)carbonyl)amino)-3-methylbutanamido)propanamido)benzyl)oxy)carbonyl)amino)-4-((S)-2-(((tert-butyldimethylsilyl)oxy)methyl)-4-methylenepyrrolidine-1-carbonyl)-2-methoxyphenoxy) propoxy)-2-((S)-2-(((tert-butyldimethylsilyl)oxy)methyl)-4-methylenepyrrolidine-1-carbonyl)-4-methoxyphenyl)carbamate (18)

Triethylamine (0.38 g, 0.53 mL, 3.8 mmol, 2.2 eq.) was added to a stirred solution of the mono-boc protected bis-aniline (12) (1.6 g, 1.72 mmol, 1.0 eq.) and triphosgene (0.184 g, 0.62 mmol. 0.36 eq.) in dry THF (25 mL) under a nitrogen atmosphere at room temperature. The reaction mixture was heated to 40° C., after 5 min a sample was treated with methanol and analysed by LCMS as the methyl carbamate. Analytical Data: RT 2.32 min; MS (ES+) m/z (relative intensity) 983 ([M+H]+, 55), 1005 ([M+Na]+, 100) A solution/suspension of the benzyl-alcohol (17) (1.52 g, 2.35 mmol, 1.4 eq.) and triethylamine (0.26 g, 0.36 mL 2.6 mmol, 1.5 eq.) in dry THF (40 mL) was run in from a dropping funnel to the freshly prepared isocyanate. The reaction mixture was stirred at 40° C. for 2.5 h. The reaction mixture was allowed to cool, filtered and the filtrate evaporated to dryness to afford the crude product as a yellow oil which was purified by flash column chromatography [n-hexane/EtOAc 50/50 v/v] which gave the product as a yellow glass (1.192 g). The mixed fractions were purified by flash column chromatography [CHCl3/MeOH 0% to 1%] to give a further amount of product (0.22 g). The material was combined to give the product as a yellow foam (1.41 g, 63%). Analytical Data: RT 2.27 min; MS (ES+) m/z (relative intensity) 1328 ([M+H]+,30), 1350 ([M+Na]+, 100)


(ii) tert-butyl (5-(3-(5-((((4-((S)-2-((S)-2-(((allyloxy)carbonyl)amino)-3-methylbutanamido)propanamido)benzyl)oxy)carbonyl)amino)-4-((S)-2-(hydroxymethyl)-4-methylenepyrrolidine-1-carbonyl)-2-methoxyphenoxy)propoxy)-2-((S)-2-(hydroxymethyl)-4-methylenepyrrolidine-1-carbonyl)-4-methoxyphenyl)carbamate (19)

A 1.0M solution of TBAF in THF (2.34 mL, 2.34 mmol, 2.2 eq.) was added to a solution of the bis-TBS compound (18) (1.41 g, 1.06 mmol, 1.0 eq.) in anhydrous THF (12 mL). The mixture was stirred at room temperature for 30 min., the solvent was removed under reduced pressure and the residue purified by flash column chromatography [CHCl3/MeOH 0% to 4% in 1% increments] to give the desired product as a white foam (0.98 g, 84%). Analytical Data: RT 1.62 min; MS (ES+) m/z (relative intensity) 1100 ([M+H]+,60), 1122 ([M+Na]+, 100)


(iii) 4-((S)-2-((S)-2-(((allyloxy)carbonyl)amino)-3-methylbutanamido)propanamido)benzyl (11S,11aS)-8-(3-(((11S,11aS)-10-(tert-butoxycarbonyl)-11-hydroxy-7-methoxy-2-methylene-5-oxo-2,3,5,10,11,11a-hexahydro-1H-benzo[e]pyrrolo[1,2-a][1,4]diazepin-8-yl)oxy)propoxy)-11-hydroxy-7-methoxy-2-methylene-5-oxo-2,3,11,11a-tetrahydro-1H-benzo[e]pyrrolo[1,2-a][1,4]diazepine-10(5H)-carboxylate (20)

IBX (45 wt %, 1.3 g, 2.09 mmol, 2.4 eq.) was added to a solution of the bis-alcohol 19 (0.959 g, 0.87 mmol, 1.0 eq.) in anhydrous DMSO (25 mL). The solution was stirred at 30° C. for 18h. LCMS analysis indicated the presence of a small amount of partially cyclised material. A further portion of IBX (45 wt %, 0.049 g, 0.17 mmol, 0.2 eq.) was added and the reaction was continued for a further 18 h. The reaction mixture was poured into water (200 mL) and the resultant precipitate was collected by filtration washing with water. The precipitate was dissolved in DCM (150 mL) and washed with saturated NaHCO3 (100 mL), water (100 mL) and brine (100 mL). The organic portion was dried (MgSO4) and evaporated to give a white solid. Purification by flash column chromatography [CHCl3/MeOH 0% to 4% in 1% increments] gave the product as a white solid (0.696 g, 73%). Analytical Data: RT 1.55 min; MS (ES+) m/z (relative intensity) 1096 ([M+H]+,20), 1118 ([M+Na]+, 100)


(iv) 4-((S)-2-((S)-2-amino-3-methylbutanamido)propanamido)benzyl (11S,11aS)-8-(3-(((11S,11aS)-10-(tert-butoxycarbonyl)-11-hydroxy-7-methoxy-2-methylene-5-oxo-2,3,5,10,11,11a-hexahydro-1H-benzo[e]pyrrolo[1, 2-a][1,4]diazepin-8-yl)oxy)propoxy)-11-hydroxy-7-methoxy-2-methylene-5-oxo-2,3,11,11a-tetrahydro-1H-benzo[e]pyrrolo[1,2-a][1,4]diazepine-10(5H)-carboxylate (21)

Pd(PPh3)4 (14 mg, 12.28 μmol, 0.02 eq.) was added to a solution of the cyclised product 20 (0.673 g, 0.61 mmol, 1.0 eq.) and pyrrolidine (55 mg, 63 μL, 0.8 mmol, 1.25 eq.) in anhydrous DCM (30 mL). The solution was stirred at room temperature for 30 min. The reaction mixture was diluted with DCM (70 mL) and washed with saturated NH4Cl (100 mL), saturated brine (100 mL), dried (MgSO4) and evaporated to give an off white foam. The product was triturated with diethyl ether and dried to give the product (0.62 g, 100%) which was used without further purification. Analytical Data: RT 1.16 min; MS (ES+) m/z (relative intensity) 1012 ([M+H]+,80), 1034 ([M+Na]+, 20) (v) tert-butyl (11S,11aS)-8-(3-(((11S,11aS)-10-(((4-((2S,5S)-37-(2,5-dioxo-2, 5-dihydro-1H-pyrrol-1-yl)-5-isopropyl-2-methyl-4,7,35-trioxo-10,13,16,19,22,25,28,31-octaoxa-3,6,34-triazaheptatriacontanamido)benzyl)oxy)carbonyl)-11-hydroxy-7-methoxy-2-methylene-5-oxo-2,3,5,10,11,11a-hexahydro-1H-benzo[e]pyrrolo[1,2-a][1,4]diazepin-8-yl)oxy)propoxy)-11-hydroxy-7-methoxy-2-methylene-5-oxo-2,3,11,11a-tetrahydro-1H-benzo[e]pyrrolo[1,2-a][1,4]diazepine-10(5H)-carboxylate (22)


EDCI.HCl (0.13 g, 0.66 mmol, 1.1 eq.) was added to a cloudy solution of compound 21 (0.61 g, 0.6 mmol, 1.0 eq.) and Mal-dPEG8®-OH (0.393 g, 0.66 mmol, 1.1 eq.) in CHCl3 (25 mL). The clear solution was stirred at room temperature for 1.5 h., diluted with CHCl3 (100 mL) washed with brine (2×100 mL), dried (MgSO4) and evaporated under reduced pressure to give a yellow foam. Purification by flash column chromatography [CHCl3/MeOH 0% to 6% in 1% increments gave the product as a white foam (0.786 g, 82%). Analytical Data: RT 1.44 min; MS (ES+) m/z (relative intensity) 1586 ([M+H]+,40), 1609 ([M+Na]+, 100)


(vi) 4-((2S,5S)-37-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)-5-isopropyl-2-methyl-4,7,35-trioxo-10,13,16,19,22,25,28,31-octaoxa-3,6,34-triazaheptatriacontanamido)benzyl (11S,11aS)-11-hydroxy-7-methoxy-8-(3-(((S)-7-methoxy-2-methylene-5-oxo-2,3,5,11a-tetrahydro-1H-benzo[e]pyrrolo[1,2-a][1,4]diazepin-8-yl)oxy)propoxy)-2-methylene-5-oxo-2,3,11,11a-tetrahydro-1H-benzo[e]pyrrolo[1,2-a][1,4]diazepine-10(5H)-carboxylate (23)

Ice cold 95% TFA(aq) solution (10 mL) was added to the Boc protected compound 22 (0.759 g, 0.48 mmol, 1.0 eq) which had been cooled to 0° C. (ice bath). The yellow solution was stirred at 0° C. for 1h. The reaction mixture was poured onto ice/water (200 mL) and the mixture was basified to pH 8 with solid NaHCO3. The mixture was extracted with DCM (4×50 mL) and the combined extracts washed with brine (100 mL), dried (MgSO4) and evaporated under reduced pressure. The product was purified by flash column chromatography [CHCl3/MeOH 0% to 8% in 1% increments] to give a pale yellow foam (0.445 g, 65%). Analytical Data: RT 1.37 min; MS (ES+) m/z (relative intensity) 1468 ([M+H]+,40)


Example 2
Alternate synthesis of tert-butyl (5-(3-(5-((((4-((S)-2-((S)-2-(((allyloxy)carbonyl)amino)-3-methylbutanamido)propanamido)benzyl)oxy)carbonyl)amino)-4-((S)-2-(hydroxymethyl)-4-methylenepyrrolidine-1-carbonyl)-2-methoxyphenoxy)propoxy)-2-((S)-2-(hydroxymethyl)-4-methylenepyrrolidine-1-carbonyl)-4-methoxyphenyl)carbamate (19)



embedded image


(i)((2S,2'S)-(4,4′-(propane-1,3-diylbis(oxy))bis(5-methoxy-2-nitrobenzoyl)) bis(4-methylenepyrrolidine-1,2-diyl))bis(methylene) diacetate (24)

A solution of acetyl chloride (21.1 mL, 23.3 g, 297 mmol) in DCM (100 mL) was added drop-wise over a period of 20 min to a stirred solution of bis-alcohol (9) (75 g, 114 mmol) and triethylamine (34.7 g, 343 mmol) in anhydrous DCM (900 mL) at 0-5° C. under a nitrogen atmosphere. The reaction mixture was allowed to warm to room temperature and stirred for a further 60 mins. The reaction mixture was washed with ice cold 0.5M HCl (500 mL), saturated aqueous sodium hydrogen carbonate (250 mL), brine (100 mL) and dried (MgSO4). Removal of the solvent by rotary evaporation gave a pale yellow foam which was used in the next step without further purification. Yield=66.7 g (79%). Analytical Data: Purity satisfactory by LC/MS (7.60 min (ES+) m/z (relative intensity) 741.2 ([M+1]+, 60) 763.3 ([M+Na]+, 100)); 1H NMR (400 MHz, CDCl3) δ δ7.73 (s, 2H), 6.83 (s, 2H), 5.12 (d, 2H, J=12 Hz), 5.02 (s, 2H), 4.89 (s, 2H), 4.79 (m, 2H), 4.61 (m, 1H), 4.35 (m, 6H), 3.98 (s, 6H), 3.87 (d, 1H, J=4.0 Hz), 3.76 (m, 3H), 2.87-2.83 (m, 2H), 2.56-2.43 (m, 4H), 2.05 (s, 4H), 1.96 (s, 2H).


(ii) ((2S,2'S)-(4,4′-(propane-1,3-diylbis(oxy))bis(2-amino-5-methoxybenzoyl)) bis(4-methylenepyrrolidine-1,2-diyl))bis(methylene) diacetate (25)

A 10% solution of formic acid in methanol (500 mL) was added in one go, via a separating funnel, to a solution of the bis-alcohol (24) (66 g, 0.09 mol) in methanol (1000 mL) containing zinc* (145 g, 2.22 mol) at room temperature. The temperature of the reaction mixture rapidly rose to 42° C. and was then cooled back to room temperature with the aid of a cold water bath. The excess zinc was removed by filtering through a short bed of celite, which was then washed with ethyl acetate (100 mL). The filtrate was diluted with ethyl acetate (1400 mL) and washed with saturated sodium hydrogen carbonate (1500 mL), water (500 mL), brine (100 mL) and dried (MgSO4). Removal of the solvent by rotary evaporation gave a yellow solid which was purified by column chromatography (4% methanol/DCM) to give the product as a pale yellow foam. Yield=38.1 g (63%). Analytical Data: Purity satisfactory by LC/MS (6.61 min (ES+) m/z (relative intensity) 681.2 ([M+1]+, 100)); 1H NMR (400 MHz, CDCl3) δ δ6.74 (s, 2H), 6.31 (s, 2H), 5.02 (bs, 2H), 4.97 (bs, 2H), 4.80 (s, 2H), 4.33-4.10 (m, 16H), 3.78 (s, 3H), 2.78 (m, 2H), 2.46 (m, 2H), 2.34 (m, 2H), 2.04 (s, 6H).


(iii) ((S)-1-(4-(3-(4-((S)-2-(acetoxymethyl)-4-methylenepyrrolidine-1-carbonyl)-5-((tert-butoxycarbonyl)amino)-2-methoxyphenoxy)propoxy)-2-amino-5-methoxybenzoyl)-4-methylenepyrrolidin-2-yl)methyl acetate (26)

Boc anhydride (21.6 g, 31.7 mmol) was added to a solution of the diamine (25) (6.92 g 31.7 mmol) in THF (200 mL) at room temperature. The resulting solution was then heated at reflux for 3 hours, cooled and evaporated to dryness under reduced pressure. The resulting residue was purified by column chromatography (70-100% ethyl acetate/hexane) to give the product as a pale yellow solid. Yield=8.4 g (34%). Analytical Data: Purity satisfactory by LC/MS (3 min run) (1.64 min (ES+) m/z (relative intensity) 781.2 ([M+Na]+, 30)); 1H NMR (400 MHz, CDCl3) δ 8.32 (bs, 1H), 7.87 (s, 1H), 6.80 (s, 1H), 6.73 (s, 1H), 5.02 (m, 3H), 4.79 (3H), 4.34-4.09 (m, 14H), 3.83 (s, 3H), 3.78 (s, 3H), 2.81-2.74 (m, 2H), 2.48-2.36 (m, 4H), 2.04 (m, 7H), 1.49 (s, 9H).


(iv) ((S)-1-(4-(3-(4-((S)-2-(acetoxymethyl)-4-methylenepyrrolidine-1-carbonyl)-5-((((4-((S)-2-((S)-2-(((allyloxy)carbonyl)amino)-3-methylbutanamido)propanamido)benzyl) oxy)carbonyl)amino)-2-methoxyphenoxy)propoxy)-2-((tert-butoxycarbonyl)amino)-5-methoxybenzoyl)-4-methylenepyrrolidin-2-yl)methyl acetate (27)

Triethyl amine (0.57 g, 5.6 mmol) was added in one go to a solution of the amine (26) (2 g, 2.56 mmol) and triphosgene (0.27 g, 0.92 mmol) in THF (30 mL) under nitrogen. The resulting mixture was heated at 40° C. for 5 min. A small aliquot was quenched with methanol, and LCMS indicated complete conversion to the methyl carbamate (m/z 983, M+1). A slurry of SG3366 (2.25 g, 3.48 mmol) and triethyl amine (0.39 g, 3.84 mmol) in THF (50 mL) was added in one go and the resulting mixture heated at 40° C. for 4 hours.


After cooling, the white solid was removed by filtration and the filtrate evaporated to dryness under reduced pressure, and purified by column chromatography (1-3% methanol/DCM) to give the product as a pale yellow solid. Yield=2.1 g (69%). Analytical Data: Purity satisfactory by LC/MS (8.26 min (ES+) m/z (relative intensity) 1184.3 ([M+1]+, 70), 1206.3 ([M+Na]+, 100)); 1H NMR (400 MHz, DMSO-d6) δ 7.62 (s, 2H), 7.30 (s, 2H), 4.29 (t, 4H, J=6.0 Hz), 3.85 (s, 6H), 2.30-2.26 (m, 2H).


(v) ((S)-1-(4-(3-(4-((S)-2-(acetoxymethyl)-4-methylenepyrrolidine-1-carbonyl)-5-((((4-((S)-2-((S)-2-(((allyloxy)carbonyl)amino)-3-methylbutanamido)propanamido)benzyl) oxy)carbonyl)amino)-2-methoxyphenoxy)propoxy)-2-((tert-butoxycarbonyl)amino)-5-methoxybenzoyl)-4-methylenepyrrolidin-2-yl)methyl acetate (19)

Potassium carbonate (1.16 g, 8.44 mmol) was dissolved in water (8.4 mL) and added to a solution of the diacetate (27) (2.0 g, 1.69 mmol) in methanol (40 mL). The resulting mixture was stirred at 25° C. for 30 mins, then evaporated to dryness under reduced pressure. The resulting residue was taken up in water (100 mL), acidified (pH 3) with 1M citric acid and extracted with ethyl acetate (3×100 mL). The combined extracts were washed with water (100 mL), brine (30 mL) and dried (MgSO4). Removal of the solvent under reduced pressure left the product as an off-white solid which was used in the next step without further purification. Yield=1.6 g (87%). Analytical Data: Purity satisfactory by LC/MS (7.32 min (ES+) m/z (relative intensity) 1100.7 ([M+1]+, 50), 1122.3 ([M+Na]+, 100)); 1H NMR (400 MHz, DMSO-d6) δ9.98 (bs, 1H), 9.09 (bs, 1H), 8.73 (bs, 1H), 8.14 (d, J=8 Hz, 1H), 7.59 (d, J=8 Hz, 2H), 7.33 (d, J=8 Hz, 2H), 7.21 (m, 3H), 6.90 (bs, 2H), 5.91 (m, 1H), 5.30 (d, J=4 Hz, 1H), 5.19 (d, J=4 Hz, 1H), 5.00 (m, 6H), 4.70-4.35 (m, 6H), 4.15-3.88 (m, 12H), 3.77 (s, 3H), 3.67 (s, 3H), 2.82-2.67 (m, 2H), 2.42 (m, 3H), 2.21 (t, J=4 Hz, 2H), 1.98 (m, 6H), 1.42 (s, 9H), 1.31 (d, J=8 Hz, 3H), 0.90 (d, J=4 Hz, 3H), 0.84 (d, J=4 Hz, 3H).


Example 3



embedded image


(a) Allyl (5-(3-(5-amino-4-((S)-2-(((tert-butyldimethylsilyl)oxy)methyl)-4-methylenepyrrolidine-1-carbonyl)-2-methoxyphenoxy) propoxy)-2-((S)-2-(((tert-butyldimethylsilyl)oxy)methyl)-4-methylenepyrrolidine-1-carbonyl)-4-methoxyphenyl)carbamate (28)

A solution of allyl chloroformate (1.05 g, 0.9 mL, 8.7 mmol, 0.9 eq.) was added drop wise to a solution of bis aniline (11)(8.02 g, 9.7 mmol, 1 eq.) and pyridine (1.15 g, 1.2 mL, 14.55 mmol, 1.9 eq.) in dry DCM (350 mL) at −78° C. (dry ice/acetone bath). The reaction mixture was stirred at −78° C. for 1 hour and then allowed to reach room temperature. The reaction mixture was washed with saturated aqueous copper sulphate solution (250 mL), water (250 mL), saturated sodium bicarbonate (250 mL), brine (250 mL) and dried (MgSO4). Rotary evaporation under reduced pressure afforded the crude product. Purification by flash chromatography [50% n-hexane/50% ethyl acetate, to 20% n-hexane/80% ethyl acetate to 100% ethyl acetate to 1% methanol/99% ethyl acetate] gave the bis-alloc product (2.066 g), the desired mono-alloc product (4.33 g, 49%) and recovered bis-aniline (1.96 g). Analytical Data: RT 2.26 min; MS (ES+) m/z (relative intensity) 909 ([M+1]+, 100); 931 ([M+Na]+, 100)


(b) 4-((S)-2-((S)-2-(((allyloxy)carbonyl)amino)-3-methylbutanamido)propanamido)benzyl (5-(3-(5-(((allyloxy)carbonyl)amino)-4-((S)-2-(((tert-butyldimethylsilyl)oxy)methyl)-4-methylenepyrrolidine-1-carbonyl)-2-methoxyphenoxy)propoxy)-2-((S)-2-(((tert-butyldimethylsilyl)oxy)methyl)-4-methylenepyrrolidine-1-carbonyl)-4-methoxyphenyl)carbamate (29)

Triethylamine (1.22 g, 1.7 mL, 12.1 mmol, 2.2 eq.) was added to a stirred solution of the mono-boc protected bis-aniline (28) (5.0 g, 5.5 mmol, 1.0 eq.) and triphosgene (0.59 g, 1.98 mmol, 0.36 eq.) in dry THF (75 mL) under a nitrogen atmosphere at room temperature. The reaction mixture was heated to 40° C., after 5 min a sample was treated with methanol and analysed by LCMS as the methyl carbamate. Analytical Data: RT 2.30 min; MS (ES+) m/z (relative intensity) 967 ([M+H]+, 25), 989 ([M+Na]+, 100)


A solution/suspension of the benzyl-alcohol (17) (3.11 g, 8.25 mmol, 1.5 eq.) and triethylamine (0.83 g, 1.1 mL 2.6 mmol, 1.5 eq.) in dry THF (75 mL) was run in from a dropping funnel to the freshly prepared isocyanate. The reaction mixture was stirred at 40° C. for 5h. then overnight at room temperature The reaction mixture was allowed to cool, filtered and the filtrate evaporated to dryness to afford the crude product as a yellow oil which was purified by flash column chromatography [50% n-hexane/50% ethyl acetate to 40% n-hexane/60% ethyl acetate] which gave the product as a yellow glass (1.25 g, 17%). Analytical Data: RT 2.26 min; MS (ES+) m/z (relative intensity) 1312 ([M+H]+, 25), 1335 ([M+Na]+, 35)


(c) 4-((S)-2-((S)-2-(((allyloxy)carbonyl)amino)-3-methylbutanamido)propanamido)benzyl (5-(3-(5-(((allyloxy)carbonyl)amino)-4-((S)-2-(hydroxymethyl)-4-methylenepyrrolidine-1-carbonyl)-2-methoxyphenoxy)propoxy)-2-((S)-2-(hydroxymethyl)-4-methylenepyrrolidine-1-carbonyl)-4-methoxyphenyl)carbamate (30)

A 1.0M solution of TBAF in THF (5.2 mL, 5.2 mmol, 2.2 eq.) was added to a solution of the bis TBS compound (29) (3.096 g, 2.36 mmol, 1.0 eq.) in anhydrous THF (25 mL). The mixture was stirred at room temperature for 30 min., the solvent was removed under reduced pressure and the residue purified by flash column chromatography [ethyl acetate/methanol 0% to 6% in 1% increments] which gave the desired product as a white foam (1.91 g, 75%). Analytical Data: RT 1.56 min; MS (ES+) m/z (relative intensity) 1084 ([M+H]+, 100), 1106 ([M+Na]+, 90)


(d) Allyl (11S,11aS)-8-(3-(((11S,11aS)-10-(((4-((S)-2-((S)-2-(((allyloxy)carbonyl)amino)-3-methylbutanamido)propanamido)benzyl)oxy)carbonyl)-11-hydroxy-7-methoxy-2-methylene-5-oxo-2,3,5,10,11,11a-hexahydro-1H-pyrrolo[2,1-c][1,4]benzodiazepin-8-yl)oxy)propoxy)-11-hydroxy-7-methoxy-2-methylene-5-oxo-2,3,11,11a-tetrahydro-1H-pyrrolo[2,1-c][1,4]benzodiazepine-10(5H)-carboxylate (31)

IBX (45 wt %, 2.06 g, 3.3 mmol, 2.4 eq.) was added to a solution of the bis-alcohol 30 (1.49 g, 1.38 mmol, 1.0 eq.) in anhydrous DMSO (40 mL). The solution was stirred at 30° C. for 18h. LCMS analysis indicated the presence of a small amount of partially cyclised material. A further portion of IBX (45 wt %, 0.171 g, 0.275 mmol, 0.2 eq.) was added and the reaction was continued for a further 24 h. The reaction mixture was poured into water (200 mL) and the resultant precipitate was collected by filtration washing with water. The precipitate was dissolved in DCM (150 mL) and washed with saturated NaHCO3 (100 mL), water (100 mL) and brine (100 mL). The organic portion was dried (MgSO4) and evaporated to give a white solid. Purification by flash column chromatography [CHCl3/MeOH 0% to 3% in 1% increments] gave the product as a white solid (1.06 g, 72%). Analytical Data: RT 6.88 min; MS (ES+) m/z (relative intensity) 1080 ([M+H]+, 50), 1102 ([M+Na]+, 100)


(e) 4-((S)-2-((S)-2-amino-3-methylbutanamido)propanamido)benzyl (11S,11aS)-11-hydroxy-7-methoxy-8-(3-(((S)-7-methoxy-2-methylene-5-oxo-2,3,5,11a-tetrahydro-1H-pyrrolo[2,1-c][1,4]benzodiazepin-8-yl)oxy)propoxy)-2-methylene-5-oxo-2,3,11,11a-tetrahydro-1H-pyrrolo[2,1-c][1,4]benzodiazepine-10(5H)-carboxylate (32)

Pd(PPh3)4 (44 mg, 38.5 μmol, 0.04 eq.) was added to a solution of the cyclised product 31 (1.04 g, 0.96 mmol, 1.0 eq.) and pyrrolidine (0.171 mg, 196 μL, 2.4 mmol, 2.5 eq.) in anhydrous DCM (30 mL). The solution was stirred at room temperature for 30 min. The reaction mixture was diluted with DCM (30 mL) and washed with saturated NH4Cl (100 mL), saturated brine (100 mL), dried (MgSO4) and evaporated to give an off white foam. The product was triturated with diethyl ether and dried to give the product (0.86 g, 100%) which was used without further purification. Analytical Data: RT 1.10 min; MS (ES+) m/z (relative intensity) 894 ([M+H]+, 30)


(f) 4-((2S,5S)-37-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)-5-isopropyl-2-methyl-4,7,35-trioxo-10,13,16,19,22,25,28,31-octaoxa-3,6,34-triazaheptatriacontanamido)benzyl (11S,11aS)-11-hydroxy-7-methoxy-8-(3-(((S)-7-methoxy-2-methylene-5-oxo-2,3,5,11a-tetrahydro-1H-pyrrolo[2,1-c][1,4]benzodiazepin-8-yl)oxy)propoxy)-2-methylene-5-oxo-2,3,11,11a-tetrahydro-1H-pyrrolo[2,1-c][1,4]benzodiazepine-10(5H)-carboxylate (23)

EDCI.HCl (0.203 g, 1.06 mmol, 1.1 eq.) was added to a solution of compound 32 (0.86 g, 0.96 mmol, 1.0 eq.) and Mal-dPEG8®-OH (0.57 g, 0.96 mmol, 1.1 eq.) in dry DCM (30 mL) and CHCl3 (to give a clear solution). The clear solution was stirred at room temperature for 18h. then a further portion of EDCI.HCl (0.037 g, 0.19 mmol, 0.2 eq.) was added and reaction continued for a further 24h. The reaction mixture was diluted with DCM (70 mL) washed with water (100 mL), brine (100 mL), dried (MgSO4) and evaporated under reduced pressure to give a yellow foam. Purification by flash column chromatography [CHCl3/MeOH 0% to 6% in 1% increments] gave the product as an off white foam (0.56 g, 40%). Analytical Data: RT 6.13 min; MS (ES+) m/z (relative intensity) 1468 ([M+H]+, 20)


Example 4



embedded image


embedded image


embedded image




    • (a) Allyl (2-((R)-2-(((tert-butyldimethylsilyl)oxy) methyl)-4-methylenepyrrolidine-1-carbonyl)-5-(3-(4-((R)-2-(((tert-butyldimethylsilyl)oxy)methyl)-4-methylenepyrrolidine-1-carbonyl)-5-5 ((((4-((10S,13S)-10-isopropyl-13-methyl-8, I1-dioxo-2,5-dioxa-9,12-diazatetradecan-14-amido)benzyl)oxy)carbonyl)amino)-2-methoxyphenoxy)propoxy)-4-methoxyphenyl)carbamate (34)





Triethylamine (0.049 g, 0.07 mL, 0.48 mmol, 2.2 eq.) was added to a stirred solution of the mono-alloc protected bis-aniline (23) (0.2 g, 0.22 mmol, 1 0.0 eq.) and triphosgene (0.024 g, 10 0.079 mmol, 0.36 eq.) in dry THF (5 mL) under an argon atmosphere at room temperature. The reaction mixture was heated to 40° C., after 5 min a sample was treated with methanol and analysed by LCMS as the methyl carbamate. Analytical Data: RT 2.27 min, MS (ES+) m/z (relative intensity) 967 ([M+H]+. 80), 989 ([M+Na]+, 100)


A solution/suspension of the benzyl-alcohol (33) (0.121 g, 0.29 mmol, 1.3 eq.) and triethylamine (0.029 g, 0.04 mL 0.29 mmol, 1.3 eq.) in dry THF (5 mL) was run in from a dropping funnel to the freshly prepared isocyanate. The reaction mixture was stirred at 40° C. for 4h. then overnight at room temperature The reaction mixture was allowed to cool, filtered and the filtrate evaporated to dryness to afford the crude product which was purified by flash column chromatography [Biotage Isolera™ CHCl3/MeOH 2% to 4%, gradient elution]. This gave the product (0.237 g, 79%). Analytical Data: RT 2.19 min; MS (ES+) m/z (relative intensity) 1358 ([M+H]+, 30), 1380 ([M+Na]+, 15)


(b) 4-((10S,13S)-10-isopropyl-13-methyl-8,11-dioxo-2,5-dioxa-9,12-diazatetradecan-14-amido)benzyl (5-(3-(5-amino-4-((R)-2-(((tert-butyldimethylsilyl)oxy)methyl)-4-methylenepyrrolidine-1-carbonyl)-2-methoxyphenoxy)propoxy)-2-((R)-2-(((tert-butyldimethylsilyl)oxy)methyl)-4-methylenepyrrolidine-1-carbonyl)-4 -methoxyphenyl)carbamate (35)

Pd(PPh3)4 (0.3 g, 0.25 mmol, 0.06 eq.) was added to a solution of the alloc protected intermediate 34 (5.89 g, 4.3 mmol, 1.0 eq.) and pyrrolidine (0.46 g, 530 μL, 6.5 mmol, 1.5 eq.) in anhydrous DCM (50 mL). The solution was stirred at room temperature for 1h. The reaction mixture was diluted with DCM and washed with saturated NH4Cl, saturated brine, dried (MgSO4) and evaporated to give crude product. The product was purified by flash column chromatography [Biotage Isolera™ DCM/MeOH 1% to 3%] to give the product (4.53 g, 83%) which had an overall purity of 80% and was used without further purification. Analytical Data: RT 2.10 min; MS (ES+) m/z (relative intensity) 1275 ([M+H]+, 40).


(c) 4-((S)-2-((S)-2-(((allyloxy)carbonyl)amino)-3-methylbutanamido)propanamido)benzyl (2-((S)-2-(((tert-butyldimethylsilyl)oxy)methyl)-4-methylenepyrrolidine-1-carbonyl)-5-(3-(4-((S)-2-(((tert-butyldimethylsilyl)oxy)methyl)-4-methylenepyrrolidine-1-carbonyl)-5-((((4-((10S,13S)-10-isopropyl-13-methyl-8,11-dioxo-2,5-dioxa-9,12-diazatetradecan-14-amido)benzyl)oxy)carbonyl)amino)-2-methoxyphenoxy)propoxy)-4-methoxyphenyl)carbamate (36)

Triethylamine (0.35 g, 48 μL, 0.34 mmol, 2.2 eq.) was added to a stirred solution of the aniline (35) (0.2 g, 0.157 mmol, 1.0 eq.) and triphosgene (0.017 g, 57 μmol, 0.36 eq.) in dry THF (5 mL) under an argon atmosphere at room temperature. The reaction mixture was heated to 40° C., after 5 min a sample was treated with methanol and analysed by LCMS as the methyl carbamate. Analytical Data: RT 2.15 min; MS (ES+) m/z (relative intensity) 1333 ([M+H]+, 40), 1354 ([M+Na]+, 35).


A solution/suspension of the benzyl-alcohol (17) (0.071 g, 0.19 mmol, 1.2 eq.) and triethylamine (19 mg, 26 μL 0.19 mmol, 1.2 eq.) in dry THF (5 mL) was run in from a dropping funnel to the freshly prepared isocyanate. The reaction mixture was stirred at 40° C. for 4 h. then overnight at room temperature The reaction mixture was filtered and the filtrate evaporated to dryness to afford the crude product which was purified by flash column chromatography [Biotage Isolera™ CHCl3/MeOH 2% to 3%, gradient elution] which gave the product (0.152 g, 58%). Analytical Data: RT 2.12 min; MS (ES+) m/z (relative intensity) 1677 ([M+H]+, 30), 1700 ([M+Na]+, 100).


(d) 4-((S)-2-((S)-2-(((allyloxy)carbonyl)amino)-3-methylbutanamido)propanamido)benzyl (2-((S)-2-(hydroxymethyl)-4-methylenepyrrolidine-1-carbonyl)-5-(3-(4-((S)-2-(hydroxymethyl)-4-methylenepyrrolidine-1-carbonyl)-5-((((4-((10S,13S)-10-isopropyl-13-methyl-8,11-dioxo-2,5-dioxa-9,12-diazatetradecan-14-amido)benzyl)oxy)carbonyl)amino)-2-methoxyphenoxy)propoxy)-4-methoxyphenyl)carbamate (37)

A 1.0M solution of TBAF in THF (3.4 mL, 3.4 mmol, 2.0 eq.) was added to a solution of the bis TBS compound (36) (2.86 g, 1.7 mmol, 1.0 eq.) in anhydrous THF (30 mL) under an argon atmosphere. The mixture was stirred at room temperature for 6h., the reaction mixture was diluted with CHCl3 and washed with water, brine, dried (MgSO4) and evaporated under reduced pressure to give a yellow solid. The residue was purified by flash column chromatography [Biotage Isolera™ CHCl3/MeOH, gradient elution with the product eluting at 4% MeOH] which gave the desired product (1.365 g) and mixed fractions which were further purified by flash column chromatography [CHCl3/MeOH 1% to 5%] to give further product (0.562 g) this gave a combined yield of desired product (1.93 g, 75%). Analytical Data: RT 1.55 min; MS (ES+) m/z (relative intensity) 1449 ([M+1]+, 25); 1471 ([M+Na]+, 20).


(e) 4-((S)-2-((S)-2-(((allyloxy)carbonyl)amino)-3-methylbutanamido)propanamido)benzyl (11S,11aS)-11-hydroxy-8-(3-(11S,11aS)-11-hydroxy(((S)-10-(((4-((10S,13S)-10-isopropyl-13-methyl-8,11-dioxo-2,5-dioxa-9,12-diazatetradecan-14-amido)benzyl)oxy)carbonyl)-7-methoxy-2-methylene-5-oxo-2,3,5,10,11,11a-hexahydro-1H-pyrrolo[2,1-c][1,4]benzodiazepin-8-yl)oxy)propoxy)-7-methoxy-2-methylene-5-oxo-2,3,11,11a-tetrahydro-1H-pyrrolo[2,1-c][1,4]benzodiazepine-10(5H)-carboxylate (38)

IBX (45 wt %, 0.236 g, 0.38 mmol, 2.2 eq.) was added to a solution of the bis-alcohol 37 (0.25 g, 0.17 mmol, 1.0 eq.) in anhydrous DMSO (12 mL). The solution was stirred at 30° C. for 3.5 d. The reaction mixture was poured into water (100 mL) and the resultant precipitate was collected by filtration washing with water. The precipitate was extracted with DCM (5×30 mL) and the combined fractions were washed with saturated NaHCO3 (60 mL), water (60 mL) and brine (60 mL). The organic portion was dried (MgSO4) and evaporated to give crude product. Purification by flash column chromatography [CHCl3/MeOH 1% to 5%] gave the product as a white solid (0.158 g, 64%). Analytical Data: RT 1.53 min; MS (ES+) m/z (relative intensity) 1445 ([M+1]+, 20); 1467 ([M+Na]+, 30).


(f) 4-((S)-2-((S)-2-amino-3-methylbutanamido)propanamido)benzyl (11S,11aS)-11-hydroxy-8-(3-(((11S,11aS)-11-hydroxy-10-(((4-((10S,13S)-10-isopropyl-13-methyl-8,11-dioxo-2,5-dioxa-9,12-diazatetradecan-14-amido)benzyl)oxy)carbonyl)-7-methoxy-2-methylene-5-oxo-2,3,5,10,11,11a-hexahydro-1H-benzo[e]pyrrolo[1,2-a][1,4]diazepin-8-yl)oxy)propoxy)-7-methoxy-2-methylene-5-oxo-2,3,11,11a-tetrahydro-1H-benzo[e]pyrrolo[1,2-a][1,4]diazepine-10(5H)-carboxylate (39)

Pd(PPh3)4 (8 mg, 6.9 μmol, 0.06 eq.) was added to a solution of the cyclised product 38 (0.158 g, 0.109 mmol, 1.0 eq.) and pyrrolidine (0.01 g, 12 μL, 0.15 mmol, 1.5 eq.) in anhydrous DCM (10 mL). The solution was stirred at room temperature for 15 min. The reaction mixture was diluted with CHCl3 and washed with saturated sodium bicarbonate solution, saturated brine, dried (MgSO4) and evaporated to give crude product. The product was triturated with diethyl ether (×3) and dried to give the product (0.136 g, 100%) which was used without further purification. Analytical Data: RT 1.21 min; MS (ES+) m/z (relative intensity) 1361 ([M+1]+, 50); 1384 ([M+Na]+, 10).


(g) 4-((2S,5S)-37-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)-5-isopropyl-2-methyl-4,7,35-trioxo-10,13,16,19,22,25,28,31-octaoxa-3,6,34-triazaheptatriacontanamido)benzyl (11S,11aS)-11-hydroxy-8-(3-(((11S,11aS)-11-hydroxy-10-(((4-((10S,13S)-10-isopropyl-13-methyl-8,11-dioxo-2,5-dioxa-9,12-diazatetradecan-14-amido)benzyl)oxy)carbonyl)-7-methoxy-2-methylene-5-oxo-2,3,5,10,11,11a-hexahydro-1H-pyrrolo[2,1-c][1,4]benzodiazepin-8-yl)oxy)propoxy)-7-methoxy-2-methylene-5-oxo-2,3,11,11a-tetrahydro-1H-pyrrolo[2,1-c][1,4]benzodiazepine-10(5H)-carboxylate (40)

A solution of compound 39 (0.136 g, 0.1 mmol, 1.0 eq.), Mal-dPEG8®-OH (0.066 g, 0.11 mmol, 1.1 eq.) and EDCI.HCl (0.022 g, 0.11 mmol, 1.1 eq.) in dry DCM (10 mL) and MeOH (1 drop) was stirred at room temperature for 1.45 h. The reaction mixture was diluted with CHCl3 and washed with water, brine, dried (MgSO4) and evaporated under reduced pressure to give the crude product. Purification by flash column chromatography [CHCl3/MeOH 1% to 9%] gave the product as a white solid (0.123 g, 63%). Analytical Data: [α]21D=+112.5° (c=0.4, hplc CHCl3); RT 6.37 min; MS (ES+) m/z (relative intensity) 1936 ([M+1]+, 35); 1958 ([M+Na]+, 15).


Example 5—Conjugation

Conjugate trastuzumab-23


A 50 mM solution of tris(2-carboxyethyl)phosphine hydrochloride (TCEP) in phosphate-buffered saline pH 7.4 (PBS) was added (50 molar equivalent/antibody, 35 micromoles, 700 L) to a 24.14 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 4.35 mg/mL. The reduction mixture was heated at +37° C. for 3 hours (or until full reduction is observed by UHPLC) in an incubator with gentle (<150 rpm) shaking. After cooling down to room temperature, the reduced antibody was buffer exchanged, via spin filter centrifugation, into a reoxidation buffer containing PBS pH 7.4 and 1 mM EDTA to remove all the excess reducing agent. A 50 mM solution of dehydroascorbic acid (DHAA, 10 molar equivalent/antibody, 7 micromoles, 140 μL) in DMSO was added and the reoxidation mixture was allowed to react for 16 hours at room temperature with gentle (<150 rpm) shaking at an antibody concentration of 2.3 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 pH 7.4, 1 mM EDTA for a final antibody concentration of 1.0-1.5 mg/mL. Compound 23 (SG3400) was added as a DMSO solution (10 molar equivalent/antibody, 1 micromole, in 1.0 mL DMSO) to 9 mL of this reoxidised antibody solution (15 mg, 100 nanomoles) for a 10% (v/v) final DMSO concentration. The solution was mixed for 1.5 hours at room temperature, then the conjugation was quenched by addition of N-acetyl cysteine (4 micromoles, 40 μL at 100 mM), diluted to >50 mL in PBS and conjugate trastuzumab-23 was 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 Phenomenex Aeris 3.6u XB-C18 150 mm×2.1 mm column eluting with a gradient of water and acetonitrile on a reduced sample of conjugate trastuzumab-A at 280 nm and 330 nm (Compound A specific) shows unconjugated light chains and a mixture of unconjugated heavy chains and heavy chains attached to a single molecule of compound 23, consistent with a drug-per-antibody ratio (DAR) of 1.71 molecules of compound 23 per antibody.


UHPLC analysis on a Shimadzu Prominence system using a Tosoh Bioscience TSKgel G3000SWXL 5 μm 7.8×300 mm column (with a 7 μm 6.0×40 mm guard column) eluting with 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 conjugate trastuzumab-23 at 280 nm shows a monomer purity of 94%. UHPLC SEC analysis gives a concentration of final conjugate trastuzumab-23 at 0.84 mg/mL in 15 mL, obtained mass of conjugate trastuzumab-23 is 12.7 mg (84% yield).


Conjugate trastuzumab-40


A 50 mM solution of tris(2-carboxyethyl)phosphine hydrochloride (TCEP) in phosphate-buffered saline pH 7.4 (PBS) was added trastuzumab, (50 molar equivalent/antibody, 50 micromoles, 1.0 mL) to a 34.5 mL solution of antibody (150 mg, 1.0 micromole) in reduction buffer containing PBS and 1 mM ethylenediaminetetraacetic acid (EDTA) at a final antibody concentration of 4.35 mg/mL. The reduction mixture was heated at +37° C. for 3 hours (or until full reduction is observed by UHPLC) in an incubator with gentle (<150 rpm) shaking. After cooling down to room temperature, the reduced antibody was buffer exchanged, via spin filter centrifugation, into a reoxidation buffer containing PBS and 1 mM EDTA to remove excess reducing agent. A 50 mM solution of dehydroascorbic acid (DHAA, 50 molar equivalent/antibody, 50 micromoles, 1.0 mL) in DMSO was added and the reoxidation mixture was allowed to react for 2 hours at room temperature (or until full reoxidation of the cysteine thiols to reform the inter-chain cysteine disulfides is observed by UHPLC) with gentle (<150 rpm) shaking at an antibody concentration of 2-3 mg/mL. The reoxidation mixture was then sterile-filtered and diluted in a conjugation buffer containing PBS and 1 mM EDTA to a final antibody concentration of ˜1.5 mg/mL. Compound 40 was added as a DMSO solution (10 molar equivalent/antibody, 1 micromole, in 0.9 mL DMSO) to 9 mL of this reoxidised antibody solution (15 mg, 100 nanomoles). The solution was mixed for 1.25 hours at room temperature, after which the conjugation reaction was quenched by addition of N-acetyl cysteine (4 micromoles, 40 μL at 100 mM) and diluted to >50 mL in PBS. The conjugation mixture was purified by spin filtration using a 15 mL Amicon Ultracell 50 kDa MWCO spin filter, sterile-filtered, analysed and stored at +4° C. The reduction and reoxidation steps are monitored by comparison of the relative amounts of individual light and heavy chains with full length antibody as observed by UHPLC analysis on a Shimadzu Prominence system using a PhenomenexAeris 3.6u XB-C18 150×2.1 mm column eluting with a gradient of water and acetonitrile. UHPLC analysis on a Shimadzu Prominence system using a Phenomenex Aeris 3.6u XB-C18 150×2.1 mm column eluting with a gradient of water and acetonitrile on a reduced sample of conjugate trastuzumab-B at 280 nm and 330 nm (Compound 40 specific) shows unconjugated light chains and a mixture of unconjugated heavy chains and heavy chains attached to a single molecule of Compound 40, consistent with a drug-per-antibody ratio (DAR) of 1.68 molecules of Compound 40 per antibody.


UHPLC analysis on a Shimadzu Prominence system using a Tosoh Bioscience TSKgel SuperSW mAb HTP 4 μm 4.6×150 mm column (with a 4 μm 3.0×20 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 conjugate trastuzumab-40 at 280 nm shows a monomer purity of 93% with no impurity detected. UHPLC SEC analysis gives a concentration of final conjugate trastuzumab-B at 0.74 mg/mL in 17 mL, obtained mass of conjugate trastuzumab-40 is 12.5 mg (83% yield).


Conjugate R347-23


A 50 mM solution of tris(2-carboxyethyl)phosphine hydrochloride (TCEP) in phosphate-buffered saline pH 7.4 (PBS) was added (42 molar equivalent/antibody, 56 micromoles, 1.12 mL at 50 mM) to a 14.09 mL solution of antibody (200 mg, 1.33 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 heated at +25° C. for 24 hours (or until full reduction observed by UHPLC) in an incubator with gentle (<100 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 cm2 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 0.45 μM membrane filter. A 50 mM solution of dehydroascorbic acid (DHAA, 15 molar equivalent/antibody, 20 micromoles, 400 μL at 50 mM) in DMSO was added and the reoxidation mixture was allowed to react for 16 hours at room temperature with gentle (<100 rpm) shaking at an antibody concentration of 2.5 mg/mL (or until full reoxidation of the cysteine thiols to reform the inter-chain cysteine disulfides is observed by UHPLC). The reoxidation mixture was centrifuged for 3 min at 4000 rpm and then sterile-filtered using 0.2 μM membrane filter. Compound 23 was added as a DMSO solution (10 molar equivalent/antibody, 13.3 micromoles, in 6.6 mL DMSO) to 80 mL of this reoxidised antibody solution (200 mg, 1.33 micromoles) for a 10% (v/v) final DMSO concentration. The solution was shaken for 3 hours at +25° C. and then the conjugation was quenched with N-acetyl cysteine (72.3 micromoles, 0.72 mL at 100 mM).


Excess free drug was removed via Tangential Flow Filtration unit (TFF) using mPES, MidiKros® 30 kDa fiber filter with 115 cm2 surface area, into buffer containing PBS pH 7.4. Extent of free drug removal was monitored by UHPLC-RP using neat conjugate. After complete removal of free drug, ADC were formulated onto 25 mM Histidine, 200 mM Sucrose, pH 6.0, via TFF using mPES, MidiKros® 30 kDa fiber filter with 115 cm2 surface area. The whole process of R347 conjugation with Compound 23 was repeated with 400 mg antibody and also purified using TFF. ADC from both batches were combined and then filtered using Mustang filter under sterile atmosphere and then further stored at −78° C.


UHPLC analysis on a Shimadzu Prominence system using a Phenomenex Aeris 3.6u XB-C18 150×2.1 mm column eluting with a gradient of water and acetonitrile on a reduced sample of Conjugate at 214 nm and 330 nm (Compound 23 specific) shows a mixture of light and heavy chains attached to several molecules of Compound 23, consistent with a drug-per-antibody ratio (DAR) of 1.71 molecules of Compound 23 per antibody.


UHPLC analysis on a Shimadzu Prominence system using a Tosoh Bioscience TSKgel SuperSW mAb HTP 4 μm 4.6×150 mm column (with a 4 μm 3.0×20 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 greater than 97%. UHPLC SEC analysis gives a concentration of final ADC at 1.92 mg/mL in 265 mL, obtained mass of ADC is 509 mg (85% yield).


Conjugate R347-40


A 50 mM solution of tris(2-carboxyethyl)phosphine hydrochloride (TCEP) in phosphate-buffered saline pH 7.4 (PBS) was added (50 molar equivalent/antibody, 20 micromoles, 0.4 mL) to a 13.25 mL solution of antibody (60 mg, 0.4 micromoles) in reduction buffer containing PBS and 1 mM ethylenediaminetetraacetic acid (EDTA) at a final antibody concentration of 4.5 mg/mL. The reduction mixture was heated at +37° C. for 3 hours (or until full reduction is observed by UHPLC) in an incubator with gentle (<150 rpm) shaking. After cooling down to room temperature, the reduced antibody was buffer exchanged, via spin filter centrifugation, into a reoxidation buffer containing PBS and 1 mM EDTA to remove excess reducing agent. A 50 mM solution of dehydroascorbic acid (DHAA, 12 molar equivalent/antibody, 4.8 micromoles, 96 μL) in DMSO was added and the reoxidation mixture was allowed to react for 17 hours at room temperature (or until full reoxidation of the cysteine thiols to reform the inter-chain cysteine disulfides is observed by UHPLC) with gentle (<150 rpm) shaking at an antibody concentration of ˜1.6 mg/mL. The reoxidation mixture was then sterile-filtered and diluted in a conjugation buffer containing PBS and 1 mM EDTA to a final antibody concentration of ˜1.5 mg/mL. Compound 40 was added as a DMSO solution (11 molar equivalent/antibody, 0.44 micromoles, in 0.45 mL DMSO) to 4.05 mL of this reoxidised antibody solution (6 mg, 40 nanomoles). The solution was mixed for 1.25 hours at room temperature, after which the conjugation reaction was quenched by addition of N-acetyl cysteine (1.76 micromoles, 17.6 μL at 100 mM). The conjugation mixture was purified by spin filtration with PBS using a 15 mL Amicon Ultracell 50 kDa MWCO spin filter, sterile-filtered, analysed and stored at +4° C.


The reduction and reoxidation steps are monitored by comparison of the relative amounts of individual light and heavy chains with full length antibody as observed by UHPLC analysis on a Shimadzu Prominence system using a PhenomenexAeris 3.6u XB-C18 150×2.1 mm column eluting with a gradient of water and acetonitrile. UHPLC analysis on a Shimadzu Prominence system using a Phenomenex Aeris 3.6u XB-C18 150×2.1 mm column eluting with a gradient of water and acetonitrile on a reduced sample of Conjugate R347-40 at 280 nm and 330 nm (Conjugate 40 specific) shows unconjugated light chains and a mixture of unconjugated heavy chains and heavy chains attached to a single molecule of Compound 40, consistent with a drug-per-antibody ratio (DAR) of 1.86 molecules of Compound 40 per antibody.


UHPLC analysis on a Shimadzu Prominence system using a Tosoh Bioscience TSKgel G3000SWXL 5 μm 7.8×300 mm column (with a 7 μm 6.0×40 mm guard column) eluting with 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 Conjugate R347-40 at 280 nm shows a monomer purity of 97% with no impurity detected. UHPLC SEC analysis gives a concentration of final Conjugate R347-40 at 0.87 mg/mL in 5.5 mL, obtained mass of Conjugate R347-40 is 4.8 mg (80% yield).


Conjugate HLL2-23


A 50 mM solution of DTT (Dithiothreitol) in phosphate-buffered saline pH 7.4 (PBS) was added (40 molar equivalent/antibody, 40 micromoles, 825 μL) to a 37.5 mL solution of antibody HLL2 (150 mg, 1 micromol) in reduction buffer containing PBS and 1 mM ethylenediaminetetraacetic acid (EDTA) and a final antibody concentration of 4 mg/mL. The reduction mixture was incubated at room temperature overnight with gentle (135 rpm) shaking. The reduced antibody was buffer-exchanged against PBS+1 mM EDTA (to remove the excess of DTT) using TFF (Tangential Flow Filtration, Spectrum Labs 115 cm2 hollow fibre cassette with 50 kDa molecular weight cut off). The sample was filtered using a 0.4 μm syringe filter to remove any debris from the TFF step and antibody concentration brought to 1.5 mg/mL before reoxidation. A 50 mM solution of dehydroascorbic acid (DHAA, 15 molar equivalent/antibody, 13.9 micromoles, 0.28 mL) in DMSO was added and the reoxidation mixture was allowed to react for 16 hours at room temperature under gentle (<150 rpm) shaking. The reoxidation mixture was then sterile-filtered; 139 mg of antibody (92.6 mL as 1.5 mg/mL solution) was obtained, 14 mL of which was taken forward for conjugation with Compound 23 (estimated ca. 21 mg antibody, 0.14 micromoles). Compound 23 was added as a DMSO solution (10 molar equivalent/antibody, 0.33 micromoles in 0.133 mL DMSO) to 14 mL of the reoxidised antibody solution. The conjugation mixture was topped with 1.27 ml of DMSO to bring the final DMSO concentration to 10% (v/v) and incubated for 3 hours at room temperature under gentle agitation (135 rpm). Free drug was then removed from the antibody-drug conjugate by extensive diafiltration in PBS using a spin filter device (Amicon Ultra-30K centrifugal filter, Millipore). The resulting conjugation mixture was sterile-filtered and analysed by UHPLC.


UHPLC analysis on a Shimadzu Prominence system using a Phenomenex Aeris 3.6u XB-C18 150×2.1 mm column eluting with a gradient of water and acetonitrile on a reduced sample of Conjugate at 214 nm (ADC) and 330 nm (Compound 23 specific) shows a mixture of heavy chains either unconjugated or attached to 1 molecule of Compound 23, consistent with a drug-per-antibody ratio (DAR) of 1.64 molecules of Compound 23 per antibody.


UHPLC analysis on a Shimadzu Prominence system using a Tosoh Bioscience TSKgel SuperSW mAb HTP 4 μm 4.6×150 mm column (with a 4 μm 3.0×20 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 greater than 97%. UHPLC SEC analysis gives a concentration of final ADC at 2.06 mg/mL in 10 mL, obtained mass of ADC is 20.6 mg.


Conjugate AntiCD79b-23


A 50 mM solution of DL-dithiothreitol (DTT) in phosphate-buffered saline pH 7.4 (PBS) was added (80 molar equivalent/antibody, 53.3 micromoles, 1.07 mL) to a 25 mL solution of antibody CD79b (100 mg, 667 nmol) in reduction buffer containing PBS and 1 mM ethylenediaminetetraacetic acid (EDTA) and a final antibody concentration of 4 mg/mL.


The reduction mixture was allowed to react at room temperature overnight with gentle 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, 9.28 micromoles, 185 μL) in DMSO was added and the reoxidation mixture was allowed to react for 16 hours at room temperature under gentle (<150 rpm) shaking. The reoxidation mixture was then sterile-filtered; Compound 23 was added as a DMSO solution (15 molar equivalent/antibody, 1.8 micromoles in 1.0 mL DMSO) to 9 mL of this reoxidised antibody solution (18 mg, 120 nanomoles) for a 10% (v/v) final DMSO concentration and a final antibody concentration of 1.8 mg/mL in PBS+1 mM EDTA. The solution was mixed for 2 hours at room temperature under gentle agitation (135 rpm), then the conjugation was quenched by addition of N-acetyl cysteine (7.2 micromoles, 72 μL 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 Phenomenex Aeris 3.6u XB-C18 150×2.1 mm column eluting with a gradient of water and acetonitrile on a reduced sample of Conjugate at 280 nm (ADC) and 330 nm (Compound 23 specific) unconjugated light chains and a mixture of unconjugated heavy chains and heavy chains attached to 1 or 2 molecules of Compound 23, consistent with a drug-per-antibody ratio (DAR) of 2.08 molecules of Compound 23 per antibody.


UHPLC analysis on a Shimadzu Prominence system using a Tosoh Bioscience TSKgel SuperSW mAb HTP 4 μm 4.6×150 mm column (with a 4 μm 3.0×20 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 greater than 98%. UHPLC SEC analysis gives a concentration of final ADC at 1.62 mg/mL in 7.6 mL, obtained mass of ADC is 12.28 mg.


Example 5—In Vitro Testing

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 37° C. 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 of trypan blue cell stained cells, using a haemocytometer. Cells were diluted to 2×105/ml, dispensed (50 μl/well) into 96 well flat bottom plates and incubated overnight before use.


A stock solution (1 ml) of antibody drug conjugate (ADC) (20 μg/ml) was made by dilution of filter-sterilised ADC into cell culture medium. A set of 8×10-fold dilutions of stock ADC were made in a 24 well plate by serial transfer of 100 μl onto 900 μl of cell culture medium.


ADC dilution was dispensed (50 μl/well) into 4 replicate wells of the 96-well plate, containing 50 μl cell suspension seeded the previous day. Control wells received 50 μl cell culture medium.


The 96-well plate containing cells and ADCs was incubated at 37° C. in a CO2-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 μl per well) into each well and incubated for 4 hours at 37° C. in the CO2-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%). IC50 was determined from the doses-response data using GraphPad Prism using the non-linear curve fit algorithm: sigmoidal, 4PL X is log(concentration).




















Cell

ADC
Cell growth



Line
Description
Exposure
medium







SKBR3
Breast
4 days
McCoys with




carcinoma

Glutamax,






10% FBS



BT474
Breast
5 days
DMEM with




carcinoma

glutamax,






10% FBS



NCIN87
Gastric
7 days
RPMI 1640 with




carcinoma

glutamax,






10% FBS













IC50 (μg/ml) in:













ADC
DAR
BT474
NCI-N87
SKBR3







trastuzumab-40
1.68
>1
0.019
0.019



trastuzumab-23
1.71
>1
0.030
0.022










Testing of AntiCD79b-23


The concentration and viability of cells from a sub-confluent (80-90% confluency) T75 flask are measured by trypan blue staining, and counted using the LUNA-II™ Automated Cell Counter. Cells were diluted to 2×105/ml, dispensed (50 μl/well) into 96-well flat-bottom plates.


A stock solution (1 ml) of antibody drug conjugate (ADC) (20 μg/ml) was made by dilution of filter-sterilised ADC into cell culture medium. A set of 8×10-fold dilutions of stock ADC were made in a 24-well plate by serial transfer of 100 μl into 900 μl of cell culture medium. ADC dilution was dispensed (50 μl/well) into 4 replicate wells of the 96-well plate, containing 50 μl cell suspension seeded the previously. Control wells received 50 μl cell culture medium. The 96-well plate containing cells and ADCs was incubated at 37° C. in a CO2-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 μl per well) into each well and incubated for 4 hours at 37° C. in the CO2-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%). IC50 was determined from the dose-response data using GraphPad Prism using the non-linear curve fit algorithm: sigmoidal dose-response curve with variable slope.


ADC incubation times were 4 days with WSUDLCL2 (B-cell non-Hodgkin lymphoma) and SUDHL4 (B-lymphocyte), 5 days for Granta519 (B-cell non-Hodgkin lymphoma) and 6 days for BJAB (Burkitt lymphoma). WSUDLCL2 and SUDHL4 were cultured in RPMI 1640 with Glutamax+10% (v/v) HyClone™ Fetal Bovine Serum, Granta519 in DMEM+Glutamax with 10% (v/v) HyClone™ Fetal Bovine Serum and BJAB in RPMI 1640+Glutamax with 20% (v/v) HyClone™ Fetal Bovine Serum.















EC50 (μg/ml) in:











ADC
SUDHL4
WSUDLCL2
GRANTA519
BJAB





AntiCD79b-23
0.05387
0.9268
0.04957
0.003158









Example 6

Mice


Female severe combined immune-deficient mice (Fox Chase SCID®, C.B-17/Icr-Prkdcscid, Charles River) were ten weeks old with a body weight (BW) range of 16.2 to 21.9 grams 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.


In Vivo Implantation and Tumour Growth


Xenografts were initiated with BT474 human breast carcinomas maintained at CR Discovery Services by serial subcutaneous transplantation into the SCID mice (see above). On the day of tumour implant, each test mouse received a 1-mm3 BT474 fragment implanted subcutaneously in the right flank, and tumour growth was monitored as the average size approached the target range of 100 to 150 mm3. Thirty-three days after tumour implantation, designated as Day 1 of the study, the animals were sorted into nine groups each consisting of ten mice with individual tumour volumes of 75 to 144 mm3 and group mean tumour volumes of 111 to 112 mm3. Tumours were measured in two dimensions using calipers, and volume was calculated using the formula:

Tumour Volume (mm3)=0.5(w2×l)

where w=width and l=length, in mm, of the tumour. Tumour weight may be estimated with the assumption that 1 mg is equivalent to 1 mm3 of tumour volume.


Treatment 1


Treatment began on Day 1 in groups of mice (n=10) with established subcutaneous BT474 tumors (75-196 mm3). Trastuzumab-23 was administered intravenously once on Day 1 (qd×1) at two dosages (0.3 and 1 mg/kg). A vehicle-treated group served as the control group for efficacy analysis. Tumors were measured twice per week until the study was ended on Day 60. Each mouse was euthanized when its tumor reached the endpoint volume of 800 mm3 or on the final day, whichever came first. The time to endpoint (TTE) was calculated for each mouse.


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. Mice were monitored for complete regression (CR) and partial regression (PR) responses.


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 44.4 days, establishing a maximum possible TGD of 15.6 days (35%) for the 60-day study. ADC regimens resulted in the maximum possible TGD, produced survival benefit that was statistically significantly different from vehicle-treated controls (P<0.01) and could not be distinguished based on logrank analysis (P>0.05). Differences within ADC treatments were only evident in the MTV on the final day and numbers and types of regression responses produced by each regimen.


Trastuzumab-23 at 1 mg/kg produced four partial regressions (PRs). The results are illustrated in FIG. 1.


Treatment 2


Treatment began on Day 1 in groups of mice (n=9 or 10) with established subcutaneous BT474 tumors (108-196 mm3). Trastuzumab-40 was administered intravenously once on Day 1 (qd×1) at two dosages (0.3 and 1 mg/kg). A vehicle-treated group served as the control group for efficacy analysis. Tumors were measured twice per week until the study was ended on Day 62. Each mouse was euthanized when its tumor reached the endpoint volume of 1000 mm3 or on the final day, whichever came first. The time to endpoint (TTE) was calculated for each mouse.


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. Mice were monitored for complete regression (CR) and partial regression (PR) responses.


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 52.9 days, establishing a maximum possible TGD of 9.1 days (17%) for the 62-day study. ADC treatment resulted in the maximum possible TGD, however only the 1 mg/kg treatment produced survival benefit that was statistically significantly different from vehicle-treated controls (P<0.001).


Trastuzumab-40, at 1 mg/kg produced four partial regressions (PRs) and one complete regression (CR) which remained a tumour-free survivor (TFS) at study end. The results are shown in FIG. 2.


Example 7

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 μg/mL streptomycin sulfate and 25 μg/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 (SCID mice as in Example 6) was injected subcutaneously in the right flank with 1×107 cells (0.1 mL cell suspension), and tumor growth was monitored as the average size approached the target range of 100 to 150 mm3. Eleven days later, designated as Day 1 of the study, mice were sorted according to calculated tumor size into eleven groups each consisting of ten animals with individual tumor volumes ranging from 88 to 144 mm3 and group mean tumor volumes of 119-121 mm3. Tumors were measured in two dimensions using calipers, and volume was calculated using the formula:

Tumour Volume (mm3)=0.5(w2×l)

where w=width and l=length, in mm, of the tumour. Tumour weight may be estimated with the assumption that 1 mg is equivalent to 1 mm3 of tumour volume


Treatment 1


Treatment began on Day 1 in groups of mice (n=10) with established subcutaneous NCI-N87 tumors (88-144 mm3). Trastuzumab-23 was administered intravenously once on Day 1 (qd×1) at two dosages (0.3 and 1 mg/kg). A vehicle-treated group served as the control group for efficacy analysis. Tumors were measured twice per week until the study was ended on Day 81. Each mouse was euthanized when its tumor reached the endpoint volume of 800 mm3 or on the final day, whichever came first. The time to endpoint (TTE) was calculated for each mouse.


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. Mice were monitored for complete regression (CR) and partial regression (PR) responses.


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 53.4 days, establishing a maximum possible TGD of 27.6 days (52%) for the 81-day study. Trastuzumab-23 tested at 1 mg/kg produced survival benefit that was statistically significantly different from vehicle-treated controls (P<0.001) and resulted in the maximum possible TGD. At 0.3 mg/kg, the median TTE was 80.5 days, which corresponds to TGD of 27.1 days (51%).


Trastuzumab-23, at 1 mg/kg produced one partial regressions (PR). The results are illustrated in FIG. 3.


Treatment 2


Treatment began on Day 1 in groups of mice (n=10) with established subcutaneous NCI-N87 tumors (75-126 mm3). Trastuzumab-40 was administered intravenously once on Day 1 (qd×1) at two dosages (0.3 and 1 mg/kg). A vehicle-treated group served as the control group for efficacy analysis. Tumors were measured twice per week until the study was ended on Day 83. Each mouse was euthanized when its tumor reached the endpoint volume of 800 mm3 or on the final day, whichever came first. The time to endpoint (TTE) was calculated for each mouse.


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. Mice were monitored for complete regression (CR) and partial regression (PR) responses. 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 44.9 days, establishing a maximum possible TGD of 38.1 days (85%) for the 83-day study. Trastuzumab-40 (0.3 mg/kg) had a median TTE of 54.2 days corresponding to a TGD of 9.3 days (21%). Trastuzumab-40 (1 mg/kg) had a median TTE of 61.6 days corresponding to a TGD of 16.7 days (37%).


The results are illustrated in FIG. 4.


Example 8—Toxicity Studies/Therapeutic Index

Rat Study:


A single dose toxicity study was used to determine the maximum tolerated dose (MTD) and safety profile of Trastuzumab-23. Male Sprague Dawley rats (Harlan, Inc) were dosed once by slow bolus intravenous injection via the tail vein with vehicle control (25 mM Histidine-HCl, 7% sucrose, 0.02% Polysorbate 80, pH 6.0) or test material (Trastuzumab-23).


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.






















Male Rats




Dose
Dose

Main Study


Group
Treatment
Route
(mg/kg)
Frequency
N




















1
Control
IV
0
Single
5


7
Trastuzumab-23
IV
3
Single
5


8
Trastuzumab-23
IV
4
Single
5


9
Trastuzumab-23
IV
5
Single
5


10
Trastuzumab-23
IV
6
Single
5


13
Trastuzumab-23
IV
7
Single
5





Control = 25 mM Histidine-HCl, 7% sucrose, 0.02% Polysorbate 80, pH 6.0






Tolerability was determined based on toxicity end points, including body weight loss (>10%) and bone marrow suppression. Based on minimal adverse findings at the high dose, the maximum tolerated dose (MTD) in the rat after a single dose of Trastuzumab-23 was determined to be >7 mg/kg, which was the highest dose level evaluated.


Therapeutic Index


The Therapeutic Index can be calculated by dividing the maximum tolerated single dose (MTD) of non-targeted ADC in rat, by the minimal effective single dose (MED) of the a targeted ADC. The MED is the single dose necessary to achieve tumour stasis in an in vivo model at 28 days (for NCI-N87 xenograft).


Thus for conjugates of compound 23, the therapeutic index is the MTD of greater than 7 mg/kg divided by the MED which is less than 1 mg/kg (see FIG. 3 at 28 days), giving a Therapeutic Index of greater than 7.


Cynomolgus Macaque Study:


A Single-Dose Toxicity study was performed in male Cynomolgus macaques monkeys (Macaca fascicularis) of Cambodian origin following a single intravenous (IV) bolus injection of ADC-SG3400. The study was conducted in 2 phases. In phase 1, animals (n=1) were treated at dose levels of 1, 3, or 6 mg/kg to determine the optimal dose level to explore in phase 2. Animals were dosed by slow bolus intravenous injection via the saphenous vein with vehicle control (25 mM Histidine, 200 mM Sucrose, pH 6.0) or test material (Trastuzumab-23). Based on observations of significant body weight loss at 6 mg/kg, a dose level of 4.5 mg/kg was chosen for phase 2 of the study. In phase 2, animals (n=3) were administered a single dose of 4.5 mg/kg Trastuzumab-23 on Day 1 and necropsied on Days 71 or 72.






















Male Cyno




Dose
Dose

Main Study


Group
Treatment
Route
(mg/kg)
Frequency
N




















1
Trastuzumab-23
IV
1
Single
1


2
Trastuzumab-23
IV
3
Single
1


3
Trastuzumab-23
IV
6
Single
1


4
Trastuzumab-23
IV
4.5
Single
3









Tolerability was determined based on toxicity end points, including body weight loss (>10%) and bone marrow suppression. There was no unscheduled mortality in any animal administered Trastuzumab-23. The major findings were body weight loss and bone marrow suppression at the highest tested dose of 6 mg/kg. Based on these data, and minimal signs of toxicity at next lowest dose, the MTD of Trastuzumab-23 in cynos was 4.5 mg/kg.


All documents and other references mentioned above are herein incorporated by reference.

Claims
  • 1. A conjugate of formula I: L-(DL)P  (I)wherein L is a Ligand unit, DL is a Drug Linker unit of formula II:
  • 2. A conjugate according to claim 1, wherein DL is DL-A:
  • 3. A conjugate according to claim 1, wherein DL is DL-B:
  • 4. The conjugate according to claim 1, wherein the antibody or antibody fragment is an antibody or antibody fragment for a tumour-associated antigen.
  • 5. The conjugate according to claim 1, 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) BMPR1B;(2) E16;(3) STEAP1;(4) 0772P;(5) MPF;(6) Napi3b;(7) Sema 5b;(8) PSCA h1g;(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) EGFRvIII;(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-MST1R;(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.
  • 6. The conjugate according to claim 1, wherein the antibody or antibody fragment is a cysteine-engineered antibody.
  • 7. The conjugate according to claim 1 wherein p is an integer from 1 to 8.
  • 8. The conjugate according to claim 7, wherein p is 1, 2, 3, or 4.
  • 9. A composition comprising a mixture of conjugates according to claim 1, wherein the average p in the mixture of conjugate compounds is about 1 to about 8.
  • 10. A pharmaceutical composition comprising the conjugate of claim 1, a pharmaceutically acceptable diluent, carrier or excipient.
  • 11. A method of medical treatment comprising administering to a patient a therapeutically effective amount of the pharmaceutical composition of claim 10, wherein the method of medical treatment is for treating cancer, wherein the cancer comprises lymphoma, breast cancer, or gastric cancer.
  • 12. The method of claim 11, wherein the patient is administered a chemotherapeutic agent, in combination with the conjugate.
  • 13. A method of treating a mammal having lymphoma, breast cancer, or gastric cancer, comprising administering an effective amount of a conjugate according to claim 1 or a pharmaceutical composition according to claim 10.
Priority Claims (1)
Number Date Country Kind
1602356 Feb 2016 GB national
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2017/052988 2/10/2017 WO
Publishing Document Publishing Date Country Kind
WO2017/137553 8/17/2017 WO A
US Referenced Citations (247)
Number Name Date Kind
3361742 Julius et al. Jan 1968 A
3523941 Leimgruber et al. Aug 1970 A
3524849 Batcho et al. Aug 1970 A
3794644 Karlyone et al. Feb 1974 A
4185016 Takanabe et al. Jan 1980 A
4239683 Takanabe et al. Dec 1980 A
4309437 Ueda et al. Jan 1982 A
4353827 Hunkeler et al. Oct 1982 A
4382032 Hunkeler et al. May 1983 A
4386028 Hunkeler et al. May 1983 A
4405516 Hunkeler et al. Sep 1983 A
4405517 Hunkeler et al. Sep 1983 A
4407752 Hunkeler et al. Oct 1983 A
4427587 Kaneko et al. Jan 1984 A
4427588 Kaneko et al. Jan 1984 A
4701325 Ueda et al. Oct 1987 A
4816567 Cabilly et al. Mar 1989 A
4923984 Matsumura et al. May 1990 A
5362852 Geoghegan Nov 1994 A
5418241 Jegham et al. May 1995 A
5440021 Chuntharapai et al. Aug 1995 A
5583024 McElroy et al. Dec 1996 A
5621002 Bosslet et al. Apr 1997 A
5644033 Seon Jul 1997 A
5674713 McElroy et al. Oct 1997 A
5700670 Yamagishi et al. Dec 1997 A
5773223 Shyamala et al. Jun 1998 A
5792616 Persico et al. Aug 1998 A
5854399 Salomon et al. Dec 1998 A
5869445 Cheever et al. Feb 1999 A
5976551 Mottez et al. Nov 1999 A
6011146 Mottez et al. Jan 2000 A
6153408 Abastado et al. Nov 2000 A
6214345 Firestone et al. Apr 2001 B1
6218519 Kenten et al. Apr 2001 B1
6268488 Barbas, III et al. Jul 2001 B1
6362331 Kamal et al. Mar 2002 B1
6518404 Li et al. Feb 2003 B1
6534482 Fikes et al. Mar 2003 B1
6555339 Liaw et al. Apr 2003 B1
6562806 Thurston et al. May 2003 B1
6602677 Wood et al. Aug 2003 B1
6608192 Thurston et al. Aug 2003 B1
6660742 Lee Dec 2003 B2
6660856 Wang Dec 2003 B2
6677435 Barbas, III et al. Jan 2004 B2
6747144 Thurston et al. Jun 2004 B1
6759509 King et al. Jul 2004 B1
6835807 Sasaki et al. Dec 2004 B1
6884799 Kamal et al. Apr 2005 B2
6909006 Thurston et al. Jun 2005 B1
7049311 Thurston et al. May 2006 B1
7067511 Thurston et al. Jun 2006 B2
7223837 De Groot et al. May 2007 B2
7244724 Liu et al. Jul 2007 B2
7265105 Thurston et al. Sep 2007 B2
7375078 Feng May 2008 B2
7407951 Thurston et al. Aug 2008 B2
7429658 Howard et al. Sep 2008 B2
7511032 Liu et al. Mar 2009 B2
7521541 Eigenbrot et al. Apr 2009 B2
7528126 Howard et al. May 2009 B2
7557099 Howard et al. Jul 2009 B2
7612062 Gregson et al. Nov 2009 B2
7704924 Thurston et al. Apr 2010 B2
7723485 Junutula et al. May 2010 B2
7741319 Howard et al. Jun 2010 B2
8034808 Delavault et al. Nov 2011 B2
8163736 Gauzy et al. Apr 2012 B2
8321774 Barthel et al. Nov 2012 B2
8487092 Howard et al. Jul 2013 B2
8501934 Howard et al. Aug 2013 B2
8592576 Howard et al. Nov 2013 B2
8633185 Howard et al. Jan 2014 B2
8637664 Howard et al. Jan 2014 B2
8697688 Howard et al. Apr 2014 B2
8829184 Howard et al. Sep 2014 B2
8940733 Howard et al. Jan 2015 B2
9102704 Howard Aug 2015 B2
9242013 Howard et al. Jan 2016 B2
9321774 Howard et al. Apr 2016 B2
9376440 Howard et al. Jun 2016 B2
9387259 Jeffrey et al. Jul 2016 B2
9388187 Howard et al. Jul 2016 B2
9399073 Howard et al. Jul 2016 B2
9399641 Howard et al. Jul 2016 B2
9415117 Howard Aug 2016 B2
9464141 Asundi et al. Oct 2016 B2
9526798 Jeffrey et al. Dec 2016 B2
9562049 Howard Feb 2017 B2
9592240 Howard et al. Mar 2017 B2
9624227 Howard et al. Apr 2017 B2
9649390 Howard et al. May 2017 B2
9707301 Jeffrey et al. Jul 2017 B2
9713647 Jeffrey et al. Jul 2017 B2
9732084 Howard et al. Aug 2017 B2
9745303 Howard et al. Aug 2017 B2
9889207 Howard Feb 2018 B2
9956298 Howard et al. May 2018 B2
20010055751 Reiter et al. Dec 2001 A1
20020034749 Billing-Medel et al. Mar 2002 A1
20020042366 Thompson et al. Apr 2002 A1
20020150573 Nussenzweig Oct 2002 A1
20020193567 Jacobs et al. Dec 2002 A1
20030060612 Goddard et al. Mar 2003 A1
20030062401 Hasz et al. Apr 2003 A1
20030064397 Spancake et al. Apr 2003 A1
20030065143 Eaton et al. Apr 2003 A1
20030091580 Mitcham et al. May 2003 A1
20030096743 Senter et al. May 2003 A1
20030096961 Baker et al. May 2003 A1
20030105292 Liaw et al. Jun 2003 A1
20030109676 Li et al. Jun 2003 A1
20030118592 Ledbetter et al. Jun 2003 A1
20030119121 Baker et al. Jun 2003 A1
20030119122 Baker et al. Jun 2003 A1
20030119125 Baker et al. Jun 2003 A1
20030119126 Baker et al. Jun 2003 A1
20030119128 Baker et al. Jun 2003 A1
20030119129 Baker et al. Jun 2003 A1
20030119130 Baker et al. Jun 2003 A1
20030119131 Baker et al. Jun 2003 A1
20030124140 Bangur et al. Jul 2003 A1
20030124579 Mack et al. Jul 2003 A1
20030129192 Chenault et al. Jul 2003 A1
20030130189 Senter et al. Jul 2003 A1
20030134790 Langenfeld Jul 2003 A1
20030143557 Penner Jul 2003 A1
20030157089 Xu et al. Aug 2003 A1
20030165504 Retter et al. Sep 2003 A1
20030185830 Xu et al. Oct 2003 A1
20030186372 Baker et al. Oct 2003 A1
20030186373 Baker et al. Oct 2003 A1
20030194704 Penn et al. Oct 2003 A1
20030195196 Thurston et al. Oct 2003 A1
20030206918 Fanger et al. Nov 2003 A1
20030219806 Glucksmann et al. Nov 2003 A1
20030224411 Stanton et al. Dec 2003 A1
20030224454 Ryseck et al. Dec 2003 A1
20030228319 Frantz et al. Dec 2003 A1
20030232056 Fanger et al. Dec 2003 A1
20030232350 Afar et al. Dec 2003 A1
20040001827 Dennis Jan 2004 A1
20040005320 Thompson et al. Jan 2004 A1
20040005538 Chen et al. Jan 2004 A1
20040005563 Mack et al. Jan 2004 A1
20040005598 DeVaux et al. Jan 2004 A1
20040018194 Francisco et al. Jan 2004 A1
20040018553 Billing-Medel et al. Jan 2004 A1
20040022727 Stanton et al. Feb 2004 A1
20040044179 Baker et al. Mar 2004 A1
20040044180 Baker et al. Mar 2004 A1
20040052793 Carter et al. Mar 2004 A1
20040101874 Ghosh et al. May 2004 A1
20040101899 Dillon et al. May 2004 A1
20040121940 De Groot et al. Jun 2004 A1
20040138269 Sun et al. Jul 2004 A1
20040197325 Law et al. Oct 2004 A1
20040198722 Thurston et al. Oct 2004 A1
20040249130 Stanton et al. Dec 2004 A1
20050271615 Shabat et al. Dec 2005 A1
20060116422 De Groot et al. Jun 2006 A1
20070072846 Vishnuvajjala et al. Mar 2007 A1
20070154906 Martin et al. Jul 2007 A1
20070185336 Rossen et al. Aug 2007 A1
20070191349 Howard et al. Aug 2007 A1
20070232592 Delavault et al. Oct 2007 A1
20070249591 Howard et al. Oct 2007 A1
20080090812 Pepper et al. Apr 2008 A1
20080092940 Nakajima Apr 2008 A1
20080112961 Stavenhagen et al. May 2008 A1
20080206239 Jones et al. Aug 2008 A1
20080213289 Francisco et al. Sep 2008 A1
20080214525 Howard et al. Sep 2008 A1
20090036431 Gauzy et al. Feb 2009 A1
20090148942 Mcdonagh et al. Jun 2009 A1
20090149449 Liu et al. Jun 2009 A1
20090274713 Chari et al. Nov 2009 A1
20090304710 Park et al. Dec 2009 A1
20100028346 Lutz et al. Feb 2010 A1
20100047257 Blanc et al. Feb 2010 A1
20100113425 Howard et al. May 2010 A1
20100203007 Li et al. Aug 2010 A1
20100316656 Bouchard et al. Dec 2010 A1
20110039969 Muratoglu et al. Feb 2011 A1
20110091372 Ghayur et al. Apr 2011 A1
20110160192 Howard et al. Jun 2011 A1
20110195021 Deckert et al. Aug 2011 A1
20110195022 Deckert et al. Aug 2011 A1
20110196148 Howard et al. Aug 2011 A1
20110201803 Howard et al. Aug 2011 A1
20110256157 Howard et al. Oct 2011 A1
20120233172 Skillcorn et al. Sep 2012 A1
20120238731 Fishkin et al. Sep 2012 A1
20130028917 Howard et al. Jan 2013 A1
20130028919 Howard et al. Jan 2013 A1
20130137659 Commercon et al. May 2013 A1
20130244171 Yamasaki et al. Sep 2013 A1
20130266595 Flygare et al. Oct 2013 A1
20130266596 Li et al. Oct 2013 A1
20130302359 Li et al. Nov 2013 A1
20130304357 Koci et al. Nov 2013 A1
20140030279 Polakis et al. Jan 2014 A1
20140030280 Polakis Jan 2014 A1
20140066435 Howard et al. Mar 2014 A1
20140088089 Chari Mar 2014 A1
20140120118 Howard May 2014 A1
20140127239 Howard May 2014 A1
20140155590 Commercon et al. Jun 2014 A1
20140234346 Howard Aug 2014 A1
20140274907 Howard et al. Sep 2014 A1
20140275522 Howard et al. Sep 2014 A1
20140286970 Jeffrey et al. Sep 2014 A1
20140294868 Howard et al. Oct 2014 A1
20140302066 Jeffrey et al. Oct 2014 A1
20150111880 Howard et al. Apr 2015 A1
20150126495 Howard et al. May 2015 A1
20150133435 Howard et al. May 2015 A1
20150158869 Howard Jun 2015 A1
20150183883 Asundi Jul 2015 A1
20150265722 Van Berkel Sep 2015 A1
20150273077 Van Berkel Oct 2015 A1
20150273078 Van Berkel Oct 2015 A1
20150274737 Howard et al. Oct 2015 A1
20150283258 Van Berkel Oct 2015 A1
20150283262 Van Berkel Oct 2015 A1
20150283263 Van Berkel Oct 2015 A1
20150297746 Van Berkel Oct 2015 A1
20150315196 Howard et al. Nov 2015 A1
20150344482 Howard et al. Dec 2015 A1
20160015828 Torgor Jan 2016 A1
20160031887 Howard et al. Feb 2016 A1
20160075787 Zheng et al. Mar 2016 A1
20160144052 Howard et al. May 2016 A1
20160074527 Flygare et al. Jul 2016 A1
20160250344 Howard et al. Sep 2016 A1
20160250345 Howard et al. Sep 2016 A1
20160250346 Howard et al. Sep 2016 A1
20160256561 Howard et al. Sep 2016 A1
20160263242 Howard et al. Sep 2016 A1
20160310611 Flygare et al. Oct 2016 A1
20170239365 Howard et al. Aug 2017 A1
20170290924 Jeffrey et al. Oct 2017 A1
20170298137 Jeffrey et al. Oct 2017 A1
20170340752 Howard Nov 2017 A1
20180125997 Howard et al. May 2018 A1
20180134717 Howard et al. May 2018 A1
Foreign Referenced Citations (298)
Number Date Country
101171257 Apr 2008 CN
0522868 Jan 1993 EP
0875569 Nov 1998 EP
1295944 Mar 2003 EP
1347046 Sep 2003 EP
1394274 Mar 2004 EP
1439393 Jul 2004 EP
1813614 Aug 2007 EP
2019104 Jan 2009 EP
2298817 Mar 2011 EP
2528625 Jul 2013 EP
2027356 Dec 1969 FR
2586683 Mar 1987 FR
1299198 Dec 1972 GB
2053894 Feb 1981 GB
5382792 Jul 1978 JP
57131791 Aug 1982 JP
2004113151 Apr 2004 JP
WO 199102536 Mar 1991 WO
WO 199207574 May 1992 WO
WO 199217497 Oct 1992 WO
WO 199219620 Nov 1992 WO
WO 199318045 Sep 1993 WO
WO 199410312 May 1994 WO
WO 199428931 Dec 1994 WO
WO 199504718 Feb 1995 WO
WO 199630514 Oct 1996 WO
WO 199707198 Feb 1997 WO
WO 199744452 Nov 1997 WO
WO 199813059 Apr 1998 WO
WO 199837193 Aug 1998 WO
WO 199840403 Sep 1998 WO
WO 199851805 Nov 1998 WO
WO 199851824 Nov 1998 WO
WO 199928468 Jun 1999 WO
WO 199946284 Sep 1999 WO
WO 199958658 Nov 1999 WO
WO 200003291 Jan 2000 WO
WO 200012506 Mar 2000 WO
WO 200012507 Mar 2000 WO
WO 200012508 Mar 2000 WO
WO 200012509 Mar 2000 WO
WO 200014228 Mar 2000 WO
WO 200020579 Apr 2000 WO
WO 200022129 Apr 2000 WO
WO 200032752 Jun 2000 WO
WO 200036107 Jun 2000 WO
WO 200040614 Jul 2000 WO
WO 200044899 Aug 2000 WO
WO 200012130 Sep 2000 WO
WO 200053216 Sep 2000 WO
WO 200055351 Sep 2000 WO
WO 200075655 Dec 2000 WO
WO 200100244 Jan 2001 WO
WO 200116104 Mar 2001 WO
WO 200116318 Mar 2001 WO
WO 200138490 May 2001 WO
WO 200140269 Jun 2001 WO
WO 200140309 Jun 2001 WO
WO 200141787 Jun 2001 WO
WO 200145746 Jun 2001 WO
WO 200146232 Jun 2001 WO
WO 200146261 Jun 2001 WO
WO 200148204 Jul 2001 WO
WO 200153463 Jul 2001 WO
WO 200157188 Aug 2001 WO
WO 200162794 Aug 2001 WO
WO 200166689 Sep 2001 WO
WO 200172830 Oct 2001 WO
WO 200172962 Oct 2001 WO
WO 200175177 Oct 2001 WO
WO 200177172 Oct 2001 WO
WO 200188133 Nov 2001 WO
WO 200190304 Nov 2001 WO
WO 200194641 Dec 2001 WO
WO 200198351 Dec 2001 WO
WO 200202587 Jan 2002 WO
WO 200202624 Jan 2002 WO
WO 200202634 Jan 2002 WO
WO 200206317 Jan 2002 WO
WO 200206339 Jan 2002 WO
WO 200210187 Feb 2002 WO
WO 200210382 Feb 2002 WO
WO 200212341 Feb 2002 WO
WO 200213847 Feb 2002 WO
WO 200214503 Feb 2002 WO
WO 200216413 Feb 2002 WO
WO 200222153 Mar 2002 WO
WO 200222636 Mar 2002 WO
WO 200222660 Mar 2002 WO
WO 200222808 Mar 2002 WO
WO 200224909 Mar 2002 WO
WO 200226822 Apr 2002 WO
WO 200230268 Apr 2002 WO
WO 200238766 May 2002 WO
WO 200254940 Jul 2002 WO
WO 200259377 Aug 2002 WO
WO 200260317 Aug 2002 WO
WO 200261087 Aug 2002 WO
WO 200264798 Aug 2002 WO
WO 200271928 Sep 2002 WO
WO 200272596 Sep 2002 WO
WO 200278524 Oct 2002 WO
WO 200281646 Oct 2002 WO
WO 200283866 Oct 2002 WO
WO 200286443 Oct 2002 WO
WO 200288170 Nov 2002 WO
WO 200288172 Nov 2002 WO
WO 200289747 Nov 2002 WO
WO 200292836 Nov 2002 WO
WO 200294852 Nov 2002 WO
WO 200298358 Dec 2002 WO
WO 200299074 Dec 2002 WO
WO 200299122 Dec 2002 WO
WO 2002101075 Dec 2002 WO
WO 2002102235 Dec 2002 WO
WO 200216429 Jan 2003 WO
WO 2003000842 Jan 2003 WO
WO 2003002717 Jan 2003 WO
WO 2003003906 Jan 2003 WO
WO 2003003984 Jan 2003 WO
WO 2003004529 Jan 2003 WO
WO 2003004989 Jan 2003 WO
WO 2003008537 Jan 2003 WO
WO 2003009814 Feb 2003 WO
WO 2003014294 Feb 2003 WO
WO 2003016475 Feb 2003 WO
WO 2003016494 Feb 2003 WO
WO 2003018621 Mar 2003 WO
WO 2003022995 Mar 2003 WO
WO 2003023013 Mar 2003 WO
WO 2003024392 Mar 2003 WO
WO 2003025138 Mar 2003 WO
WO 2003025148 Mar 2003 WO
WO 2003025228 Mar 2003 WO
WO 2003026493 Apr 2003 WO
WO 2003026577 Apr 2003 WO
WO 2003029262 Apr 2003 WO
WO 2003029277 Apr 2003 WO
WO 2003029421 Apr 2003 WO
WO 2003034984 May 2003 WO
WO 2003035846 May 2003 WO
WO 2003042661 May 2003 WO
WO 2003043583 May 2003 WO
WO 2003045422 Jun 2003 WO
WO 2003048202 Jun 2003 WO
WO 2003054152 Jul 2003 WO
WO 2003055439 Jul 2003 WO
WO 2003055443 Jul 2003 WO
WO 2003060612 Jul 2003 WO
WO 2003062401 Jul 2003 WO
WO 2003072035 Sep 2003 WO
WO 2003072036 Sep 2003 WO
WO 2003077836 Sep 2003 WO
WO 2003081210 Oct 2003 WO
WO 2003083041 Oct 2003 WO
WO 2003083047 Oct 2003 WO
WO 2003083074 Oct 2003 WO
WO 2003087306 Oct 2003 WO
WO 2003087768 Oct 2003 WO
WO 2003088808 Oct 2003 WO
WO 2003089624 Oct 2003 WO
WO 2003089904 Oct 2003 WO
WO 2003093444 Nov 2003 WO
WO 2003097803 Nov 2003 WO
WO 2003101283 Dec 2003 WO
WO 2003101400 Dec 2003 WO
WO 2003104270 Dec 2003 WO
WO 2003104275 Dec 2003 WO
WO 2003104399 Dec 2003 WO
WO 2003105758 Dec 2003 WO
WO 2004000221 Dec 2003 WO
WO 2004000997 Dec 2003 WO
WO 2004001004 Dec 2003 WO
WO 2004005598 Jan 2004 WO
WO 2004009622 Jan 2004 WO
WO 2004011611 Feb 2004 WO
WO 2004015426 Feb 2004 WO
WO 2004016225 Feb 2004 WO
WO 2004020583 Mar 2004 WO
WO 2004020595 Mar 2004 WO
WO 2004022709 Mar 2004 WO
WO 2004022778 Mar 2004 WO
WO 2004027049 Apr 2004 WO
WO 2004031238 Apr 2004 WO
WO 2004032828 Apr 2004 WO
WO 2004032842 Apr 2004 WO
WO 2004040000 May 2004 WO
WO 2004042346 May 2004 WO
WO 2004043361 May 2004 WO
WO 2004043963 May 2004 WO
WO 2004044178 May 2004 WO
WO 2004045516 Jun 2004 WO
WO 2004045520 Jun 2004 WO
WO 2004045553 Jun 2004 WO
WO 2004046342 Jun 2004 WO
WO 2004047749 Jun 2004 WO
WO 2004048938 Jun 2004 WO
WO 2004053079 Jun 2004 WO
WO 2004058309 Jul 2004 WO
WO 2004063355 Jul 2004 WO
WO 2004063362 Jul 2004 WO
WO 2004063709 Jul 2004 WO
WO 2004065576 Aug 2004 WO
WO 2004065577 Aug 2004 WO
WO 2004074320 Sep 2004 WO
WO 2005023814 Mar 2005 WO
WO 2005040170 May 2005 WO
WO 2005042535 May 2005 WO
WO 2005079479 Sep 2005 WO
WO 2005082023 Sep 2005 WO
WO 2005085177 Sep 2005 WO
WO 2005085250 Sep 2005 WO
WO 2005085251 Sep 2005 WO
WO 2005085259 Sep 2005 WO
WO 2005085260 Sep 2005 WO
WO 2005105113 Nov 2005 WO
WO 2005110423 Nov 2005 WO
WO 2006111759 Oct 2006 WO
WO 2007039752 Apr 2007 WO
WO 2007044515 Apr 2007 WO
WO 2007085930 Aug 2007 WO
WO 2008010101 Jan 2008 WO
WO 2008022152 Feb 2008 WO
WO 2008047242 Apr 2008 WO
WO 2008050140 May 2008 WO
WO 2008070593 Jun 2008 WO
WO 2009016516 Feb 2009 WO
WO 2009052249 Apr 2009 WO
WO 2009060208 May 2009 WO
WO 2009060215 May 2009 WO
WO 2009117531 Sep 2009 WO
WO 2010010347 Jan 2010 WO
WO 2010043877 Apr 2010 WO
WO 2010043880 Apr 2010 WO
WO 2010091150 Aug 2010 WO
WO 2010095031 Aug 2010 WO
WO 2011023883 Mar 2011 WO
WO 2011038159 Mar 2011 WO
WO 2011100227 Aug 2011 WO
WO 2011128650 Oct 2011 WO
WO 2011130598 Oct 2011 WO
WO 2011130613 Oct 2011 WO
WO 2011130615 Oct 2011 WO
WO 2011130616 Oct 2011 WO
WO 2011133039 Oct 2011 WO
WO 2012014147 Feb 2012 WO
WO 2012112687 Aug 2012 WO
WO 2012112708 Aug 2012 WO
WO 2012128868 Sep 2012 WO
WO 2013041606 Mar 2013 WO
WO 2013053871 Apr 2013 WO
WO 2013053872 Apr 2013 WO
WO 2013053873 Apr 2013 WO
WO 2013055987 Apr 2013 WO
WO 2013055990 Apr 2013 WO
WO 2013055993 Apr 2013 WO
WO 2013164592 Nov 2013 WO
WO 2013164593 Nov 2013 WO
WO 2013177481 Nov 2013 WO
WO 2014011518 Jan 2014 WO
WO 2014011519 Jan 2014 WO
WO 2014022679 Feb 2014 WO
WO 2014031566 Feb 2014 WO
WO 2014057072 Apr 2014 WO
WO 2014057073 Apr 2014 WO
WO 2014057074 Apr 2014 WO
WO 2014057113 Apr 2014 WO
WO 2014057114 Apr 2014 WO
WO 2014057115 Apr 2014 WO
WO 2014057117 Apr 2014 WO
WO 2014057118 Apr 2014 WO
WO 2014057119 Apr 2014 WO
WO 2014057120 Apr 2014 WO
WO 2014057122 Apr 2014 WO
WO 2014080251 May 2014 WO
WO 2014096365 Jun 2014 WO
WO 2014096368 Jun 2014 WO
WO 2014130879 Aug 2014 WO
WO 2014140174 Sep 2014 WO
WO 2014140862 Sep 2014 WO
WO 2014159981 Oct 2014 WO
WO 2014174111 Oct 2014 WO
WO 2005052533 Apr 2015 WO
WO 2015052321 Apr 2015 WO
WO 2015052322 Apr 2015 WO
WO 2015052332 Apr 2015 WO
WO 2015052333 Apr 2015 WO
WO 2015052334 Apr 2015 WO
WO 2015052335 Apr 2015 WO
WO 2015052532 Apr 2015 WO
WO 2015052534 Apr 2015 WO
WO 2015052535 Apr 2015 WO
WO 2015095124 Jun 2015 WO
WO 2015159076 Oct 2015 WO
WO 2016037644 Mar 2016 WO
WO 2016040868 Mar 2016 WO
WO 2016044560 Mar 2016 WO
Non-Patent Literature Citations (402)
Entry
Gregson et al.—Linker length modulates DNA crosslinking reactivity and cytotoxic potency of C8/C8′ether-linked C2-exo unsaturated pyrrolo[2, 1-c][1,4]benzodiazepine (PDB) dimers, J. Med. Chem., 47, 1161-1174, 2004. (Year: 2004).
Hartley J.A., The development of pyrrolobenzodiazepines as antitumor agents, Exp. Op. Inv. Drugs, 20, 733-744, 2011. (Year: 2011).
Adair, J.R et al., “Antibody-drug conjugates—a perfect synergy,” Exp. Opin. Biol. Ther. (2012), pp. 1-16.
Adams et al., “Molecular modelling of a sequence-specific DNA-binding agent based on the pyrrolo[2,1-c][1,4]benzodiazepines,” Pharm. Pharmacol. Commun. (1999) 5:555-560.
Aird, R.E. et al., “ABCB1 genetic polymorphism influences the pharmacology of the new pyrrolobenzodiazepine derivative SJG-136,” Pharmacogenomics Journal (2008) 8(4):289-296.
“Alley, M.C. et al., ““SJG-136 (NSC 694501), a novel rationally designed DNA minor groove interstrand cross-linking agent with potent and broad spectrum antitumor activity. Part 2: Efficacy evaluations,”” Cancer Res. (2004) 64:6700-6706”.
Alley, M.C., “Efficacy evaluations of SJG-136 (NSC 694501), a novel pyrrolobenzodiazepine dimer with broad spectrum antitumor activity,” Proceedings of the American Association for Cancer Research Annual Meeting (Mar. 2002) 43:63.
Alley, S. C., “Contribution of Linker Stability to the Activities of Anticancer Immunoconjugates” Bioconjugate Chem 2008, 19, 759-765.
Althius, T. H. and Hess, H. J., “Synthesis and Identification of the Major Metabolites of Prazosin Formed in Dog and Rat,” J. Medicinal Chem. (1977) 20(1):146-148.
Altuvia et al., “Ranking potential binding peptides to MHC molecules by a computational threading approach.” J. Mol. Biol., 249, 244-250 (1995).
Amiel J., et al., “Heterozygous endothelin receptor B {EDNRB) mutations in isolated Hirschsprung disease,” Hum. Mol. Genet. 5, 355-357, 1996.
Amir et al., “Self-Immolative Dendrimers,” (2003) Angew. Chem. Int. Ed. 42:4494-4499.
Amsberry, et al., “The Lactonization of 2′-Hydroxyhydrocinnamic Acid Amides: A Potential Prodrug for Amines,” (1990) J. Org. Chem. 55:5867-5877.
Antonow, D. et al., ““Synthesis of DNA-Interactive Pyrrolo [2,1-c][1,4] benzodiazepines (PBDs)”” Chemical Reviews, 2011, 111(4):2815-2864.
Antonow, D. et al., “Structure-activity relationships of monomeric C2-aryl pyrrolo[2,1-c][1,4]benzodiazepine (PBD) antitumor agents.”J Med Chem. Apr. 8, 2010;53(7):2927-41.
Antonow, D. et al..“Parallel synthesis of a novel C2-aryl pyrrolo[2,1-c][1,4]benzodiazepine (PBD) library,” J. Comb. Chem. (2007) 9:437-445.
Arai H., et al., ““Molecular cloning of human endothelin receptors and their expressiOn in vascular endothelial cells and smooth muscle cells,”” Jpn. Circ. J. 56, 1303-1307, 1992.
Arai H., et al., “The Human Endotbelin-B Receptor Gene. Structural Organization and Chromosomal Assignment,” J. Biol. Chem. 268, 3463-3470, 1993.
Arima et al., “Studies on Tomaymycin, a New Antibiotic. I. Isolation and Properties of Tomaymycin,” J. Antibiotics (1972) 25:437-444.
Arnould, S., ““Impact on the cell cycle and involvement of ATM, ATR, chk1 and chk2 kinases in the cytotoxicity of SJG-136, a new pyrrolobenzodiazepine dimer,”” Proceedings of the American Association for Cancer Research Annual Meeting (Mar. 2004) 45:1298, Abstract No. 5618.
Arnould, S., “Time-dependent cytotoxicity induced by SJG-136 (NSC 694501): influence of the rate of interstrand cross-link formation on DNA damage signaling,” Mol. Canc. Therap. 5(6):1602-1609 (2006).
Attie T., et al., “Mutation of the endothelin-receptor B gene in Waardenburg-Hirschsprung disease,” Hum. Mol. Genet. 4, 2407-2409, 1995.
Auricchio A., et al., “Endothelin-B receptor mutations in patients with isolated Hirschsprung disease from a non-inbred population,” Hum. Mol. Genet. 5:351-354, 1996.
Axup et al., “Synthesis of site-specific antibody-drug conjugates using unnatural amino acids.” Proc Natl Acad Sci USA. Oct. 2, 2012; 109(40):16101-6.
Bahrenberg et al., ““Reduced Expression of PSCA, a Member of the LY-6 Family of Cell Surface Antigens, in Bladder, Esophagus, and Stomach Tumors,”” Biochem. Biophys. Res. Commun. (2000) 275(3):783-788.
Banker, G.S. et al., “Modern Pharmaceutics”, Third edition, Marcel Dekker, New York (1996) 451 and 596.
Barel M., et al., “Evidence for a new transcript of the Epstein-Barr virus/C3d receptor (CR2, CD21) which is due to alternative exon usage,” Mol. Immunol. 35, 1025-1031, 1998.
Barella et al., “Sequence variation of a novel heptahelical leucocyte receptor through alternative transcript formation,” (1995) Biochem. J. 309:773-779.
Barnett T., et al., “Carcinoembryonic Antigen Family: Characterization of cDNAs Coding for NCA and CEA and Suggestion of Nonrandom Sequence Variation in Their Conserved Loop-Domains,” Genomics 3, 59-66, 1988.
Batisse, et al., “A new delivery system for Auristatin in STxB-drug conjugate therapy.” European J. Medicinal Chemistry, 2015, 95: 483-491.
Beck et al., “DNA Sequence Analysis of 66 kb of the Human MHC Class II Region Encoding a Cluster of Genes for Antigen Processing,” (1992) J. Mol. Biol. 228:433-441.
Beck et al., “Evolutionary Dynamics of Non-coding Sequences Within the Class II Region of the Human MHC,” (1996) J. Mol. Biol. 25 255:1-13.
Berge et al., “Pharmaceutical Salts,” J. Pharm. Sci. (1977) 66:1-19.
Berry, J. M. et al., “Solid-phase synthesis of DNA-interactive pyrrolo[2,1-c][1,4]benzodiazepines,” Tetrahedron Letters (2000) 41:6171-6174.
Blanc et al., “SAR3419: an anti-CD19-Maytansinoid Immunoconjugate for the treatment of B-cell malignancies,” Clin Cancer Res., 2011, 17(20):6448-58.
Blumberg H., et al., ““Interleukin 20: Discovery, Receptor Identification, and Role in Epidermal Function,”” Cell 104, 9-19, 2001.
Bose et al., “New Approaches to Pyrrolo[2,1-c][1,4]benzodiazepines: Synthesis, DNA-binding and cytotoxicity of DC-81,” Tetrahedron, 48, 751-758 (1992).
Bose, D.S. et al., “Effect of linker length on DNA-binding affinity, cross-linking efficiency and cytotoxicity of C8 linked pyrrolobenzodiazepine dimers,” J. Chem. Soc. Chem. Commun. (1992) 20:1518-1520.
Bose, D.S et al., “Rational Design of a Highly Efficient Irreversible DNA Interstrand Cross-Linking Agent Based on the Pyrrolobenzodiazepine Ring System,” J. Am. Chem. Soc., 114, 4939-4941 (1992).
Bourgeois C., et al., “Endothelin-1 and ETA Receptor Expression in Vascular Smooth Muscle Cells from Human Placenta: A New ETA Receptor Messenger Ribonucleic Acid Is Generated by Alternative Splicing of Exon 3,” J. Clin. Endocrinol. Metab. 82,3116-3123, 1997.
Brand et al., Prospect for anti-HER2 receptor therapy in breast cancer. Anticancer Res. Jan.-Feb. 2006;26(1B):463-70.
Brinster et al., ““Introits increase transcriptional efficiency in transgenic mice,”” (1988) Proc. Natl. Acad. Sci. USA 85:836-840.
Buchman et al., “Comparison of Intron-Dependent and Intron-Independent Gene Expression,” (1988) Mol. Cell. Biol. 8:4395-4405.
Buhrow, S.A., “LC-MS/MS assay and dog pharmacokinetics of the dimeric pyrrolobenzodiazepine SJG-136 (NSC 694501),” J. Chromat. B: Anal. Tech. Biomed. Life Sci. (2006) 840(1):56-62.
Burke, P.J. et al., “Anti-CD70 antibody-drug conjugates containing pyrrolobenzodiazepine dimers demonstrate robust antitumor activity,” AACR National Meeting, Apr. 2012, Chicago, Illinois, Abstract No. 4631.
Burke, P.J. et al., “Novel immunoconjugates comprised of streptonigrin and 17-amino-geldanamycin attached via a dipeptide-p-aminobenzyl-amine linker system,” Bioorg. Med. Chem. Lett., Apr. 2009, 19(10):2650-2653.
Calcutt, M.W., ““Determination of chemically reduced pyrrolobenzodiazepine SJG-136 in human plasma by HPLC-MS/MS: application to an anticancer phase I dose escalation study,”” J. Mass Spectrom. (2008) 43(1):42-52.
Carl et al., “A Novel Connector Linkage Applicable in Prodrug Design,” (1981) J. Med. Chem. 24:479-480.
Carlsson et al., “Protein Thiolation and Reversible Protein-Protein Conjugation,” (1978) Biochem. J. 173:723-737.
Carter, P., “Potent antibody therapeutics by design,” (2006) Nature Reviews Immunology 6:343-357.
Casset et al., “A peptide mimetic of an anti-CD4 monoclonal antibody by rational design.” Biochem Biophys Res Commun. Jul. 18, 2003; 307(1):198-205.
CellTiter-Glo Luminescent Cell Viability Assay, Promega Corp. Technical Bulletin TB288, dated Jan. 13, 2012 (14 pages).
Chakravarty et al., ““Plasmin-Activated Prodrugs for Cancer Chemotherapy. 2. Synthesis and Biological Activity of Peptidyl Derivatives of Doxorubicin,”” (1983) J. Med. Chern. 26:638-644.
Chan, J. and Watt, V.M., “eek and erk, new members of the eph subclass of receptor protein-tyrosine kinases,” Oncogene 6 (6), 1057-1061 (1991).
Chang et al., “Molecular cloning of mesothelin, a differentiation antigen present on mesothelium, mesotheliomas, and ovarian cancers,” Proc. Natl. Acad. Sci. U.S.A. 93(1):136-140 (1996).
Chen, Z. et al., ““A novel approach to the synthesis of cytotoxic C2-C3 unsaturated pyrrolo[2,1- c][1,4]benzodiazepines (PBDs) with conjugated acrylyl C2-substituents,”” Biorg. Med. Chem. Lett. (2004) 14:1547-1549.
Cheung, A., “Direct liquid chromatography determination of the reactive imine SJG-136 (NSC 694501),” J. Chromat. B: Anal. Techn. Biomed. Life Sci. (2005) 822(1 -2):10-20.
Child et al., “Translational Control by an Upstream Open Reading Frame in the HER-2/neu Transcript,” (1999) J. Bioi. Chern. 274: 24335-24341.
Cho et al., ““Structure of the extracellular region of HER2 alone and in complex with the Herceptin Fab,”” Nature 421,756-760, 2003.
Ciccodicola, A , et al., ““Molecular characterization of a gene of the ‘EGF family’ expressed in undifferentiated human NTERA2 teratocarcinoma cells,”” EMBO J. 8(7):1987-1991 (1989).
Cipolla, L., “Pyrrolo[2,1-c][1,4]benzodiazepine as a scaffold for the design and synthesis of anti-tumour drugs,” Anti-Cancer Agents in Medicinal Chemistry (Jan. 2009) 9(1):1-31.
Clackson et al., “Making antibody fragments using phage display libraries,” (1991) Nature, 352:624-628.
Clark H.F., et al., “The Secreted Protein Discovery Initiative (SPDI], a Large-Scale Effort to Identify Novel Human Secreted and Transmembrane Proteins: A Bioinformatics Assessment,” Genome Res. 13, 2265-2270, 2003.
Clingen, P.H., “The role of nucleotide excision repairand homologous recomination in the sensitivity of mammalian cells to the minor groove crosslinking pyrrolo[2,1-c][1,4]benzodiazepine dimer SJG-136 (NSC694501),” Proceedings of the American Association for Cancer Research Annual Meeting (Jul. 2003) 44:524.
Clingen, P.H., “the XPF-ERCC1 endonuclease and homologous recombination contribute to the repair of minor groove DNA interstrand crosslinks in mammalian cells produced by the pyrrolo[2,1-c][1,4]benzodiazepine dimer SJG-136,” Nucl. Acids Res. (2005) 33(10):3283-3291.
Clinical Trial, “Translational research: 4 ways to fix the clinical trial.” 2011, http://www.nature.com/news/2011/110928/full/477526a.html.
Collins, “Generation and initial analysis of more than 15,000 full-length human and mouse eDNA sequences,” Proc. Natl. Acad. Sci. U.S.A. 99 (26):16899-16903 (2002).
Cooper, N et al., “Synthesis of novel PBDs as anti-tumour agents,” Chem. Commun. (2002) 16:1764-1765.
Cooper, N., “Design, Synthesis and Evaluation of Novel C2-Aryl Pyrrolobenzodiazepines as Potential Anticancer Agents,” Thesis submitted to School of Pharmacy, University of London, Dated Oct. 5, 2006.
Corey E. Quinn JE, Buhler Kr, et al., “LuCap35: a new model of prostate cancer progression to androgen Rindependence.” The Prostate 2003;55:239-46.
Courtney, S. M. et al., “A new convenient procedure for the synthesis of pyrrolo[2,1-c][1,4]benzodiazepines”, Tetrahedron Letters, vol. 34, No. 33, 5327-28 (1993).
Coussens L., et al., “Tyrosine Kinase Receptor with Extensive Homology to EGF Receptor Shares Chromosomal Location With neu Oncogene,” Science (1985) 230(4730):1132-1139.
Cragg et al., “The alternative transcript of CD79b is overexpressed in B-CLL and inhibits signaling for apoptosis,” Blood (2002) 100 (9):3068-3076.
Cree et al., “Methotrexate chemosensitivity by ATP luminescence in human leukemia cell lines and in breast cancer primary cultures: comparison of the TCA-100 assay with a clonogenic assay,” (1995) AntiCancer Drugs 6:398-404.
Crouch et al., “The use⋅ of ATP bioluminescence as a measure of cell proliferation and cytotoxicity,” (1993) J. Immunol. Meth. 160:81-88.
Dall'Acqua, W. F. et al., “Antibody humanization by framework shuffling” Methods, 36, 43-60 (2005).
Dattolo, G. et al., “Polycondensed nitrogen heterocycles. IX. 5,6-dihydro-7H-pyrrolo[1,2-d][1,4]benzodiazepin-6-one,” J. Heterocyclic. Chem. (1980) 17:701-703.
Davis et al., “Identification of a family of Fe receptor homo logs with preferential B cell expression,” (2001) Proc. Natl. Acad. Sci. USA 98(17):9772-9777.
De Groot et al., ““Cascade-Release Dendrimers”” Liberate All End Groups upon a Single Triggering Event in the Dendritic Core, (2003) Angew. Chern. Int. Ed. 42:4490-4494.
De Groot et al., “Elongated Multiple Electronic Cascade and Cyclization Spacer Systems in Activatible Anticancer Prodrug for Enhanced Drug Release,” (2001) J. Org. Chern. 66:8815-8830.
De Pascalis et al., “Grafting of “abbreviated” complementarity-determining regions containing specificity-determining residues essential for ligand contact to engineer a less immunogenic humanized monoclonal antibody.” J Immunol. Sep. 15, 2002;169(6):3076-84.
Dennis et al., (2002) “Albumin Binding as a General Strategy for Improving the Pharmacokinetics of Proteins” J Biol Chem. 277:35035-35043.
Dijke, P., et al., “Characterization of Type I Receptors for Transforming Growth Factor-beta and Activin,” Science 264 (5155):101-104 (1994).
Dobner et al., “Differentiation-specific expression of a novel G protein-coupled receptor from Burkitt's lymphoma,” (1992) Eur. J. Immunol. 22:2795-2799.
Dong, Q. et al., “Reductive cleavage of TROC groups under neutral conditions with cadmium-lead couple,” Tetrahedron Lett. (1995) 36(32):5681-5682.
Dono et al., “Isolation and Characterization of the CRI PTO Autosomal Gene and Its X-linked Related Sequence,” Am. J. Hum. Genet. 49:555-565, 1991.
Dornan et al., “Therapeutic potential of an anti -CD79b antibody-drug conjugate, anti-CD79b-vc-MMAE, for the treatment of non-Hodgkin lymphoma,” (2009) Blood 114(13):2721-2729.
Doronina et al., “Enhanced Activity of Monomethylauristatin F through Monoclonal Antibody Delivery: Effects of Linker Technology on Efficacy and Toxicity,” (2006) Bioconj. Chem. 17:114-124.
Doronina, S.O. et al., “Development of potent monoclonal antibody auristatin conjugates for cancer therapy,” Nature Biotech. (2003) 21:778-784.
Dorwald, F.Z., Side Reactions in Organic Synthesis: a Guide to Successful Synthesis Design, Weinheim: Wiley-VCH Verlag GmbH & Co., KGaA (2005) Preface.
Doyle, M., “Response of Staphylococcus aureus to subinhibitory concentrations of a sequence-selective, DNA minor groove cross-linking pyrrolobenzodiazepine dimer,” J. Antimicrob. Chemo., Aug. 2009, 64(5):949-959.
Dubowchik et al., “Cathepsin B-sensitive dipeptide prodrugs. 2. Models of anticancer drugs paclitaxel (Taxol), mitomycin C and doxorubicin.” Bioorganic & Medicinal Chemistry Letters, 8:3347-3352, (1998).
Dubowchik et al., “Cathepsin B-sensitive dipeptide prodrugs. 1. A model study of structural requirements for efficient release of doxorubicin.” Bioorg Med Chem Lett. Dec. 1, 1998;8(23):3341-6.
Dubowchik et al., “Cathepsin B-Labile Dipeptide Linkers for Lysosomal Release of Doxorubicin from Internalizing Immunoconjugates: Model Studies of Enzymatic Drug Release and Antigen-Specific In Vitro Anticancer Activity,” Bioconjugate Chem. (2002) 13, 855-869.
Dubowchik, et al., “Monomethoxytrityl (MMT) as a Versatile Amino Protecting Group for Complex Prodrugs of Anticancer Compounds Sensitive to Strong Acids, Bases and Nucleophiles,” (1997) Tetrahedron Letters. 38:5257-5260.
Dumoutier L., et al., “Cutting Edge: STAT Activation by IL-19, IL-20 and mda-7 Through IL-20 Receptor Complexes of Two Types,” J. Immunol. 167, 3545-3549, 2001.
Dupont, C. et al., “Synthesis of rhazinilam analogue acting as an inhibitor of tubulin assembly,” Tetrahedron Lett. (2000) 41:5853-5856.
Ehsani A., et al., “Characterization of a New Allele of the Human ERBB2 Gene by Allele-Specific Competition Hybridization,” (1993) Genomics 15, 426-429.
Elshourbagy N.A., et al., “Molecular Characterization and Regulation of the Human Endothelin Receptors,” J. Biol. Chem. 268, 3873-3879, 1993.
Erickson et al., “Antibody-Maytansinoid Conjugates Are Activated in Targeted Cancer Cells by Lysosomal Degradation and Linker-Dependent Intracellular Processing,” (2006) Cancer Res. 66(8): 4426-4433.
Farmer, J.D. et al., “Synthesis and DNA crosslinking ability of a dimeric anthramycin analog,” Tetrahedron Letters (1988) 29(40):5105-5108, Abstract only.
Farmer, J.D. et al., “DNA binding properties of a new class of linked anthramycin analogs,”, Chemical Abstracts, Abstract No. 239940r, vol. 114, No. 25, 25 899-903 (1991).
Feild, J.A., et al., “Cloning and Functional Characterization of a Sodium-Dependent Phosphate Transporter Expressed in Human Lung and Small Intestine,” (1999) Biochem. Biophys. Res. Commun. 258 (3):578-582.
Fey, T. et al., “Silica-supported TEMPO catalyst: synthesis and application in the anelli oxidation of alcohols,” J. Org. Chem. (2001) 66:8154-8159.
Fields, G. and Noble, R. (1990) “Solid phase peptide synthesis utilizing 9-fluoroenylmethoxycarbonyl amino acids”, Int. J. Peptide Protein Res. 35:161-214.
Firsching, A. et al., “Antiproliferative and angiostatic activity of suramin analogues,” Cancer Res. (1995) 55:4957-4961.
Flanagan et al., “The ephrins and Eph receptors in neural development,” Annu. Rev. Neurosci. 21:309-345 (1998).
Foloppe, M.P. et al., “DNA-binding properties of pyrrolo[2,1-c][1,4]benzodiazephine N10-C11 amidines,” Eur. J. Med. Chem., 31, 407-410 (1996).
Fox et al., “cDNA cloning and tissue distribution of five human EPH-like receptor protein-tyrosine kinases,” Oncogene 10 (5):897-905 (1995).
Fuchs S., et al., “Functional Characterization of Three Mutations of the Endothelin B Receptor Gene in Patients With Hirschsprung's Disease: Evidence for Selective Loss of Gi Coupling,” Mol. Med. 7, 115-124, 2001.
Fujisaku et al., “Genomic Organization and Polymorphisms of the Human C3d/Epstein-Barr Virus Receptor,” (1989) J. Biol. Chem. 264 (4):2118-2125.
Fujisawa Pharmaceutical Co. Ltd., “Benzodiazepine derivatives,” SciFinder Scholar, 2-3 (2002).
Fujisawa Pharmaceutical Co., Ltd., Abstract No. 139983k, “Benzodiazepine derivatives”, Chemical Abstracts, vol. 99, No. 17, 603 (1983).
Fujisawa Pharmaceutical Co., Ltd., Abstract No. 72145x, “Benzodiazepine derivatives”, Chemical Abstracts, vol. 98, No. 9, 638 (1983).
Fukuyama, T. et al., “Total Synthesis of (+)-Porothramycin B,” Tetrahedron Letters, vol. 34, 16, 2577-2580 (1993).
Gallmeier, E., “Targeted disruption of FANCC and FANCG in human cancer provides a preclinical model for specific therapeutic options,” Gastroenterology (2006) 130(7):2145-2154.
Gary S.C., et al., “cDNA cloning chromosomal localization, and expression analysis of human BEHAB/brevican, a brain specific proteoglycan regulated during cortical development and in glioma,” Gene 256, 139-147, 2000.
Gaugitsch, H.W., et al., “A novel transiently expressed, integral membrane protein linked to cell activation. Molecular cloning via the rapid degradation signal AUUUA.,” (1992) J. Biol. Chem. 267 (16):11267-11273.
Gavezzotti, A., “Are crystal structures predictable?” Acc. Chem. Res. (1994) 27:309-314.
Genbank accession No. AF116456 (1999).
Genbank accession No. AF179274 (2001).
Genbank accession No. AF229053 (2000).
Genbank accession No. AF343662 (2001).
Genbank accession No. AF343663 (2001).
Genbank accession No. AF343664 (2001).
Genbank accession No. AF369794 (2001).
Genbank accession No. AF397453 (2001).
Genbank accession No. AF455138 (2003).
Genbank accession No. AJ297436 (2008).
Genbank accession No. AK026467 (2006).
Genbank accession No. AK089756 (2010).
Genbank accession No. AL834187 (2008).
Genbank accession No. AX092328 (2001).
Genbank accession No. AY158090 (2003).
Genbank accession No. AY260763 (2003).
Genbank accession No. AY275463 (2003).
Genbank accession No. AY358085 (2003).
Genbank accession No. AY358628 (2003).
Genbank accession No. AY358907 (2003).
Genbank accession No. AY506558 (2004).
Genbank accession No. BC017023 (2006).
Genbank accession No. CAF85723 (2004).
Genbank accession No. CQ782436 (2004).
Genbank accession No. M11730 (1995).
Genbank accession No. M18728 (1995).
Genbank accession No. M26004 (1993).
Genbank accession No. NM_000626 (2013).
Genbank accession No. NP_005573.1 (2007).
Genbank accession No. NP_112571.1 (2007).
Getz et al., “A Comparison between the Sulfhydryl Reductants Tris(2-carboxyethyl)phosphine and Dithiothreitol for Use in Protein Biochemistry,” (1999) Anal. Biochem. vol. 273:73-80.
Glynne-Jones et al., “TENB2, a Proteoglycan Identified in Prostate Cancer That is Associated With Disease Progression and Androgen Independence,” (2001) Int J Cancer. Oct 15; 94(2): 178-184.
Gordon et al., “Somatic hypermutation of the B cell receptor genes B29 (Igβ, CD79b) and mb1 (Igα, CD79a),” PNAS, Apr. 1, 2003, vol. 100, No. 7, 4126-4131.
Greene, T.W. and Wuts, P.G.M., Protective Groups in Organic Synthesis, John Wiley & Sons, 2nd ed., Ch 7, 315-345 (1991).
Greene, T.W. et al., Protective Groups in Organic Synthesis, John Wiley & Sons (1999) 3rd Edition, 23-200.
Greene, T.W. et al., Protective Groups in Organic Synthesis, John Wiley & Sons (1999) 3rd Edition, 503-549.
Gregson, S. et al., “Synthesis of a novel C2/C2′-exo unsaturated pyrrolobenzodiazepine cross-linking 40agent with remarkable DNA binding affinity and cytotoxicity,” Chemical Communications, 797-798 (1999).
Gregson, S.J. et al., “Effect of C2/C3-endo unsaturation on the cytotoxicity and DNA-binding reactivity of pyrrolo-[2,1-c][1,4]-benzodiazepines,” Bioorg. Med. Chem. Lett. (2000) 10(16):1849-1851.
Gregson, S.J. et al., “Linker length modulates DNA cross-linking reactivity and cytotoxic potency of C8/C8′ ether-linked C2-exo-unsaturated pyrrolo[2,1-c][1,4]benzodiazepine (PBD) dimers,” J. Med. Chem. (2004) 1161-1174.
Gregson, S.J. et al., “Synthesis of the first example of a C2-C3/C2′-C3′-endo unsaturated pyrrolo[2,1 c][1,4]benzodiazepine dimer,” Biorg. Med. Chem. Lett. (2001) 11:2859-2862.
Gregson, S.J. et al., “Synthesis of the first examples of A-C8/C-C2 amide-linked pyrrolo[2,1-c][1,4]benzodiazepine dimers,” Biorg. Med. Chem. Lett. (2003) 13:2277-2280.
Gregson, S.J. et al., “Design, Synthesis and Evaluation of a Novel Pyrrolobenzodiazepine DNA-Interactive Agent with Highly Efficient Cross-Linking Ability and Potent Cytotoxicity”, J. Med. Chem., 44: 737-748 (2001).
Gregson, S.J. et al., “Effect of C2-exo Unsaturation on the Cytotoxicity and DNA-Binding Reactivity of Pyrrolo[2,1-c]1,4]benzodiazepines”, Bioorganic & Medicinal Chemistry Letters, 10:1845-1847 (2000).
Gu Z., et al., “Prostate stem cell antigen (PSCA) expression increases with high gleason score, advanced stage and bone metastasis in prostate cancer,” Oncogene 19, 1288-1296, 2000.
Guichard, S.M., “Influence of P-glycoprotein expression on in vitro cytotoxicity and in vivo antitumour activity of the novel pyrrolobenzodiazepine dimer SJG-136,” Eur. J. Cancer (2005) 41 (12):1811-1818.
Guiotto, A. et al., “Synthesis of novel C7-aryl substituted pyrrolo[2,1-c][1,4]benzodiazepines (PBDs) via Pro-N10-troc protection and Suzuki coupling,” Bioorganic & Medicinal Chemistry Letters, 8, No. 21, 3017-3018 (1998).
Guselnikov et al., “A family of highly diverse human and mouse genes structurally links leukocyte FcR, gp42 and PECAM-1,” Immunogenetics 54 (2):87-95 (2002).
Ha et al., “Molecular Cloning and Expression Pattern of a Human Gene Homologous To the Murine mb-1 GENE,” (1992) J. Immunol. 148(5):1526-1531.
Hadjivassileva, T., “Antibacterial activity of pyrrolobenzodiazepine dimers, a novel group of DNA-binding compounds,” Abstracts of the Interscience Conference on Antimicrobial Agents and Chemotherapy (Oct.-Nov. 2004) 44:202.
Hadjivassileva, T., “Interactions of pyrrolobenzodiazepine dimers and duplex DNA from methicillin-resistant Staphylococcus aureus,” Int. J. Antimicrob. Agents (2007) 29(6):672-678.
Hadjivassileva, T., “Pyrrolobenzodiazepine dimers: novel sequence-selective, DNA-interactive, cross-linking agents with activity against Gram-positive bacteria,” J. Antimicrob. Chemo. (2005) 56(3):513-518.
Haendler B., et al., “Molecular Cloning of Human Endothelin (ET) Receptors ETA and ETB,” J. Cardiovasc. Pharmacal. 20, s1-S4, 1992.
Hamaguchi, A., “DNA cross-linking and in vivo antitumour activity of the extended pyrrolo[2,1-c][1,4]benzodiazepine dimer SG2057 (DRG-16),” Ejc Supplements (Nov. 2006) 4(12):96.
Hamann P. “Monoclonal antibody-drug conjugates,” (2005) Expert Opin. Ther. Patents 15(9):1087-1103.
Hamblett et al., “Effects of Drug Loading on the Antitumor Activity of a Monoclonal Antibody Drug Conjugate,” (2004) Clin. Cancer Res. 10:7063-7070.
Handbook of Food Additives, 2nd Ed. (eds. M. Ash and I. Ash), Synapse Information Resources, Inc., Endicott, New York, USA (2001).
Handbook of Pharmaceutical Excipients, 2nd edition, 1994, Edited by Ain ley Wade and Paul J. Weller.
Hara et al., “DC 102, a new glycosidic pyrrolo(1,4)benzodiazepine antibiotic produced by streptomyces sp.”, J. Antibiotics, 41, 702-704 (1988).
Hartley J A: “The development of pyrrolobenzodiazepines as antitumour agents”, Expert Opinion on Investigational Drugs, Ashley Publications Ltd., vol. 28, No. 6, Jan. 1, 2011, pp. 733-744.
Hartley, J.A. et al., “SG2285, a novel C2-aryl-substituted pyrrolobenzodiazepine dimer prodrug that cross-links DNA and exerts highly potent antitumor activity,” Cancer Res., Sep. 2010, 70(17):6849-6858.
Hartley, J.A. et al., “SJG-136 (Nsc 694501), a novel rationally designed DNA minor groove interstrand cross-linking agent with potent and broad spectrum antitumor activity. Part 1: Cellular pharmacology, in vitro and initial in vivo antitumor activity,” Cancer Res. (2004) 64:6693-6699.
Hartley, J.A., “In vitro antitumor activity and in vivo DNA interstrand crosslinking by the novel pyrrolobenzodiazepine dimer SJG-136 (NSC 694501),” Proceedings of the American Association for Cancer Research Annual Meeting (Mar. 2002) 43:489.
Hartley, J.A., “SJG-136 (NSC-D694501)—a novel DNA sequence specific minor groove crosslinking agent with significant antitumor activity,” Proceedings of the American Association for Cancer Research Annual Meeting (Mar. 2000) 41:425.
Hashimoto et al., “Chromosomal localization, genomic structure, and allelic polymorphism of the human CD79a (Ig-alpha/mb-1) gene,” (1994) Immunogenetics 40(4 ):287-295.
Hay et al., “A 2-Nitroimidazole Carbamate Prodrug of 5-Amin0-1-(Chloromethyl)-3-[(5,6,7-Trimethoxyindol-2-yl)Carbonyl]-1,2-Dihydr0-3H-Benz[E]Indole (Amino-Seco-Cbi-Tmi) for Use With Adept and Gdept,” (1999) Bioorg. Med. Chern. Lett. 9:2237-2242.
Herdwijn et al., “Synthesis of trans(+ )6-phenoxyacetamido-1-methylene-3,3-dicarboxymethyl-1-carbapenam,” Canadian Journal of Chemistry. 1982, 60, 2903-2907.
Hermanson, G.T., “Heterobifunctional Cross-linkers,” (1996) Bioconjugate Techniques; Academic Press: New York, p. 228-286.
Hochhauser, D., “Phase I study of sequence-selective minor groove DNA binding agent SJG-136 in patients with advanced solid tumors,” Clin. Cancer Res., Mar. 2009, 15(6):2140-2147.
Hochlowski, J. et al., “Abbeymycin, a new anthramycin-type antibiotic produced by a streptomycete,” J. Antibiotics, 40, 145-148 (1987).
Hofstra R.M.W., et al., “A homozygous mutation in the endothelin-3 gene associated with a combined Waardenburg type 2 and Hirschsprung phenotype (Shah-Waardenburg syndrome)” Nat. Genet. 12, 445-447, 1996.
Hofstra R.M.W., et al., “Mutations in Hirschsprung Disease: When Does a Mutation Contribute to the Phenotype,” Eur. J. Hum. Genet. 5, 180-185, 1997.
Horie et al., “Identification and Characterization of TMEFF2, a Novel Surviv Factor for Hippocampal and Mesencephalic Neurons,” (2000) Genomics 67: 146-152.
Howard, P.W. et al., “Design, synthesis and biological evaluation of ZC-423, a novel C2-aryl substituted pyrrolobenzodiazepine (PBD) dimer,” Clinical Cancer Research (2005) 11(23):9015S-9016S (A205).
Howard, P.W. et al., “Synthesis of a novel C2/C2′-aryl-substituted pyrrolo[2,1-c][1,4]benzodiazepine dimer prodrug with improved water solubility and reduced DNA reaction rate,” Bioorg. Med. Chem., Sep. 2009, In Press, 4 pages now: Sep. 2009, 19:6463-6466.
Howard, P.W. et al., “The design, synthesis and biological evaluation of a set of C2-aryl substituted pyrrolo[2,1-c][1,4]benzodiazepine dimers,” EJC Supplements (2006) 4(12):95—Poster Abstract 301.
Howard, P.W., “Design, synthesis and biological evaluation of novel C2-aryl-substituted pyrrolo[2,1-c][1,4]benzodiazepine monomers,” Proceedings of the American Association for Cancer Research Annual Meeting (Apr. 2006) 47:132.
Hubert, R.S., et al., “STEAP: A prostate-specific cell-surface antigen highly expressed in human prostate tumors,” (1999) Proc. Natl. Acad. Sci. U.S.A. 96(25):14523-14528.
Hurley, L. and Needham-Vandevanter, D “Covalent Binding of Antitumor Antibiotics in the Minor Groove of DNA. Mechanism of Action of CC-1065 and the yrrolo(1,4)benzodiazepines,” Acc. Chem. Res., 19, 230-237 (1986).
Ide et al., “Cloning of human bone morphogenetic protein type IB receptor (BMPRIB) and its expression in prostate cancer in comparison with other BMPRs,” Oncogene (1997) 14, 1377-1382.
International Search Report and Written Opinion for Application No. PCT/EP2012/070233 dated Jan. 28, 2013 (8 pages).
International Search Report and Written Opinion for Application No. PCT/EP2013/071346 dated Feb. 5, 2014 (11 pages).
International Search Report and Written Opinion for Application No. PCT/EP2014/054958 dated Jul. 2, 2014 (13 pages).
International Search Report and Written Opinion for Application No. PCT/US2011/032664 dated Aug. 19, 2011 (13 pages).
International Search Report and Written Opinion for Application No. PCT/US2011/032668 dated May 26, 2011 (12 pages).
Itoh et al., “Sibanomicin, a new pyrrolo(1,4)benzodiazepine antitumor antibiotic produced by a micromonospora sp.” J. Antibiotics, 41, 1281-1284 (1988).
Janjigian, Y.Y., “A phase I trial of SJG-136 (NSC#694501) in advanced solid tumors,” Cancer Chemotherapy and Pharmacology , Aug. 2009, 65(5):833-838.
Jeffrey et al., “Development and properties of beta-glucuronide linkers for monoclonal antibody-drug conjugates.” Bioconjugate Chemistry, 5, 2006, 17, 831-840. (Abstract).
Jeffrey, S.C. et al., “Development of Pyrrolobenzodiazepine-Based Antibody-Drug Conjugates for Cancer,” AACR Annual Meeting 2013, Abstract No. 4321.
Jeffrey, S.C., “Design, synthesis, and in vitro evaluation of dipeptide-based antibody minor groove binder conjugates,” J. Med. Chem. (2005) 48(5):1344-1358.
Jespers, L. S., “Guiding the Selection of Human Antibodies from Phage Display Repertoires to a Single Epitope of an Antigen” Nature Biotech., 12, 899-903 (1994).
Jia, L., “Interspecies differences in pharmacokinetics and time-dissociated toxicokinetics of SJG-136,” Proceedings of the American Association for Cancer Research Annual Meeting (Mar. 2004) 45:487.
Jia, L., “Use of the comet assay as a surrogate biomarker for the in vivo measurement of DNA damage to lymphocytes,” Proceedings of the American Association for Cancer Research Annual Meeting (Mar. 2004) 45:452-453.
Johnson & Goldin, “The clinical impact of screening and other experimental tumor studies.” Cancer Treat Rev. Mar. 1975; 2(1):1-31.
Jordan, V.C., ““Tamoxifen: a most unlikely pioneering medicine,”” Nature Reviews: Drug Discovery (2003) 2:205-213.
Junutula, et al., “Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index,” 2008b Nature Biotech., 26(8):925-932.
Kamal et al., “Synthesis and DNA-binding affinity of A-C8/C-C2 alkoxyamido-linked pyrrolo[2,1-c][1,4]benzodiazepine dimers” Biorg. Med. Chem. Lett. (2003) 13(22):3955-3958.
Kamal, A. et al., “Design, synthesis and evaluation of new noncross-linking pyrrolobenzodiazepine dimers with efficient DNA binding ability and potent antitumor activity,” J. Med. Chem. (2002) 45:4679-4688.
Kamal, A., “Development of pyrrolo[2,1-c][1,4]benzodiazepine beta-galactoside prodrugs for selective therapy of cancer by ADEPT and PMT,” Chemmedchem (2008) 3:794-802.
Kamal, A., “Remarkable DNA binding affinity and potential anticancer activity of pyrrolo[2,1-c][1,4]benzodiazepine-naphthalamide conjugates linked through piperazine side-armed alkane spacers,” Bioorg. Med. Chem. (2008) 16(15):7218-7224.
Kamal, A., “Remarkable enhancement in the DNA-binding ability of C2-fluoro substituted pyrrolo[2,1-c][1,4]benzodiazepines and their anticancer potential,” Bioorg. Med. Chem., Jan. 2009, 17(4):1557-1572.
Kamal, A., “Synthesis of fluorinated analogues of SJG-136 and their DNA-binding potential,” Bioorg. Med. Chem. Lett (2004) 14(22):5699-5702.
Kamal, A., et al., “An Efficient Synthesis of Pyrrolo[2,1-c][1,4] Benzodiazepine Antibiotics via Reductive Cyclization,” Bioorg. Med. Chem. Ltrs, 7, No. 14, 1825-1828 (1997).
Kamal, A., et al., “Synthesis of Pyrrolo [2,1-c][1,4]-Benzodiazepene Antibiotics: Oxidation of Cyclic Secondary Amine with TPAP”, Tetrahedron, v. 53, No. 9, 3223-3230 (1997).
Kamal, et al., “Synthesis of pyrrolo[2,1-c][1,4]benzodiazepines via reductive cyclization of w-azido carbonyl compounds by TMSI: an efficient preparation of antibiotic DC-81 and its dimers,” Biorg. Med. Chem. Lett. (2000) 10:2311-2313.
Kaneko, T. et al., “Bicyclic and tricyclic analogues of anthramycin,” J. Med. Chem. (1985) 28:388-392.
Kang, G.-D. et al., “Synthesis of a novel C2-aryl substituted 1,2-unsaturated pyrrolobenzodiazepine,” Chem. Commun. (2003) 1680-1689.
Kasahara et al., “Nucleotide sequence of a chimpanzee DOB eDNA clone,” (1989) Immunogenetics 30(1):66-68.
King et al., “Facile synthesis of maleimide bifunctional Jinkers,” (2002) Tetrahedron Letters 43:1987-1990.
Kingsbury et al., ““A Novel Peptide Delivery System Involving Peptidase Activated Prodrugs as Antimicrobial Agents. Synthesis and Biological Activity of Peptidyl Derivatives of 5 Fluorouracil,”” (1984) J. Med. Chern. 27:1447-1451.
Kohn, K., “Anthramycin,” Antibiotics III, Springer-Verlag, NY, 3-11 (1975).
Kojima et al., “Molecular Cloning and Expression of Megakaryocyte Potentiating Factor eDNA,” The Journal of Biological Chemistry, vol. 270, No. 37, Issue of Sep. 15, pp. 21984-21990, 1995.
Konishi, M. et al., “Chicamycin, a new antitumor antibiotic II. Structure determination of chicamycins A and B,” J. Antibiotics, 37, 200-206 (1984).
Kovtun et al., “Antibody-Drug Conjugates Designed to Eradicate Tumors with Homogeneous and Heterogeneous Expression of the Target Antigen,” (2006) Cancer Res. 66(6):3214-3121.
Kuhns J.J., et al., “Poor Binding of a HER-2/neu Epitope (GP2) to HLA-A2.1 is due to a Lack of Interactions with the Center of the Peptide,” J. Biol. Chem. 274, 36422-36427, 1999.
Kunimoto et al., “Mazethramycin, a new member of anthramycin group antibiotics,” J. Antibiotics, 33, 665-667 (1980).
Kurebayashi et al., “Isolation and characterization of a new human breast cancer cell line, KPL-4, expressing the Erb B family receptors and interleukin 6,” (1999) Brit. Jour. Cancer 79(5-6):707-717.
Lambert J., “Drug-conjugated monoclonal antibodies for the treatment of cancer,” (2005) Current Opin. in Pharmacal. 5:543-549.
Langley, D.R. and Thurston, D.E., “A versatile and efficient synthesis of carbinolamine-containing pyrrolo[1,4]benzodiazepines via the cyclization of N-92-aminobenzoyl)pyrrolidine-2-carboxaldehyde diethyl thioacetals: total synthesis of prothracarcin,” J. Org. Chem., 52, 91-97 (1987).
Langlois, N. et al., “Synthesis and cytotoxicity on sensitive and doxorubicin-resistant cell lines of new pyrrolo[2,1-c][1,4]benzodiazepines related to anthramycin,” J. Med. Chem. (2001) 44:3754-3757.
Larhammar et al., “Sequence of Gene and eDNA Encoding Murine Major Histocompatibility Complex Class II Gene AP2*,” (1985) J. Biol. Chem. 260(26):14111-14119.
Launay et al., “TRPM4 is a Ca2+ -Activated Nonselective Cation Channel Mediating Cell Membrane Depolarization,” Cell 109 (3):397-407 (2002).
Law et al., “Lymphocyte Activation Antigen CD70 Expressed by Renal Cell Carcinoma is a Potential Therapeutic Target for Anti-CD70 Antibody-Drug Conjugates,” (2006) Cancer Res. 66(4):2328-2337.
Lazar et al., “A molecular immunology approach to antibody humanization and functional optimization” Molecular Immunology, 2007, 44(8), 1986-1998.
Le et al., “Primary structure and expression of a naturally truncated human P2X Atp receptor subunit from brain and immune system,” (1997) FEBS Lett. 418(1-2):195-199.
Leber, J.D et al., “A revised structure forsibiromycin,” J. Am. Chem. Soc., 110, 2992-2993 (1988).
Leimgruber, W. et al., “Isolation and characterization of anthramycin, a new antitumor antibiotic,” J. Am. Chem. Soc., 87, 5791-5793 (1965).
Leimgruber, W. et al., “The structure of anthramycin,” J. Am. Chem. Soc., 87, 5793-5795 (1965).
Leimgruber, W. et al., “Total synthesis of anthramycin,” J. Am. Chem. Soc., 90, 5641-5643 (1968).
Levenson et al., “MCF-7: The First Hormone-responsive Breast Cancer Cell Line,” (1997) Cancer Res. 57(15):3071-3078.
Liang et al., “The Gene fora Novel Transmembrane Protein Containing Epidermal Growth Factor and Follistatin Domains is Frequently Hypermethylated in Human Tumor Cells,” (2000) Cancer Res. 60:4907-4912.
Lonberg, “Fully Human antibodies from transgenic mouse and phage display platforms” Curr. Opinion, 20(4), 450-459 (2008).
Lown et al., “Molecular Mechanism of Binding of Pyrrolo(1,4)benzodiazepine antitumour agents to deoxyribonucleic acid—anthramycin and tomaymycin,” Biochem. Pharmacol. (1979), 28 (13), 2017-2026.
Manfre et al., “Syntheses of Proline Analogues as Potential Mechanism-Based Inhibitors of Proline Dehydrogenase: 4-Methylene-L-, (E)- and (Z)-4-(Fluoromethylene)-L-, cis- and trans-5-Ethynyl-(±)-, and cis- and trans -5-Vinyl-L- proline,” J. Org. Chem. 1992, 57, 2060-2065.
Marin, D., “Voltammetric studies of the interaction of pyrrolo[2,1-c][1,4]benzodiazepine (PBD) monomers and dimers with Dna,” J. Electroanal. Chem. (2006) 593(1-2):241 -246.
Marks et al., “By-passing Immunization, Human Antibodies from V-gene Libraries Displayed on Phage,” (1991) J. Mol. Biol., 222:581-597.
Martin, C. et al., “Sequence-selective interaction of the minor-groove interstrand cross-linking agent SJG-136 with naked and cellular DNA: footprinting and enzyme inhibition studies,” Biochem. (2005) 44(11):4135-4147.
Mastroberardino et al., “Amino-acid transport by heterodimers of 4F2hc/CD98 and members of a permease family,” Nature 395 (6699):288-291 (1998).
McDonagh, “Engineered antibody-drug conjugates with defined sites and stoichiometries of drug attachment,” (2006) Protein Eng. Design & Sel. 19(7): 299-307.
Mendoza et al., “Inhibition of Ligand-mediated HER2 Activation in Androgen-independent Prostate Cancer,” (2002) Cancer Res. 62:5485-5488.
Miller et al., “Design, Construction, and In Vitro Analyses of Multivalent Antibodies,” (2003) Jour, of Immunology 170:4854-4861.
Miller et al., “IRTAs: a new family of immunoglobulinlike receptors differentially expressed in B cells,” Blood 99 (8):2662-2669 (2002).
Miura et al., “Molecular cloning of a human RP105 homologue and chromosomal localization of the mouse and human RP105 genes (Ly64 and LY64).” Genomics. Dec. 15, 1996;38(3):299-304.
Miura et al., “RPIOS is Associated With MD-1 and Transmits an Activation Signal in Human B Cells,” (1998) Blood 92:2815-2822.
Moore M., et al., “Molecular cloning of the eDNA encoding the Epstein-Barr virus/C3d receptor (complement receptor type 2) of human B lymphocytes,” Proc. Natl. Acad. Sci. U.S.A. 84, 9194-9198, 1987.
Mori, M. et al., “Total syntheses of prothracarcin and tomaymycin by use of palladium catalyzed carbonylation,” Tetrahedron (1986) 42(14):3793-3806.
Morrison et al., “Chimeric human antibody molecules: Mouse antigen-binding domains with human constant region domains,” (1984) Proc. Natl. Acad. Sci. USA, 81:6851-6855.
Mountzouris, J.A. et al., “Comparison of a DSB-120 DNA interstrand cross-linked adduct with the corresponding bis-Tomamycin adduct,” J. Med. Chem. (1994) 37:3132-3140.
Muller et al., “Cloning and sequencing of the eDNA encoding the human homologue of the murine immunoglobulin-associated protein B29,” (1992) Eur. J. Immunol. 22 (6): 1621-1625.
Mungall A.J., et al., “The DNA sequence and analysis of human chromosome 6,” Nature 425, 805-811, 2003.
Nadler et al., “B4, a human B lymphocyte-associated antigen expressed on normal, mitogen-activated, and malignant B lymphocytes,” Journal of Immunology, 1983, 131(1):244-250.
Nagasaka, T. and Koseki, Y, “Stereoselective Synthesis of Tilivalline,” Journal of Organic Chemistry, vol. 63, No. 20, 6797-6801 (1998).
Nagasaka, T. et al., “Stereoselective Synthesis of Tilivalline,” Tetrahedron Letters, 30:14, 1871-1872 (1989).
Nagase T., et al., “Prediction of the Coding Sequences of Unidentified Human Genes. XVII. The Complete Sequences of 100 New eDNA Clones from Brain Which Code for Large Proteins in vitro,” (2000) DNA Res. 7 (2):143-150.
Nakamuta M., et al., “Cloning and Sequence Analysis of A cDNA Encoding Human Non-Selective Type of Endothelin Receptor,” Biochem. Biophys. Res. Commun. 177, 34-39, 1991.
Nakayama et al., “Altered Gene Expression upon BCR Cross-Linking in Burkitt's Lymphoma B Cell Line,” (2000) Biochem. Biophys. Res. Commun. 277(1):124-127.
Narayanaswamy, M., “A novel HPLC/MS assay to measure DNA interstrand cross-linking efficacy in oligonucleotides of varying sequence,” EJC Supplements (Nov. 2006) 4(12):92-93.
Narayanaswamy, M., “An assay combinding high-performance liquid chromatography and mass spectrometry to measure DNA interstrand cross-linking efficiency in oligonucleotides of varying sequences,” Anal. Biochem. (2008) 374(1):173-181.
Narayanaswamy, M., “Use of HPLC-MS to characterize novel mono and intrastrand cross-linked DNA adducts formed by the sequence-selective DNA-interactive agent SJG-136,” Proceedings of the American Association for Cancer Research Annual Meeting (Apr. 2007) 48:760-761.
Narukawa, Y., “General and efficient synthesis of 2-alkylcarbapenems synthesis of dethia carba analogs of clinically useful carbapenems via palladium-catalyzed cross-coupling reaction,” Tetrahedron (1997) 53:539-556.
Naruse et al., “The HLA-DOB gene displays limited polymorphism with only one amino acid substitution,” (2002) Tissue Antigens 59:512-519.
Neuberger and Williams, “The intron requirement for immunoglobulin gene expression is dependent upon the promoter,” (1988) Nucleic Acids Res. 16:6713-6724.
Nicolaou et al., “Calicheamicin θ: A Rationally Designed Molecule with Extremely Potent and Selective DNA Cleaving Properties and Apoptosis Inducing Activity,” Angew Chem. Intl. Ed. Engl. (1994) 33:183-186.
Nilius et al., “Voltage Dependence of the Ca2+ -activated Cation Channel TRPM4,” The Journal of Biological Chemistry, vol. 278, No. 33, Issue of Aug. 15, pp. 30813-30820, 2003.
O'Neil, Chemical Abstract No. 171573p, “The synthesis of Functionalized Pyrrolo-[2,1-c][1,4]-Benzodiazepines”, Chemical Abstracts, vol. 126, No. 13, 618 (1997).
O'Neil, I.A., et al., “The Synthesis of Functionalized Pyrrolo-[2,1-c][1,4]-Benzodiazepines,” Synlett, 75-78 (1997).
O'Neil, I.A. et al., “Dppe: A Convenient Replacement for Triphenylphosphine in the Staudingerand Mitsunobu Reactions”, Tetrahedron Letters, vol. 39, No. 42, 7787-7790 (1998).
Ogawa Y., et al., “Molecular Cloning of a Non-Isopeptide-Selective Human Endothelin Receptor,” Biochem. Biophys. Res. Commun. 178, 248-255, 1991.
Okamoto Y., et al. “Palmitoylation of Human Endothelin B,” Biol. Chem. 272, 21589-21596, 1997.
Parrish-Novak J., et al., “Interleukins 19, 20, and 24 Signal through Two Distinct Receptor Complexes,” J. Biol. Chem. 277, 47517-47523, 2002.
Paul, Fundamental Immunology, 3rd Edition, pp. 292-295.
Payne, G. “Progress in immunoconjugate cancer therapeutics,” (2003) Cancer Cell 3:207-212.
PCT/US2012/059864 International Search Report and Written Opinion dated Dec. 21, 2012 (7 pages).
Pepper, C., “Fludarabine-mediated suppression of the excision repair enzyme ERCC1 contributes to the cytotoxic synergy with the DNA minor groove crosslinking agent SJG-136 (NSC 694501) in chronic lymphocytic leukaemia cells,” Br. J. Cancer (2007) 97(2):253-259.
Pepper, C.J. et al., “The novel sequence-specific DNA cross-linking agent SJG-136 (NSC 694501) has potent and selective in vitro cytotoxicity in human B-cell chronic lymphocytic leukemia cells with evidence of a p53-independent mechanism of cell kill,” Cancer Res. (2004) 64:6750-6755.
Phillips et al., “Targeting HER2-Positive Breast Cancer with Trastuzumab-DM1, an Antibody-Cytotoxic Drug Conjugate,” (2008) Cancer Res. 68(22):9280-9290.
Pingault V., et al., “SOX10 mutations in chronic intestinal pseudo-obstruction suggest a complex physiopathological mechanism,” (2002) Hum. Genet. 111, 198-206.
Pletnev S., et al., “Characterization of the Recombinant Extracellular Domains of Human Interleukin-20 Receptors and Their Complexe with Interleukin-19 and Interleukin-20,” (2003) Biochemistry 42:12617-12624.
Porkaa et al., “Cloning and Characterization of a Novel Six-Transmembrane Protein STEAP2, Expressed in Normal and Malignant Prostate,” Lab. Invest. 82 (11):1573-1582 (2002).
Prasad et al..“Human LAT1, a Subunit of System L Amino Acid Transporter: Molecular Cloning and Transport Function,” Biochem. Biophys. Res. Commun. 255 (2), 283-288 (1999).
Preud'Homme et al., “Structure and expression of the mb-1 transcript in human lymphoid cells,” (1992) Clin. Exp. Immunol. 90(1):141-146.
Puffenberger E.G., et al., “A Missense Mutation of the Endothelin-B Receptor Gene in Multigenic Hirschsprung's Disease,” Cell 79, 1257-1266, 1994.
Puzanov, I., “Phase I and pharmacokinetic trial of SJG-136 administered on a daily x5 schedule,” EJC Supplements (Nov. 2006) 4(12):93.
Quintas-Cardama, A., “Sequencing of subcloned PCR products facilitates earlier detetction of BCR-ABL1 and other mutants compared to direct sequencing of the ABL1 kinase domain,” Leukemia (2008) 22(4):877-878.
Rahman et al. “Antistaphylococcal activity of DNA-interactive pyrrolobenzodiazepine (PBD) dimers and PBD-biaryl conjugates.” J Antimicrob Chemother. Jul. 2012; 67(7):1683-96.
Rahman, K.M., “Effect of microwave irradiation on covalent ligand-DNA interactions,” Chem. Commun. (Cambridge, UK), Apr. 2009, 20:2875-2877.
Rahman, K.M., “Rules of DNA adduct formation for pyrrolobenzodiazepine (PBD) dimers,” Proceedings of the American Association for Cancer Research Annual Meeting (Apr. 2010) 51:851.
Rahman, K.M., “The pyrrolobenzodiazepine dimer SJG-136 forms sequencedependent intrastrand DNA cross-lins and monoalkylated adducts in addition to interstrand cross-links,” J. Am. Chem. Soc., Sep. 2009, 131 (38):13756-13766.
Rao et al., “Influence of diet on mammary cancer in transgenic mice bearing an oncogene expressed in mammary tissue,” (1997) Breast Cancer Res. and Treatment 45:149-158.
Reid, J.M., “LC-MS/MS assay and rat pharmacokinetics and metabolism of the dimeric pyrrolobenzodiazepine SJG-136,” Proceedings of the American Association for Cancer Research Annual Meeting (Mar. 2004) 45:1248.
Reiter R.E., et al., “Prostate stem cell antigen: A cell surface marker overexpressed in prostate cancer,” Proc. Natl. Acad. Sci. U.S.A. 95, 1735-1740, 1998.
Remmers et al., “Conformations of complexes between pyrrolo[1,4]benzodiazepines and DNA segments,” J Med Chem. Dec. 1986;29(12):2492-2503.
Rich, I.N., “Validation and development of a predictive paradigm for hemotoxicology using a multifunctional bioluminescence colony-forming proliferation assay,” Toxicological Sci. (2005) 87(2):427-441.
Rodrigues et al., “Synthesis and beta-lactamase-mediated activation of a cephalosporin-taxol prodrug,” (1995) Chemistry Biology 2:223-227.
Roguska et al., “A comparison of two murine monoclonal antibodies humanized by CDR-grafting and variable domain resurfacing,” Protein Eng, 1996, 9(10):895-904.
Roguska et al., “Humanization of murine monoclonal antibodies through variable domain resurfacing,” PNAS, 1994, 91(3):969-973.
Ross et al., “Prostate Stem Cell Antigen as Therapy Target: Tissue Expression and in Vivo Efficacy of an Immunoconjugate,” (2002) Cancer Res. 62:2546-2553.
Rudikoff et al., Single amino acid substitution altering antigen-binding specificity. Proc Natl Acad Sci U S A. Mar. 1982; 79(6):1979-83.
Sagnou, M.J. et al., “Design and Synthesis of Novel Pyrrolobenzodiazepine (PDB) Prodrugs for ADEPT and GDEPT,” Bioorganic & Medicinal Chemistry Letters, 10, 2083-2086 (2000).
Sakaguchi et al., “8 lymphocyte lineage-restricted expression of mb-1, a gene with CD3-like structural properties,” (1988) EMBO J. 7(11):3457-3464.
Sakamoto A, Yanagisawa M., et al., “Cloning and Functional Expression of Human cDNA For the ETB Endothelin Receptor,” Biochem. Biophys. Res. Commun. 178, 656-663, 1991.
Sanderson et al., “In vivo Drug-Linker Stability of an Anti-CD30 Dipeptide-Linked Auristatin Immunoconjugate,” (2005) Clin. Cancer Res. 11:843-852.
Scholler et al., “Soluble member(s) of the mesothelin/ megakaryocyte potentiating factor family are detectable in sera from patients with ovarian carcinoma,” Proc. Natl. Acad. Sci. USA vol. 96, p. 11531-11536, Sep. 1999.
Schroder and Lubke, The Peptides, vol. 1. pp 76-136 (1965) Academic Press.
Schweikart, K., “In vitro myelosuppression of SJG-136, a pyrrolobenzodiazepine dimer: comparison to bizelesin,” Proceedings of the American Association for Cancer Research Annual Meeting (Mar. 2004) 45:486.
Segawa et al., “Growth-related Renal Type II Na/Pi Cotransporter,” The Journal of Biolocjcal Chemistry, vol. 277. No. 22, Issue of May 31, pp. 19665-19672, 2002.
Semba K., et al., “A v-erbB-related protooncogene, c-erbB-2, is distinct from the c-erbB-1 /epidermal growth factor-receptor gene and is amplified in a human salivary gland adenocarcinoma,” 15 Proc. Natl. Acad. Sci. U.S.A 82, 6497-6501, 1985.
Servenius et al., “Class II Genes of the Human Major Histocompatibility Complex, THE DOBeta Gene is a Divergent Member of the Class II P Gene Family,” (1987) J. Biol. Chem. 262:8759-8766.
Shamis et al., “Bioactivation of Self-Immolative Dendritic Prodrugs by Catalytic Antibody 38C2,” (2004) J . Am. Chem. Soc. 126:1726-1731.
Sheikh F., et al., “Cutting Edge: IL-26 Signals through a Novel Receptor Complex Composed of IL-20 Receptor 1 and IL-10 Receptor 21,” (2004) J.Immunol, 172, 2006-2010.
Shen et al., “Conjugation site modulates the in vivo stability and therapeutic activity of antibody-drug conjugates,” (2012) Nature Biotech., 30(2):184-191.
Shimizu, K et al., “Prothracarcin, a Novel Antitumor Antibiotic,” J. Antibiotics, 35, 972-978 (1982).
Sinha S.K., et al., “Characterization of the EBV/C3d Receptor on the Human Jurkat T Cell Line: Evidence fora Novel Transcript,” (1993) J. Immunol. 150, 5311-5320.
Smellie, M. et al., “Cellular pharmacology of novel C8-linked anthramycin-based sequence-selective DNA minor groove cross-linking agents,” Br. J. Cancer (1994) 70:48-53.
Smellie, M. et al., “Sequence selective recognition of duplex DNA through covalent interstrand cross-linking,” Biochem. (2003) 42:8232-8239.
Smith, P. K. et al., “Measurement of protein using bicinchoninic acid.” Anal Biochem. Oct. 1985; 150(1):76-85.
Souillac, P. et al., “Characterization of delivery systems, differential scanning calorimetry,” Encyclopedia of Controlled Drug Delivery (1999) 212-227 (pp. 217-218).
Storm et al., “Effect of Small Changes in Orientation on Reaction Rate,” (1972) J. Amer. Chem. Soc. 94:5815-5825.
Strausberg et al., “Generation and initial analysis of more than 15,000 full-length human and mouse eDNA sequences,” (2002) Proc. Natl. Acad. Sci USA 99:16899-16903.
Suggitt, M., “The hollow fibre model—facilitating anti-cancer pre-clinical pharmacodynamics and improving animal welfare,” Int. J. Oncol. (2006) 29(6):1493-1499.
Suggs, J.W. et al., “Synthesis and structure of anthramycin analogs via hydride reduction of dilactams,” Tetrahedron Letters, 26, No. 40, 4871-4874 (1985).
Sun et al., “Enabling ScFvs as Multi-Drug Carriers: A Dendritic Approach,” (2003) Bioorganic & Medicinal Chemistry 11:1761-1768.
Sun et al., “Syntheses of Dendritic Linkers Containing Chlorambucil Residues for the Preparation of Antibody-Multidrug Immunoconjugates,” (2002) Bioorganic & Medicinal Chemistry Letters 12:2213-2215.
Sutherland, M.S.K. et al., “SGN-CD33A: a novel CD33-directed antibody-drug conjugate, utilizing pyrrolobenzodiazepine dimers, demonstrates preclinical anti-tumor activity against multi-drug resistant human AML,” American Society of Hematology (Dec. 8-12, 2012) Atlanta, Georgia, Abstract No. 3589.
Svensson P.J., et al., “Phenotypic variation in a family with mutations in two Hirschsprung-related genes (RET and endothelin receptor B),” Hum. Genet. 103, 145-148, 1998.
Swiercz J.M., et al., “Plexin-81 /RhoGEF-mediated Rho A activation involves the receptor tyrosine kinase ErbB-2,” J. Cell Biol. 165, 869-880, 2004.
Syrigos and Epenetos, “Antibody Directed Enzyme Prodrug Therapy (ADEPT): A Review of the Experimental and Clinical Considerations,” (1999) Anticancer Research 19:605-614.
Takeuchi, T. et al., “Neothramycins A and B, New Antitumor Antibiotics,” J. Antibiotics, 29, 93-96 (1976).
Tawaragi Y., et al., “Primary Structure of Nonspecific Crossreacting Antigen (NCA), a Member of Carcinoembryonic Antigen (CEA) Gene Family, Deduced From cDNA SEQUENCE,” Biochem. Biophys. Res. Commun. 150, 89-96, 1988.
Tercel, M. et al., “Unsymmetrical DNA cross-linking agents: combination of the CBI and PBD pharmacophores,” J. Med. Chem. (2003) 46:2132-2151.
Thompson J.S., et al., “BAFF-R, a Newly Identified TNF Receptor That Specifically Interacts with BAFF,” Science 293 (5537), 2108-2111 (2001 ).
Thurston, D. E., “Advances in the study of Pyrrolo[2,1-c][1,4] benzodiazepine (PBD) Antitumour Antibiotics”, Molecular Aspects of Anticancer Drug-DNA Interaction, Neidle, S. and Waring, M.J., Eds.; Macmillan Press Ltd, 1:54-88 (1993).
Thurston, D.E. and Bose, D.S., “Synthesis of DNA-lnteractive Pyrrolo[2,1-c][1,4]benzodiazepines,” Chem. Rev., 94:433-465 (1994).
Thurston, D.E. and Thompson, A.S., “The molecular recognition of DNA,” Chem. Brit., 26, 767-772 (1990).
Thurston, D.E. et al., “Effect of A-ring modifications on the DNA-binding behavior and cytotoxicity of pyrrolo[2,1-c][1,4]benzodiazepines”, Journal of Medicinal Chemistry, 42:1951-1964 (1999).
Thurston, D.E. et al., “Synthesis of Sequence-selective C8-linked Pyrrolo [2,1-c][1,4] Benzodiazepine DNA Interstrand Cross-linking Agent,” J. Org. Chem., 61:8141 -8147 (1996).
Thurston, D.E et al., “Synthesis of a novel GC-specific covalent-binding DNA affinity-cleavage agent based on pyrrolobenzodiazepines (PBDs),” Chemical Communications, 563-565 (1996).
Thurston, D.E., “Nucleic acid targeting: therapeutic strategies for the 21st century,” Brit. J. Cancer (1999) 80(1):65-85.
Tiberghien, A.C. et al., “Application of the stille coupling reaction to the synthesis of C2-substituted endo-exo unsaturated pyrrolo[2,1-c][1,4]benzodiazepines (PBDs),” Biorg. Med. Chem. Lett. (2004) 14:5041-5044.
Tiberghien, A.C., “Application of the stille coupling reaction to the synthesis of C2-substituted endo-exo unsaturated pyrrolo[2,1-c][1,4]benzodiazepines (PBDs),” Bioorg. Med. Chem. Lett. (2008) 18(6):2073-2077.
Toki et al., “Protease-Mediated Fragmentation of p-Amidobenzyl Ethers: A New Strategy for the Activation of Anticancer Prodrugs,” (2002) J. Org. Chem. 67:1866-1872.
Tonnelle et al., “DO Beta a new chain gene in HLA-D with a distinct regulation of expression,” (1985) EMBO J. 4(11):2839-2847.
Touchman et al., “The Genomic Region Encompassing the Nephropathic Cystinosis Gene (CTNS): Complete Sequencing of a 200-kb Segment and Discovery of a Novel Gene within the Common Cystinosis-Causing Deletion,” (2000) Genome Res. 10:165-173.
Tozuka et al., “Studies on tomaymycin. II. Total synthesis of the antitumor antibiotics, E-and Z-tomaymycins,” J. Antibiotics (Tokyo) (1983) 36:276-282.
Trail et al., “Monoclonal antibody drug immunoconjugates for targeted treatment of cancer,” (2003) Cancer Immunol. Immunother. 52:328-337.
Tsunakawa, M. et al., “Porothramycin, a new antibiotic of the anthramycin group: Production, isolation, structure and biological activity,” J. Antibiotics, 41:1366-1373 (1988).
Tsutsumi M., et al., “Novel endothelin B receptor transcripts with the potential of generating a new receptor,” Gene 228, 43-49, 1999.
Uchida et al., “A Novel Epidermal Growth Factor-like Molecule Containing Two Follistatin Modules Stimulates Tyrosine Phosphorylation of erbB-4 in MKN28 Gastric Cancer Cells,” (1999) Biochem. Biophys. Res. Commun. 266:593-602.
Umezawa, H. et al., “Mazethramycins,” SciFinder Scholar, 2-3 (2002).
Umezawa, H. et al., Chemical Abstract No. 4427a, “Mazethramycins” Chemical Abstracts, vol. 90, No. 1, 428 (1979).
Verheij J.B., et al., “ABCD Syndrome is Caused by a Homozygous Mutation in the EDNRB Gene,” Am. J. Med. 15 Genet. 108, 223-225, 2002.
Vippagunta, S.R. et al., “Crystalline solids,” Adv. Drug Delivery Rev. (2001) 48:3-26.
Von Hoegen et al., “Identification of a Human Protein Homologous to the Mouse Lyb-2 B Cell Differentiation Antigen and Sequence of the Corresponding cDNA,” (1990) J. Immunol. 144(12):4870-4877.
Wang, J.H., “Determination of antitumor agent AJG-136 in human serum by HPLC with tandem mass spectrometric detection (HPLC-MS/MS),” Abstracts of Papers American Chemical Society (Mar. 13, 2005) 229(1):U119.
Webster et al., “Mammary tumorigenesis and metastasis in transgenic mice,” (1994) Semin. Cancer Biol. 5:69-76.
Weidner-Wells, M.A. et al., “Photochemical approach to the synthesis of the pyrrolo[1,4]benzodiazepine antibiotics,” J. Org. Chem. (1989) 54:5746-5758.
Weis J.J., et al., “Identification of a partial eDNA clone for the C3d/Epstein-Barr virus receptor of human B lymphocytes: Homology with the receptor for fragments C3b and C4b of the third and fourth components of complement,” Proc. Natl. Acad. Sci. U.S.A. 83, 5639-5643, 1986.
Weis J.J., et al., “Structure of the Human B Lymphocyte Receptor for C3d and the Epstein-Barr Virus and Relatedness to Other Members of the Family of C3/C4 Binding Proteins,” J. Exp. Med. 167, 1047-1066, 1988.
Wells, G. et al., “Design, synthesis and biophysical and biological evaluation of a series of pyrrolobenzodiazepine-poly(N-methylpyrrole) conjugates,” J. Med. Chem. (2006) 49:5442-5461.
Wikipedia, “How many types of cancer are there?”, 2012, 3 pages; http://wiki.answers.com/Q/How-many-different-types_of_cancer_are_there.
Wikipedia, “Management of Cancer,” 2012, 1 page; http://en.wikipedia.org/wiki/Management of cancer.
Wilkinson “Eph Receptors and Ephrins: Regulators of Guidance and Assembly,” Int. Rev. Cytol. 196:177-244 (2000).
Wilkinson, G.P., “Pharmacokinetics and intracellular pharmacological characteristics of the novel pyrrolobenzodiazepine (PBD) dimer SJG-136,” Proceedings of the American Association for Cancer Research Annual Meeting (Jul. 2003) 44:320.
Wilkinson, G.P., “Pharmacokinetics, metabolism and glutathione reactivity of SJG-136,” Br. J. Cancer (2003) 88(Supp. 1):S29.
Wilkinson, G.P., “Preliminary pharmacokinetic and bioanalytical studies of SJG-136 (NSC 694501), a sequence-selective pyrrolobenzodiazepine dimer DNA-cross-linking agent,” Investigational New Drugs (2004) 22(3):231-240.
Wilson et al., “eDNA Cloning of the B Cell Membrane Protein CD22: A Mediator of B-B Cell Interactions,” (1991) J. Exp. Med. 173:137-146.
Wilson, S.C. et al., “Design and Synthesis of a Novel Epoxide-Containing Pyrrolo[2,1-c][1,4]benzodiazepine (PBD) via a New Cyclization Procedure,” Tetrahedron Letters, 36, No. 35, 6333-6336 (1995).
Wilson, S.C. et al., “Design, Synthesis, and Evaluation of a Novel Sequence-Selective Epoxide-Containing DNA Cross-Linking Agent Based on the Pyrrolo[2,1-c][1,4]benzodiazepine System”, J. Med. Chem. 42: 4028-4041 (1999).
Wolff, M.E., Burger's Medicinal Chemistry, 4th Edition, Part I, Wiley: New York (1979) 336-337.
Wolff, M.E., Burger's Medicinal Chemistry, 5th Edition, Part I, John Wiley & Sons (1995) 975-977.
Workman, P. et al., “United Kingdom Co-ordinating Committee on Cancer Research (UKCCCR) guidelines for the welfare of animals in experimental neoplasia (second edition),” Br. J. Cancer (1998) 77(1):1-10.
Wu et al., “Arming antibodies: prospects and challenges for immunoconjugates,” (2005) Nature Biotech. 23(9):1137-1145.
Xie et al., “In vivo behaviour of antibody-drug conjugates for the targeted treatment of cancer,” (2006) Expert. Opin Biol. Ther. 6(3):281-291.
Xie, G. et al., “Bisindolylmaleimides linked to DNA minor groove binding lexitropsins: synthesis, inhibitory activity against topoisomerasel, and biological evaluation,” J. Med. Chem. (1996) 39:1049-1055.
Xu et al., “Molecular Cloning, Functional Characterization, Tissue Distribution, and Chromosomal Localization of a Human, Small Intestinal Sodium-Phosphate (Na + -Pi) Transporter (SLC34A2),” Genomics 62 (2):281-284 (1999).
Xu, M.J., et al., “Molecular Cloning and Characterization of SPAP1, an Inhibitory Receptor,” (2001) Biochem. Biophys. Res. Commun. 280 (3):768-775.
Xu, X.Z., et al., “Regulation of melastatin, a TRP-related protein, through interaction with a cytoplasmic isoform,” Proc. Natl. Acad. Sci. U.S.A. 98 (19):10692-10697 (2001).
Yamaguchi, N., et al., “A Novel Cytokine Exhibiting Megakaryocyte Potentiating Activity From a Human Pancreatic Tumor Cell Line HPC-Y5,” Biol. Chem. 269 (2), 805-808 (1994).
Yamamoto T., et al., “Similarity of protein encoded by the human c-erb-B-2 gene to epidermal growth factor receptor,” Nature 319, 230-234, 1986.
Yang et al., “Murine Six-Transmembrane Epithelial Antigen of the Prostate, Prostate Stem Cell Antigen, and Prostate-specific Membrane Antigen: Prostate-specific Cell-Surface Antigens Highly Expressed in Prostate Cancer of Transgenic Adenocarcinoma Mouse Prostate Mice,” Cancer Research, 61, 5857-5860. Aug. 1, 2001.
Yin & Lloyd, “Molecular Cloning of the CA125 Ovarian Cancer Antigen,” J. Biol. Chem. 276 (29):27371-27375 (2001).
Younes et al., “Phase I multidose-escalation study of the anti-CD19 maytansinoid immunoconjugate SAR3419 administered by intravenous infusion every 3 weeks to patients with relapsed/refractory B-cell lymphoma,” J Clin Oncol., 2012, 30(22):2776-82.
Yu et al., “Human mb-1 Gene: Complete eDNA Sequence and its Expression in B Cells Bearing Membrane Ig of Various Isotypes,” (1992) J. Immunol. 148(2) 633-637.
Zammarchi et al., “Pre-Clinical Development of Adct-402, a Novel Pyrrolobenzodiazepine (PBD)—Based Antibody Drug Conjugate (ADC) Targeting CD19-Expressing B-Cell Malignancies,” Blood, 2015, 126:1564, Abstract.
Related Publications (1)
Number Date Country
20210187117 A1 Jun 2021 US