COMMON LIGHT CHAINS AND METHODS OF USE

- Hoffmann-La Roche Inc.

The present invention generally relates to novel bispecific antigen binding molecules for T cell activation and re-direction to specific target cells comprising a common light chain. In addition, the present invention relates to polynucleotides encoding such bispecific antigen binding molecules, and vectors and host cells comprising such polynucleotides. The invention further relates to methods for producing the bispecific antigen binding molecules of the invention, and to methods of using these bispecific antigen binding molecules in the treatment of disease.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS REFERENCE TO RELATED APPLICATIONS

This application is a continuation of International Application No. PCT/EP2015/076745, Publication No. WO2016/079081, filed Nov. 17, 2015, which claims priority to European Patent Application No. 14194097.3 filed Nov. 20, 2014, the disclosures of which are incorporated herein by reference in their entirety.

SEQUENCE LISTING

The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on May 18, 2017, is named P32398US_ST25.txt and is 537,716 bytes in size.

FIELD OF THE INVENTION

The present invention generally relates to bispecific antigen binding molecules for activating T cells. In addition, the present invention relates to polynucleotides encoding such bispecific antigen binding molecules, and vectors and host cells comprising such polynucleotides. The invention further relates to methods for producing the bispecific antigen binding molecules of the invention, and to methods of using these bispecific antigen binding molecules in the treatment of disease.

BACKGROUND

The selective destruction of an individual cell or a specific cell type is often desirable in a variety of clinical settings. For example, it is a primary goal of cancer therapy to specifically destroy tumor cells, while leaving healthy cells and tissues intact and undamaged.

An attractive way of achieving this is by inducing an immune response against the tumor, to make immune effector cells such as natural killer (NK) cells or cytotoxic T lymphocytes (CTLs) attack and destroy tumor cells. CTLs constitute the most potent effector cells of the immune system, however they cannot be activated by the effector mechanism mediated by the Fc domain of conventional therapeutic antibodies.

In this regard, bispecific antibodies designed to bind with one “arm” to a surface antigen on target cells, and with the second “arm” to an activating, invariant component of the T cell receptor (TCR) complex, have become of interest in recent years. The simultaneous binding of such an antibody to both of its targets forces a temporary interaction between target cell and T cell, causing activation of any cytotoxic T cell and subsequent lysis of the target cell. Hence, the immune response is re-directed to the target cells and is independent of peptide antigen presentation by the target cell or the specificity of the T cell as would be relevant for normal MHC-restricted activation of CTLs. In this context it is crucial that CTLs are only activated when the bispecific antibody binds to a target cell and the CTL, i.e. the immunological synapse is mimicked. Particularly desirable are bispecific antibodies that do not require lymphocyte preconditioning or co-stimulation in order to elicit efficient lysis of target cells.

Several bispecific antibody formats have been developed and their suitability for T cell mediated immunotherapy investigated. Out of these, the so-called BiTE (bispecific T cell engager) molecules have been very well characterized and already shown some promise in the clinic (reviewed in Nagorsen and Bäuerle, Exp Cell Res 317, 1255-1260 (2011)). BiTEs are tandem scFv molecules wherein two scFv molecules are fused by a flexible linker. Further bispecific formats being evaluated for T cell engagement include diabodies (Holliger et al., Prot Eng 9, 299-305 (1996)) and derivatives thereof, such as tandem diabodies (Kipriyanov et al., J Mol Biol 293, 41-66 (1999)). A more recent development are the so-called DART (dual affinity retargeting) molecules, which are based on the diabody format but feature a C-terminal disulfide bridge for additional stabilization (Moore et al., Blood 117, 4542-51 (2011)). The so-called triomabs, which are whole hybrid mouse/rat IgG molecules and also currently being evaluated in clinical trials, represent a larger sized format (reviewed in Seimetz et al., Cancer Treat Rev 36, 458-467 (2010)).

The variety of formats that are being developed shows the great potential attributed to T cell re-direction and activation in immunotherapy. The task of generating bispecific antibodies suitable therefor is, however, by no means trivial, but involves a number of challenges that have to be met related to efficacy, toxicity, applicability and produceability of the antibodies.

Small constructs such as, for example, BiTE molecules—while being able to efficiently crosslink effector and target cells—have a very short serum half life requiring them to be administered to patients by continuous infusion. IgG-like formats on the other hand—while having the great benefit of a long half life—suffer from toxicity associated with the native effector functions inherent to IgG molecules. Their immunogenic potential constitutes another unfavorable feature of IgG-like bispecific antibodies, especially non-human formats, for successful therapeutic development. Finally, a major challenge in the general development of bispecific antibodies has been the production of bispecific antibody constructs at a clinically sufficient quantity and purity, due to the mispairing of antibody heavy and light chains of different specificities upon co-expression, which decreases the yield of the correctly assembled construct and results in a number of non-functional side products from which the desired bispecific antibody may be difficult to separate.

Given the difficulties and disadvantages associated with currently available bispecific antibodies for T cell mediated immunotherapy, there remains a need for novel, improved formats of such molecules.

SUMMARY OF THE INVENTION

In a first aspect the present invention provides a T cell activating bispecific antigen binding molecule comprising a first and a second antigen binding moiety, wherein the first antigen binding moiety comprises a first light chain and wherein the first antigen binding moiety is capable of specific binding to an activating T cell antigen and the second antigen binding moiety comprises a second light chain and wherein the second antigen binding moiety is capable of specific binding to a target cell antigen, wherein the amino acid sequence of the first and the second light chain is identical. In one embodiment, the first antigen binding moiety is a Fab. In one embodiment, the second antigen binding moiety is a Fab. In one embodiment, the first and the second antigen binding moiety is a Fab.

In one aspect the invention provides a T cell activating bispecific antigen binding molecule comprising a first and a second antigen binding moiety, one of which is a Fab molecule capable of specific binding to an activating T cell antigen and the other one of which is a Fab molecule capable of specific binding to a target cell antigen, wherein the first and the second Fab molecule have identical VLCL light chains.

In one embodiment said T cell activating bispecific antigen binding molecule further comprises an Fc domain composed of a first and a second subunit capable of stable association.

In one embodiment said T cell activating bispecific antigen binding molecule comprises a light chain comprising the light chain CDRs of SEQ ID NO: 32, SEQ ID NO: 33 and SEQ ID NO: 34.

In one embodiment said T cell activating bispecific antigen binding molecule comprises a light chain comprising SEQ ID NO: 31.

In one embodiment said Fab molecule capable of specific binding to an activating T cell antigen comprises a heavy chain comprising the heavy chain CDR of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39.

In a particular embodiment, not more than one antigen binding moiety capable of specific binding to an activating T cell antigen is present in the T cell activating bispecific antigen binding molecule (i.e. the T cell activating bispecific antigen binding molecule provides monovalent binding to the activating T cell antigen).

In some embodiments, the first and the second antigen binding moiety of the T cell activating bispecific antigen binding molecule are fused to each other, optionally via a peptide linker. In one such embodiment, the second antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety. In another such embodiment, the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding moiety. In yet another such embodiment, the second antigen binding moiety is fused at the C-terminus of the Fab light chain to the N-terminus of the Fab light chain of the first antigen binding moiety.

In yet another such embodiment, the first antigen binding moiety is fused at the C-terminus of the Fab light chain to the N-terminus of the Fab light chain of the second antigen binding moiety.

In one embodiment, the first antigen binding moiety of the T cell activating bispecific antigen binding molecule is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or the second subunit of the Fc domain.

In one embodiment, the second antigen binding moiety of the T cell activating bispecific antigen binding molecule is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or the second subunit of the Fc domain.

In another embodiment, the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or second subunit of the Fc domain.

In one embodiment, the first and the second antigen binding moiety of the T cell activating bispecific antigen binding molecule are each fused at the C-terminus of the Fab heavy chain to the N-terminus of one of the subunits of the Fc domain.

In certain embodiments, the T cell activating bispecific antigen binding molecule comprises a third antigen binding moiety which is a Fab molecule capable of specific binding to a target cell antigen. In one embodiment said third antigen binding moiety is a Fab molecule comprising an identical VLCL light chain as the first and the second antigen binding moiety.

In one such embodiment the first, second and third antigen binding moiety are each a Fab molecule comprising the light chain CDRs of SEQ ID NO: 32, SEQ ID NO: 33 and SEQ ID NO: 34.

In one such embodiment the first, second and third antigen binding moiety are each a Fab molecule comprising a light chain comprising SEQ ID NO: 31.

In one embodiment, the third antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or second subunit of the Fc domain. In a particular embodiment, the second and the third antigen binding moiety of the T cell activating antigen binding molecule are each fused at the C-terminus of the Fab heavy chain to the N-terminus of one of the subunits of the Fc domain, and the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding moiety. In another particular embodiment, the first and the third antigen binding moiety of the T cell activating antigen binding molecule are each fused at the C-terminus of the Fab heavy chain to the N-terminus of one of the subunits of the Fc domain, and the second antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety. The components of the T cell activating bispecific antigen binding molecule may be fused directly or through suitable peptide linkers. In one embodiment the second and the third antigen binding moiety and the Fc domain are part of an immunoglobulin molecule. In one embodiment the first and the third antigen binding moiety and the Fc domain are part of an immunoglobulin molecule. In a particular embodiment the immunoglobulin molecule is an IgG class immunoglobulin. In an even more particular embodiment the immunoglobulin is an IgG1 subclass immunoglobulin. In another embodiment, the immunoglobulin is an IgG4 subclass immunoglobulin.

In a particular embodiment, the Fc domain is an IgG Fc domain. In a specific embodiment, the Fc domain is an IgG1 Fc domain. In another specific embodiment, the Fc domain is an IgG4 Fc domain. In an even more specific embodiment, the Fc domain is an IgG4 Fc domain comprising the amino acid substitution S228P. In an even more specific embodiment, the Fc domain is an IgG4 Fc domain comprising the amino acid substitutions L235E and S228P (SPLE). In particular embodiments the Fc domain is a human Fc domain.

In particular embodiments the Fc domain comprises a modification promoting the association of the first and the second Fc domain subunit. In a specific such embodiment, an amino acid residue in the CH3 domain of the first subunit of the Fc domain is replaced with an amino acid residue having a larger side chain volume, thereby generating a protuberance within the CH3 domain of the first subunit which is positionable in a cavity within the CH3 domain of the second subunit, and an amino acid residue in the CH3 domain of the second subunit of the Fc domain is replaced with an amino acid residue having a smaller side chain volume, thereby generating a cavity within the CH3 domain of the second subunit within which the protuberance within the CH3 domain of the first subunit is positionable.

In a particular embodiment the Fc domain exhibits reduced binding affinity to an Fc receptor and/or reduced effector function, as compared to a native IgG1 Fc domain. In certain embodiments the Fc domain is engineered to have reduced binding affinity to an Fc receptor and/or reduced effector function, as compared to a non-engineered Fc domain. In one embodiment, the Fc domain comprises one or more amino acid substitution that reduces binding to an Fc receptor and/or effector function. In one embodiment, the one or more amino acid substitution in the Fc domain that reduces binding to an Fc receptor and/or effector function is at one or more position selected from the group of L234, L235, and P329 (Kabat numbering). In particular embodiments, each subunit of the Fc domain comprises three amino acid substitutions that reduce binding to an Fc receptor and/or effector function wherein said amino acid substitutions are L234A, L235A and P329G. In one such embodiment, the Fc domain is an IgG1 Fc domain, particularly a human IgG1 Fc domain. In other embodiments, each subunit of the Fc domain comprises two amino acid substitutions that reduce binding to an Fc receptor and/or effector function wherein said amino acid substitutions are L235E and P329G. In one such embodiment, the Fc domain is an IgG4 Fc domain, particularly a human IgG4 Fc domain.

In one embodiment the Fc receptor is an Fcγ receptor. In one embodiment the Fc receptor is a human Fc receptor. In one embodiment, the Fc receptor is an activating Fc receptor. In a specific embodiment, the Fc receptor is human FcγRIIa, FcγRI, and/or FcγRIIIa. In one embodiment, the effector function is antibody-dependent cell-mediated cytotoxicity (ADCC).

In a particular embodiment, the activating T cell antigen that the bispecific antigen binding molecule is capable of binding is CD3. In other embodiments, the target cell antigen that the bispecific antigen binding molecule is capable of binding is a tumor cell antigen. In one embodiment, the target cell antigen is selected from the group consisting of: Folate Receptor 1 (FolR1), Mucin-1 (MUC1), and B Cell Maturation Antigen (BCMA). In one specific embodiment, the target cell antigen is not BCMA.

In another aspect, the invention provides for a light chain comprising the amino acid sequence of SEQ ID NO: 32, SEQ ID NO: 33 and SEQ ID NO: 34 for use in a T cell activating bispecific antigen binding molecule. In one embodiment, the light chain comprises the amino acid sequence of SEQ ID NO: 31. In one embodiment, the light chain comprises the amino acid sequence of SEQ ID NO: 35.

In another aspect, the invention provides for a light chain comprising the amino acid sequence of SEQ ID NO: 32, SEQ ID NO: 33 and SEQ ID NO: 34 for use in a library for production of T cell activating bispecific antigen binding molecule. In one embodiment, the light chain comprises the amino acid sequence of SEQ ID NO: 31. In one embodiment, the light chain comprises the amino acid sequence of SEQ ID NO: 35.

In another aspect, the invention provides for an isolated polypeptide comprising the amino acid sequence of SEQ ID NO: 31.

In another aspect, the invention provides for an isolated polypeptide comprising the amino acid sequence of SEQ ID NO: 35.

According to another aspect of the invention there is provided an isolated polynucleotide encoding a T cell activating bispecific antigen binding molecule of the invention or a fragment thereof. The invention also encompasses polypeptides encoded by the polynucleotides of the invention. The invention further provides an expression vector comprising the isolated polynucleotide of the invention, and a host cell comprising the isolated polynucleotide or the expression vector of the invention. In some embodiments the host cell is a eukaryotic cell, particularly a mammalian cell.

In another aspect is provided a method of producing the T cell activating bispecific antigen binding molecule of the invention, comprising the steps of a) culturing the host cell of the invention under conditions suitable for the expression of the T cell activating bispecific antigen binding molecule and b) recovering the T cell activating bispecific antigen binding molecule. The invention also encompasses a T cell activating bispecific antigen binding molecule produced by the method of the invention.

The invention further provides a pharmaceutical composition comprising the T cell activating bispecific antigen binding molecule of the invention and a pharmaceutically acceptable carrier. Also encompassed by the invention are methods of using the T cell activating bispecific antigen binding molecule and pharmaceutical composition of the invention. In one aspect the invention provides a T cell activating bispecific antigen binding molecule or a pharmaceutical composition of the invention for use as a medicament. In one aspect is provided a T cell activating bispecific antigen binding molecule or a pharmaceutical composition according to the invention for use in the treatment of a disease in an individual in need thereof. In a specific embodiment the disease is cancer.

Also provided is the use of a T cell activating bispecific antigen binding molecule of the invention for the manufacture of a medicament for the treatment of a disease in an individual in need thereof; as well as a method of treating a disease in an individual, comprising administering to said individual a therapeutically effective amount of a composition comprising the T cell activating bispecific antigen binding molecule according to the invention in a pharmaceutically acceptable form. In a specific embodiment the disease is cancer. In any of the above embodiments the individual preferably is a mammal, particularly a human.

The invention also provides a method for inducing lysis of a target cell, particularly a tumor cell, comprising contacting a target cell with a T cell activating bispecific antigen binding molecule of the invention in the presence of a T cell, particularly a cytotoxic T cell.

In another aspect, the invention provides for a method for identifying a variable heavy chain for use in a bispecific antigen binding molecule specific for a T cell activation antigen and a target cell antigen, comprising the step of screening a combinatorial library comprising variable heavy chains with a light chain comprising the amino acid sequence of SEQ ID NO: 32, SEQ ID NO: 33 and SEQ ID NO: 34. In one embodiment, the light chain comprises the amino acid sequence of SEQ ID NO: 31. In one embodiment, the light chain comprises the amino acid sequence of SEQ ID NO: 35.

BRIEF DESCRIPTION OF THE DRAWINGS

FIGS. 1A-I illustrate exemplary configurations of the T cell activating bispecific antigen binding molecules (TCBs) disclosed herein. All constructs except the kappa-lambda format in (FIG. 1I) have P329G LALA mutations and comprise knob-into-hole Fc fragments with knob-into-hole modifications. (FIG. 1A) Illustration of the “FolR1 TCB 2+1 inverted (common light chain)”. The FolR1 binder is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first subunit of the Fc domain comprising the knob modification. These constructs are not crossed and have three times the same VLCL light chain. (FIG. 1B) Illustration of the “FolR1 TCB 1+1 head-to-tail (common light chain)”. These constructs are not crossed and have two times the same VLCL light chain. (FIG. 1C) Illustration of the “FolR1 TCB 1+1 classical (common light chain)”. These constructs are not crossed and have two times the same VLCL light chain. (FIG. 1D) Illustration of the “FolR1 TCB 2+1 classical (common light chain)”. The CD3 binder is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first subunit of the Fc domain comprising the knob modification. These constructs are not crossed and have three times the same VLCL light chain. (FIG. 1E) Illustration of the “FolR1 TCB 2+1 crossfab classical”. These constructs comprise a Ck-VH chain for the CD3 binder instead of the conventional CH1-VH chain. The CD3 binder is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first subunit of the Fc domain comprising the knob modification. (FIG. 1F) Illustration of the “FolR1 TCB 2+1 crossfab inverted”. These constructs comprise a Ck-VH chain for the CD3 binder instead of the conventional CH1-VH chain. The FolR1 binder is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first subunit of the Fc domain comprising the knob modification. (FIG. 1G) Illustration of the “FolR1 TCB 1+1 crossfab head-to-tail”. These constructs comprise a Ck-VH chain for the CD3 binder instead of the conventional CH1-VH chain. (FIG. 1H) Illustration of the “FolR1 TCB 1+1 crossfab classical”. These constructs comprise a Ck-VH chain for the CD3 binder instead of the conventional CH1-VH chain. FIG. 11 illustrates the CD3/FolR1 kappa-lambda antibody format. These constructs comprise a crossed common light chain VLCH1 and one crossed VHCL chain specific for CD3 and one crossed VHCL chain specific for FolR1.

FIGS. 2A-C depict graphs summarizing Binding of FoLR1 IgG binders to HeLa cells. Binding of newly generated FolR1 binders to FolR1 expressed on HeLa cells were determined by flow cytometry. Bound antibodies were detected with a fluorescently labeled anti-human secondary antibody.

FIGS. 3A-B depict graphs summarizing specificity of FolR1 binders for FolR1. Binding of FolR1 IgGs to HEK cells transiently transfected with either FolR1 or FolR2 was analyzed by flow cytometry to identify clones which bind specifically to FolR1 and not to FolR2. The antibodies were detected with a fluorescently labeled anti-human secondary antibody.

FIGS. 4A-B depict graphs summarizing cross-reactivity of FolR1 binders to cyFoLR1. Cross-reactivity of the FolR1 antibodies to cyno FolR1 was addressed on HEK cells transiently transfected with cyFolR1 by flow cytometry. The antibodies were detected with a fluorescently labeled anti-human secondary antibody.

FIG. 5 depicts a graph illustrating internalization of FolR1 TCBs after binding. Internalization of the four FolR1 TCBs after binding to FolR1 was tested on HeLa cells. Remaining FolR1 TCBs on the surface were detected with a fluorescently labeled anti-human secondary antibody after indicated time points of incubation at 37° C. Percentage of internalization was calculated.

FIGS. 6A-E depict graphs summarizing binding of FolR1 IgGs to cells with different FolR1 expression levels. Binding of 9D11, 16D5 and Mov19 IgG to tumor cells with different FolR1 expression levels was analyzed by flow cytometry. DP47 IgG was included as isotype control and MKN-45 were included as FolR1 negative cell line. The antibodies were detected with a fluorescently labeled anti-human secondary antibody.

FIGS. 7A-L depict graphs summarizing T cell mediated killing of HT-29 and SKOV3 cells. FolR1 TCBs were used to test T cell mediated killing of HT-29 and SKOV3 tumor cells and upregulation of activation marker on T cells upon killing. (FIGS. 7A-D) T cell mediated killing of HT-29 and SKOV3 cells in the presence of 9D11 FolR1 TCB and 16D5 FolR1 TCB was measured by LDH release after 24 h and 48 h. DP47 TCB was included as negative control. After 48 h incubation upregulation of the activation marker CD25 and CD69 on CD8 T cells and CD4 T cells upon killing of SKOV3 (FIGS. 7E-H) or HT-29 (FIG. 71-L) tumor cells was assessed by flow cytometry.

FIG. 8 depicts a graph showing absence of anti-FolR1 binding to erythrocytes. Erythrocytes were gated as CD235a positive population and binding of 9D11 IgG, 16D5 IgG, Mov19 IgG and DP47 IgG to this population was determined by flow cytometry. The antibodies were detected with a fluorescently labeled anti-human secondary antibody.

FIGS. 9A-D depict graphs summarizing activation marker upregulation in whole blood. CD25 and CD69 activation marker upregulation of CD4 T cells and CD8 T cells 24 h after addition of 9D11 FolR1 TCB, 16D5 FolR1 TCB, Mov19 FolR1 TCB and DP47 TCB was analyzed by flow cytometry.

FIG. 10 Binding of 9D11 TCB a-glyco variants to HeLa cells. Binding of 9D11 FolR1 TCB a-glyco variants to Hela cells was compared to binding of the original 9D11 TCB on HeLa cells. The antibodies were detected with a fluorescently labeled anti-human secondary antibody and binding was determined by flow cytometry.

FIGS. 11A-F depict graphs summarizing T cell mediated killing with 9D11 FolR1 TCB a-glyco variants of tumor cells. 9D11 FolR1 TCB a-glyco variants were used to test T cell mediated killing of (FIG. 11A-D) SKOV3, MKN-45 (as FolR1 negative control) and (FIG. 11E-F) HT-29 tumor cells in comparison to killing with the original 9D11 FolR1 TCB. As read-out LDH release after 24 h and 48 h was used.

FIGS. 12A-X depict graphs summarizing T cell mediated killing of primary epithelial cells. Primary epithelial cells with very low levels of FolR1 were used to test T cell mediated killing with 16D5 FolR1 TCB and 9D11 FolR1 TCB, DP47 TCB was included as a negative control and HT29 cells were included as positive control. (FIGS. 12A-H) LDH release of human retinal pigment (HRP), human renal cortical (HRC), human bronchial (HB) and of HT29 cells was determined after 24 h and 48 h. CD25 and CD69 activation marker upregulation on CD4 T cells and CD8 T cells upon killing of (FIGS. 12I-L) HRP, (FIGS. 12M-P) HRC, (FIGS. 12Q-T) HB and (FIGS. 12 U-X) HT29 was determined after 48 h by flow cytometry.

FIGS. 13A-C show a comparison of different TCB formats with 16D5. Four different TCB formats containing the FolR1 binder 16D5 were compared in FIG. 13A binding to HeLa cells, in FIG. 14 B T cell mediated killing of SKOV3 cells after 24 h and 48 h and in FIG. 14C CD25 and CD69 activation marker upregulation on CD4 T cells and CD8 T cells 48 h after killing.

FIGS. 14A-C depict a comparison of different TCB formats with 9D11. Three different TCB formats containing the FolR1 binder 9D11 were compared in A) binding to HeLa cells, in B) T cell mediated killing of SKOV3 cells after 24 h and 48 h and in C) CD25 and CD69 activation marker upregulation on CD4 T cells and CD8 T cells 48 h after killing.

FIG. 15 depicts a PK-profile of FOLR1 TCB in NOG mice for three different doses.

FIG. 16 illustrates an experimental protocol for efficacy study with FOLR1 TCB.

FIGS. 17A-B depict tumor growth curves. (FIG. 17A) Mean values and SEM of tumor volumes in the different treatment groups. (FIG. 17B) Tumor growth of single mice in all treatment groups. TGI (tumor growth inhibition) give the percentage of the Mean tumor volume compared to vehicle group.

FIG. 18 shows tumor weights at study termination.

FIGS. 19A-B show FACS analysis of tumor infiltrating T-cells at study day 32. (FIG. 19A) Tumor single cells suspensions were stained with anti-human CD3/CD4/CD8 and analyzed by flow cytometry. (FIG. 19B) Mean values and SEM of T-cell counts per mg tumor tissue in different treatment groups.

FIGS. 20A-B show FACS analysis for T-cell activation/degranulation and cytokine secretion at study day 32. CD4+ (FIG. 20A) and CD8+ (FIG. 20B) tumor infiltrating T-cells were stained for cytokines, activation and degranulation markers. Displayed are the mean values and SEM of T-cell counts per mg tumor tissue in different treatment groups.

FIGS. 21A-B show percent tumor lysis. SKOV3 cells were incubated with PBMCs in the presence of either kappa lambda FoLR1 TCB or DP47 TCB. After 24 h (FIG. 21A) and 48 h (FIG. 21B) killing of tumor cells was determined by measuring LDH release

FIGS. 22A-D show CD25 and CD69 upregulation on CD4 T cells. SKOV3 cells were incubated with PBMCs in the presence of either kappa lambda FoLR1 TCB or DP47 TCB. After 48 h CD25 and CD69 upregulation on CD4 T cells (FIG. 22A-B) and CD8 T cells (FIG. 22C-D) was measured by flow cytometry.

FIGS. 23A-B show percent tumor lysis. T-cell killing of SKov-3 cells (medium FolR1) induced by 36F2 TCB, Mov19 TCB and 21A5 TCB after 24 h (FIG. 23A) and 48 h (FIG. 23B) of incubation (E:T=10:1, effectors human PBMCs).

FIGS. 24A-C show T-cell killing induced by 36F2 TCB, 16D5 TCB, 16D5 TCB classical, 16D5 TCB 1+1 and 16D5 TCB HT of Hela (high FolR1) (FIG. 24A), Skov-3 (medium FolR1) (FIG. 24B) and HT-29 (low FolR1) (FIG. 24C) human tumor cells (E:T=10:1, effectors human PBMCs, incubation time 24 h). DP47 TCB was included as non-binding control.

FIGS. 25A-C show upregulation of CD25 and CD69 on human CD8+ (FIG. 25A, B) and CD4+ (FIG. 25C), T cells after T cell-mediated killing of Hela cells (high FolR1) (FIG. 25A), SKov-3 cells (medium FolR1) (FIG. 25B) and HT-29 cells (low FolR1) (FIG. 25C) (E:T=10:1, 48 h incubation) induced by 36F2 TCB, 16D5 TCB and DP47 TCB (non-binding control).

FIGS. 26A-F show T-cell killing induced by 36F2 TCB, 16D5 TCB and DP47 TCB of human Renal Cortical Epithelial Cells (FIG. 26A, B), human Retinal Pigment Epithelial Cells (FIG. 26C, D) and HT-29 cells (FIG. 26E, F) cells after 24 h (FIG. 26A, C, E) and 48 h (FIG. 26B, D, F) of incubation (E:T=10:1, effectors human PBMCs).

FIG. 27 depicts a table summarizing quantification of FolR1 binding sites on various normal and cancer cells lines.

FIGS. 28A-B show binding of 16D5 TCB and its corresponding CD3 deamidation variants 16D5 TCB N100A and 16D5 TCB S100aA and 9D11 TCB and its demidation variants 9D11 TCB N100A and 9D11 TCB S100aA to human CD3 expressed on Jurkat cells.

FIGS. 29A-B show T-cell killing of SKov-3 (medium FolR1) human tumor cells induced by 16D5 TCB and its corresponding CD3 deamidation variants 16D5 TCB N100A and 16D5 TCB S100aA (FIG. 29A) and 9D11 TCB and its demidation variants 9D11 TCB N100A and 9D11 TCB S100aA (FIG. 29B) (E:T=10:1, effectors human PBMCs, incubation time 24 h). DP47 TCB was included as non-binding control.

FIG. 30A-B show T-cell killing of HT-29 (low FolR1) human tumor cells induced by 16D5 TCB and its corresponding CD3 deamidation variants 16D5 TCB N100A and 16D5 TCB S100aA (FIG. 30A) and 9D11 TCB and its demidation variants 9D11 TCB N100A and 9D11 TCB S100aA (FIG. 30B) (E:T=10:1, effectors human PBMCs, incubation time 24 h). DP47 TCB was included as non-binding control.

FIG. 31 shows a sequence alignment of the VH domains of the 3 identified MUC1-specific binders. All three clones are derivatives of the IGHV3-23 germline (SEQ ID NO: 136). Clone 58D6 (SEQ ID NO: 60) and 110A5 (SEQ ID NO: 64) originate from a library that was only randomized in CDR3, while clone 106D2 (SEQ ID NO: 62) was identified from a library randomized in all 3 CDRs. Positions in CDR1 and 2 that deviate from the germline sequence are printed italic.

FIGS. 32A-B shows results of characterization of CLC binders. (FIG. 32A) SPR analysis. SPR-based kinetic analyses of 3 clones specifically binding to MUC1. Smooth lines represent a global fit of the data to a 1:1 interaction model. (FIG. 32B) Summary of kinetic and thermodynamic parameters.

FIG. 33 depicts a schematic diagram of the generated TCB construct. The CLC TCB construct consists of 3 different immunoglobulin chains: 1) an IgG heavy chain harbouring the “hole mutations” in the Fc part and containing the target-specific VH domain; 2) an Ig chain consisting of the target-specific VH and a CH1 domain, followed by the CD3-specific VH domain and a CH1 domain, followed by the Fc part containing the “knob” mutations; and 3) the common light chain that anneals to both the MUC1-specific and the CD3-specific sequences.

FIGS. 34A-B depicts purification and analytical characterization of the produced MUC1-specific TCBs (FIG. 34A and FIG. 34B). The purification method involved an affinity step (protein A) followed by size exclusion chromatography (Superdex 200, GE Healthcare). The final product was analyzed and characterized by analytical size exclusion chromatography (Superdex 200 column) and by capillary electrophoresis.

FIG. 35 depicts SPR analysis of the MUC1-specific binders in the TCB format. Shown is the binding of 2 MUC1-specific TCBs at different concentrations (see text) to either MUC1 or an unrelated antigen. Smooth lines represent a global fit of the data to a 1:1 interaction model

FIGS. 36A-G depict bispecific bivalent and trivalent antibodies comprising only the Fab fragments (specific to CD3 and BCMA) with or without an Fc part as specified: (A) Fab BCMA-Fc-Fab CD3; (B) Fab BCMA-Fc-Fab CD3-Fab BCMA; (C) Fab BCMA-Fc-Fab BCMA-Fab CD3; (D) Fc-Fab CD3-Fab BCMA; (E) Fc-Fab BCMA-Fab CD3; (F) Fab CD3-Fab BCMA-Fab BCMA; (G) Fab CD3-Fab BCMA. Preferably, the LC of Fab CD3 and Fab BCMA are identical (common LC) to avoid LC mispairing and reduce side-products. Fab CD3 and Fab BCMA are linked to each other with flexible linkers.

FIG. 37 depicts lack of binding of BCMA IgG antibody to TACI receptor as detected by surface plasmon resonance (SPR). Curve 1 corresponds to the signal on reference channel, curve 2 to the channel where the binding occurs (binding channel) and the curve 2−1 is the subtracted signal (binding channel—reference channel), meaning that this is the signal due to the binding event. SPR binding assay clearly demonstrated that pSCHLI372 IgG did not bind to human TACI receptor.

FIGS. 38A-C show production and purification of BCMA-TCB CLC. CE-SDS graphs (non-reduced (top) and reduced (bottom)) of the final protein preparations after Protein A (PA) affinity chromatography and size exclusion chromatographic (SEC) purification steps applied to (A) pSCHLI333-TCB CLC, (B) pSCHLI372-TCB CLC, (C) pSCHLI373-TCB CLC. All three molecules are of molecular format as described in FIG. 36B.

FIGS. 39A-B show binding of BCMA-TCB CLC antibodies on BCMAhi-positive H929 cells by flow cytometry. The median fluorescence intensity of BCMA-TCB CLC antibodies were plotted in function of antibody concentrations (0.12 to 500 nM); (A) pSCHLI372-TCB CLC and pSCHLI373-TCB CLC on H929 cells (A) and MKN45 cells (B). DP47-TCB is a negative control TCB which did not bind to BCMA at concentrations below 100 nM (see Example 7).

FIGS. 40A-B show binding of BCMA-TCB CLC antibodies on CD3-positive Jurkat T cells as measured by flow cytometry. Median fluorescence intensity for BCMA-TCB CLC antibodies (pSCHLI372-TCB CLC and pSCHLI373-TCB CLC) binding to Jurkat T cells and plotted in function of antibody concentration. Non-binding to BCMA-negative and CD3-negative MKN45 cells at concentrations below 100 nM.

FIG. 41 shows T-cell activation mediated by BCMA-TCB CLC antibodies in presence of H929 cells as detected by flow cytometry. Expression level of the early activation marker CD69 and the late activation marker CD25 on CD4+ and CD8+ T cells after 48 hours of incubation. pSCHLI372-TCB CLC and pSCHLI373-TCB CLC antibodies induced an up-regulation of CD69 and CD25 activation markers in a concentration-dependent and specific manner in the presence of BCMA-positive target cells. E:T ratio used as 10 PBMCs:1 H929 cell; cells were incubated for 48 h before measurement of CD69 and CD25 upregulation. DP47-TCB which is a negative control TCB did not induce T-cell activation. Representative results are from two independent experiments.

FIGS. 42A-B show BCMA-TCB CLC antibodies induce T-cell redirected killing of BCMAhi-positive H929 myeloma cells as detected by colorimetric LDH release assay. BCMA-TCB CLC antibodies pSCHLI372-TCB CLC (A, B) and pSCHLI373-TCB CLC (A) induced a concentration-dependent killing of BCMAhi-positive H929 myeloma cells as measured by LDH release. DP47-TCB which is a negative control TCB that does not bind to BCMA but only to CD3 did not induce H929 cell killing. E:T ratio used as 10 PBMCs:1 H929 cell; cells were incubated for 24 h before measurement of LDH release. Representative results are from three independent experiments.

FIGS. 43 shows BCMA-TCB CLC antibodies induce T-cell redirected killing of BCMAmed/lo-positive U266 myeloma cells as detected by colorimetric LDH release assay. BCMA-TCB CLC antibodies pSCHLI372-TCB CLC and pSCHLI373-TCB CLC induced a concentration-dependent killing of BCMAmed/lo-positive U266 myeloma cells as measured by LDH release. DP47-TCB which is a negative control TCB that does not bind to BCMA but only to CD3 did not induce H929 cell killing. E:T ratio used as 10 PBMCs:1 U266 cell; cells were incubated for 24 h before measurement of LDH release. Representative results are from two independent experiments.

DETAILED DESCRIPTION OF THE INVENTION

Definitions

Terms are used herein as generally used in the art, unless otherwise defined in the following. As used herein, the term “antigen binding molecule” refers in its broadest sense to a molecule that specifically binds an antigenic determinant. Examples of antigen binding molecules are immunoglobulins and derivatives, e.g. fragments, thereof.

The term “bispecific” means that the antigen binding molecule is able to specifically bind to at least two distinct antigenic determinants. Typically, a bispecific antigen binding molecule comprises two antigen binding sites, each of which is specific for a different antigenic determinant. In certain embodiments the bispecific antigen binding molecule is capable of simultaneously binding two antigenic determinants, particularly two antigenic determinants expressed on two distinct cells.

The term “valent” as used herein denotes the presence of a specified number of antigen binding sites in an antigen binding molecule. As such, the term “monovalent binding to an antigen” denotes the presence of one (and not more than one) antigen binding site specific for the antigen in the antigen binding molecule.

An “antigen binding site” refers to the site, i.e. one or more amino acid residues, of an antigen binding molecule which provides interaction with the antigen. For example, the antigen binding site of an antibody comprises amino acid residues from the complementarity determining regions (CDRs). A native immunoglobulin molecule typically has two antigen binding sites, a Fab molecule typically has a single antigen binding site.

As used herein, the term “antigen binding moiety” refers to a polypeptide molecule that specifically binds to an antigenic determinant. In one embodiment, an antigen binding moiety is able to direct the entity to which it is attached (e.g. a second antigen binding moiety) to a target site, for example to a specific type of tumor cell or tumor stroma bearing the antigenic determinant. In another embodiment an antigen binding moiety is able to activate signaling through its target antigen, for example a T cell receptor complex antigen. Antigen binding moieties include antibodies and fragments thereof as further defined herein. Particular antigen binding moieties include an antigen binding domain of an antibody, comprising an antibody heavy chain variable region and an antibody light chain variable region. In certain embodiments, the antigen binding moieties may comprise antibody constant regions as further defined herein and known in the art. Useful heavy chain constant regions include any of the five isotypes: α, δ, ε, γ, or μ. Useful light chain constant regions include any of the two isotypes: κ and λ.

As used herein, the term “antigenic determinant” is synonymous with “antigen” and “epitope,” and refers to a site (e.g. a contiguous stretch of amino acids or a conformational configuration made up of different regions of non-contiguous amino acids) on a polypeptide macromolecule to which an antigen binding moiety binds, forming an antigen binding moiety-antigen complex. Useful antigenic determinants can be found, for example, on the surfaces of tumor cells, on the surfaces of virus-infected cells, on the surfaces of other diseased cells, on the surface of immune cells, free in blood serum, and/or in the extracellular matrix (ECM). The proteins referred to as antigens herein (e.g. MCSP, FAP, CEA, EGFR, CD33, CD3) can be any native form the proteins from any vertebrate source, including mammals such as primates (e.g. humans) and rodents (e.g. mice and rats), unless otherwise indicated. In a particular embodiment the antigen is a human protein. Where reference is made to a specific protein herein, the term encompasses the “full-length”, unprocessed protein as well as any form of the protein that results from processing in the cell. The term also encompasses naturally occurring variants of the protein, e.g. splice variants or allelic variants. Exemplary human proteins useful as antigens include, but are not limited to Folate Receptor 1 (FolR1, Folate receptor alpha (FRA); Folate binding protein (FBP); human FolR1 UniProt no.: P15328; murine FolR1 UniProt no.: P35846; cynomolgus FolR1 UniProt no.: G7PR14), Mucin-1 (MUC1), and B Cell Maturation Antigen (BCMA), and CD3, particularly the epsilon subunit of CD3 (see UniProt no. P07766 (version 130), NCBI RefSeq no. NP_000724.1 for the human sequence; or UniProt no. Q95LI5 (version 49), NCBI GenBank no. BAB71849.1 for the cynomolgus [Macaca fascicularis] sequence).

In certain embodiments the T cell activating bispecific antigen binding molecule of the invention binds to an epitope of an activating T cell antigen or a target cell antigen that is conserved among the activating T cell antigen or target antigen from different species.

By “specific binding” is meant that the binding is selective for the antigen and can be discriminated from unwanted or non-specific interactions. The ability of an antigen binding moiety to bind to a specific antigenic determinant can be measured either through an enzyme-linked immunosorbent assay (ELISA) or other techniques familiar to one of skill in the art, e.g. surface plasmon resonance (SPR) technique (analyzed on a BIAcore instrument) (Liljeblad et al., Glyco J 17, 323-329 (2000)), and traditional binding assays (Heeley, Endocr Res 28, 217-229 (2002)). In one embodiment, the extent of binding of an antigen binding moiety to an unrelated protein is less than about 10% of the binding of the antigen binding moiety to the antigen as measured, e.g., by SPR. In certain embodiments, an antigen binding moiety that binds to the antigen, or an antigen binding molecule comprising that antigen binding moiety, has a dissociation constant (KD) of ≦1μM, ≦100 nM, ≦10 nM, ≦1 nM, ≦0.1 nM, ≦0.01 nM, or ≦0.001 nM (e.g. 10−8M or less, e.g. from 10−8M to 10−13M, e.g., from 10−9M to 10−13 M).

“Affinity” refers to the strength of the sum total of non-covalent interactions between a single binding site of a molecule (e.g., a receptor) and its binding partner (e.g., a ligand). Unless indicated otherwise, as used herein, “binding affinity” refers to intrinsic binding affinity which reflects a 1:1 interaction between members of a binding pair (e.g., an antigen binding moiety and an antigen, or a receptor and its ligand). The affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (KD), which is the ratio of dissociation and association rate constants (koff and kon, respectively). Thus, equivalent affinities may comprise different rate constants, as long as the ratio of the rate constants remains the same. Affinity can be measured by well established methods known in the art, including those described herein. A particular method for measuring affinity is Surface Plasmon Resonance (SPR).

“Reduced binding”, for example reduced binding to an Fc receptor, refers to a decrease in affinity for the respective interaction, as measured for example by SPR. For clarity the term includes also reduction of the affinity to zero (or below the detection limit of the analytic method), i.e. complete abolishment of the interaction. Conversely, “increased binding” refers to an increase in binding affinity for the respective interaction.

An “activating T cell antigen” as used herein refers to an antigenic determinant expressed on the surface of a T lymphocyte, particularly a cytotoxic T lymphocyte, which is capable of inducing T cell activation upon interaction with an antigen binding molecule. Specifically, interaction of an antigen binding molecule with an activating T cell antigen may induce T cell activation by triggering the signaling cascade of the T cell receptor complex. In a particular embodiment the activating T cell antigen is CD3.

“T cell activation” as used herein refers to one or more cellular response of a T lymphocyte, particularly a cytotoxic T lymphocyte, selected from: proliferation, differentiation, cytokine secretion, cytotoxic effector molecule release, cytotoxic activity, and expression of activation markers. The T cell activating bispecific antigen binding molecules of the invention are capable of inducing T cell activation. Suitable assays to measure T cell activation are known in the art described herein.

A “target cell antigen” as used herein refers to an antigenic determinant presented on the surface of a target cell, for example a cell in a tumor such as a cancer cell or a cell of the tumor stroma. As used herein, the terms “first” and “second” with respect to antigen binding moieties etc., are used for convenience of distinguishing when there is more than one of each type of moiety. Use of these terms is not intended to confer a specific order or orientation of the T cell activating bispecific antigen binding molecule unless explicitly so stated.

The term “BCMA” as used herein relates to human B cell maturation target, also known as BCMA; TR17_HUMAN, TNFRSF17 (UniProt Q02223), which is a member of the tumor necrosis receptor superfamily that is preferentially expressed in differentiated plasma cells. The extracellular domain of BCMA consists according to UniProt of amino acids 1-54 (or 5-51). The term “antibody against BCMA, anti BCMA antibody” as used herein relates to an antibody specifically binding to BCMA.

The term “CD3ε or CD3” as used herein relates to human CD3ε described under UniProt P07766 (CD3E_HUMAN). The term “antibody against CD3, anti CD3 antibody” relates to an antibody binding to CD3ε.

A “Fab molecule” refers to a protein consisting of the VH and CH1 domain of the heavy chain (the “Fab heavy chain”) and the VL and CL domain of the light chain (the “Fab light chain”) of an immunoglobulin. The term “Fab molecules having identical VLCL light chains” as used therein refers to binders that share one light chain but still have separate specificities. T-cell activating bispecific molecules of the invention comprise at least two Fab molecules having identical VLCL light chains. The corresponding heavy chains are remodeled and confer specific binding to a T cell activating bispecific antigen and a target cell antigen, respectively.

By “fused” is meant that the components (e.g. a Fab molecule and an Fc domain subunit) are linked by peptide bonds, either directly or via one or more peptide linkers.

The term “common light chain” as used herein refers to a light chain that within a bispecific or multispecific molecule pairs with more than one heavy chain or fragment thereof to form at least a first and a second antigen binding site, e.g., a Fab, each specific for a different antigen. For example, the common light chain pairs with a first heavy chain or fragment thereof within an antigen binding molecule to form a first binding site specific for a tumor antigen, and another copy of the common light chain pairs with a second heavy chain or fragment thereof within an antigen binding molecule to form a second binding site specific for a T cell activating antigen, e.g., CD3.

The term “immunoglobulin molecule” refers to a protein having the structure of a naturally occurring antibody. For example, immunoglobulins of the IgG class are heterotetrameric glycoproteins of about 150,000 daltons, composed of two light chains and two heavy chains that are disulfide-bonded. From N- to C-terminus, each heavy chain has a variable region (VH), also called a variable heavy domain or a heavy chain variable domain, followed by three constant domains (CH1, CH2, and CH3), also called a heavy chain constant region. Similarly, from N- to C-terminus, each light chain has a variable region (VL), also called a variable light domain or a light chain variable domain, followed by a constant light (CL) domain, also called a light chain constant region. The heavy chain of an immunoglobulin may be assigned to one of five types, called α (IgA), δ (IgD), ε (IgE), γ (IgG), or μ (IgM), some of which may be further divided into subtypes, e.g. γ1 (IgG1), γ2 (IgG2), γ3 (IgG3), γ4 (IgG4), α1 (IgA1) and α2 (IgA2). The light chain of an immunoglobulin may be assigned to one of two types, called kappa (κ) and lambda (λ), based on the amino acid sequence of its constant domain. An immunoglobulin essentially consists of two Fab molecules and an Fc domain, linked via the immunoglobulin hinge region.

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

An “antibody fragment” refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds. Examples of antibody fragments include but are not limited to Fv, Fab, Fab′, Fab′-SH, F(ab′)2, diabodies, linear antibodies, single-chain antibody molecules (e.g. scFv), and single-domain antibodies. For a review of certain antibody fragments, see Hudson et al., Nat Med 9, 129-134 (2003). For a review of scFv fragments, see e.g. Plückthun, in The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., Springer-Verlag, New York, pp. 269-315 (1994); see also WO 93/16185; and U.S. Pat. Nos. 5,571,894 and 5,587,458. For discussion of Fab and F(ab′)2 fragments comprising salvage receptor binding epitope residues and having increased in vivo half-life, see U.S. Pat. No. 5,869,046. Diabodies are antibody fragments with two antigen-binding sites that may be bivalent or bispecific. See, for example, EP 404,097; WO 1993/01161; Hudson et al., Nat Med 9, 129-134 (2003); and Hollinger et al., Proc Natl Acad Sci USA 90, 6444-6448 (1993). Triabodies and tetrabodies are also described in Hudson et al., Nat Med 9, 129-134 (2003). Single-domain antibodies are antibody fragments comprising all or a portion of the heavy chain variable domain or all or a portion of the light chain variable domain of an antibody. In certain embodiments, a single-domain antibody is a human single-domain antibody (Domantis, Inc., Waltham, Mass.; see e.g. U.S. Pat. No. 6,248,516 B1). Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as production by recombinant host cells (e.g. E. coli or phage), as described herein.

The term “antigen binding domain” refers to the part of an antibody that comprises the area which specifically binds to and is complementary to part or all of an antigen. An antigen binding domain may be provided by, for example, one or more antibody variable domains (also called antibody variable regions). Particularly, an antigen binding domain comprises an antibody light chain variable region (VL) and an antibody heavy chain variable region (VH).

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

The term “hypervariable region” or “HVR”, as used herein, refers to each of the regions of an antibody variable domain which are hypervariable in sequence and/or form structurally defined loops (“hypervariable loops”). Generally, native four-chain antibodies comprise six HVRs; three in the VH (H1, H2, H3), and three in the VL (L1, L2, L3). HVRs generally comprise amino acid residues from the hypervariable loops and/or from the complementarity determining regions (CDRs), the latter being of highest sequence variability and/or involved in antigen recognition. With the exception of CDR1 in VH, CDRs generally comprise the amino acid residues that form the hypervariable loops. Hypervariable regions (HVRs) are also referred to as “complementarity determining regions” (CDRs), and these terms are used herein interchangeably in reference to portions of the variable region that form the antigen binding regions. This particular region has been described by Kabat et al., U.S. Dept. of Health and Human Services, Sequences of Proteins of Immunological Interest (1983) and by Chothia et al., J Mol Biol 196:901-917 (1987), where the definitions include overlapping or subsets of amino acid residues when compared against each other. Nevertheless, application of either definition to refer to a CDR of an antibody or variants thereof is intended to be within the scope of the term as defined and used herein. The appropriate amino acid residues which encompass the CDRs as defined by each of the above cited references are set forth below in Table A as a comparison. The exact residue numbers which encompass a particular CDR will vary depending on the sequence and size of the CDR. Those skilled in the art can routinely determine which residues comprise a particular CDR given the variable region amino acid sequence of the antibody.

TABLE A CDR Definitions1 CDR Kabat Chothia AbM2 VH CDR1 31-35 26-32 26-35 VH CDR2 50-65 52-58 50-58 VH CDR3  95-102  95-102  95-102 VL CDR1 24-34 26-32 24-34 VL CDR2 50-56 50-52 50-56 VL CDR3 89-97 91-96 89-97 1Numbering of all CDR definitions in Table A is according to the numbering conventions set forth by Kabat et al. (see below). 2“AbM” with a lowercase “b” as used in Table A refers to the CDRs as defined by Oxford Molecular's “AbM” antibody modeling software.

Kabat et al. also defined a numbering system for variable region sequences that is applicable to any antibody. One of ordinary skill in the art can unambiguously assign this system of “Kabat numbering” to any variable region sequence, without reliance on any experimental data beyond the sequence itself. As used herein, “Kabat numbering” refers to the numbering system set forth by Kabat et al., U.S. Dept. of Health and Human Services, “Sequence of Proteins of Immunological Interest” (1983). Unless otherwise specified, references to the numbering of specific amino acid residue positions in an antibody variable region are according to the Kabat numbering system.

The polypeptide sequences of the sequence listing are not numbered according to the Kabat numbering system. However, it is well within the ordinary skill of one in the art to convert the numbering of the sequences of the Sequence Listing to Kabat numbering.

“Framework” or “FR” refers to variable domain residues other than hypervariable region (HVR) residues. The FR of a variable domain generally consists of four FR domains: FR1, FR2, FR3, and FR4. Accordingly, the HVR and FR sequences generally appear in the following sequence in VH (or VL): FR1-H1(L1)-FR2-H2(L2)-FR3-H3(L3)-FR4.

The “class” of an antibody or immunoglobulin refers to the type of constant domain or constant region possessed by its heavy chain. There are five major classes of antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgG1, IgG2, IgG3, IgG4, IgA1, and IgA2. The heavy chain constant domains that correspond to the different classes of immunoglobulins are called α, δ, ε, γ, and μ, respectively.

The term “Fc domain” or “Fc region” herein is used to define a C-terminal region of an immunoglobulin heavy chain that contains at least a portion of the constant region. The term includes native sequence Fc regions and variant Fc regions. Although the boundaries of the Fc region of an IgG heavy chain might vary slightly, the human IgG heavy chain Fc region is usually defined to extend from Cys226, or from Pro230, to the carboxyl-terminus of the heavy chain. However, the C-terminal lysine (Lys447) of the Fc region may or may not be present. Unless otherwise specified herein, numbering of amino acid residues in the Fc region or constant region is according to the EU numbering system, also called the EU index, as described in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md., 1991. A “subunit” of an Fc domain as used herein refers to one of the two polypeptides forming the dimeric Fc domain, i.e. a polypeptide comprising C-terminal constant regions of an immunoglobulin heavy chain, capable of stable self-association. For example, a subunit of an IgG Fc domain comprises an IgG CH2 and an IgG CH3 constant domain.

A “modification promoting the association of the first and the second subunit of the Fc domain” is a manipulation of the peptide backbone or the post-translational modifications of an Fc domain subunit that reduces or prevents the association of a polypeptide comprising the Fc domain subunit with an identical polypeptide to form a homodimer. A modification promoting association as used herein particularly includes separate modifications made to each of the two Fc domain subunits desired to associate (i.e. the first and the second subunit of the Fc domain), wherein the modifications are complementary to each other so as to promote association of the two Fc domain subunits. For example, a modification promoting association may alter the structure or charge of one or both of the Fc domain subunits so as to make their association sterically or electrostatically favorable, respectively. Thus, (hetero)dimerization occurs between a polypeptide comprising the first Fc domain subunit and a polypeptide comprising the second Fc domain subunit, which might be non-identical in the sense that further components fused to each of the subunits (e.g. antigen binding moieties) are not the same. In some embodiments the modification promoting association comprises an amino acid mutation in the Fc domain, specifically an amino acid substitution. In a particular embodiment, the modification promoting association comprises a separate amino acid mutation, specifically an amino acid substitution, in each of the two subunits of the Fc domain.

The term “effector functions” refers to those biological activities attributable to the Fc region of an antibody, which vary with the antibody isotype. Examples of antibody effector functions include: C1q binding and complement dependent cytotoxicity (CDC), Fc receptor binding, antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), cytokine secretion, immune complex-mediated antigen uptake by antigen presenting cells, down regulation of cell surface receptors (e.g. B cell receptor), and B cell activation.

As used herein, the terms “engineer, engineered, engineering”, are considered to include any manipulation of the peptide backbone or the post-translational modifications of a naturally occurring or recombinant polypeptide or fragment thereof. Engineering includes modifications of the amino acid sequence, of the glycosylation pattern, or of the side chain group of individual amino acids, as well as combinations of these approaches.

The term “amino acid mutation” as used herein is meant to encompass amino acid substitutions, deletions, insertions, and modifications. Any combination of substitution, deletion, insertion, and modification can be made to arrive at the final construct, provided that the final construct possesses the desired characteristics, e.g., reduced binding to an Fc receptor, or increased association with another peptide. Amino acid sequence deletions and insertions include amino- and/or carboxy-terminal deletions and insertions of amino acids. Particular amino acid mutations are amino acid substitutions. For the purpose of altering e.g. the binding characteristics of an Fc region, non-conservative amino acid substitutions, i.e. replacing one amino acid with another amino acid having different structural and/or chemical properties, are particularly preferred. Amino acid substitutions include replacement by non-naturally occurring amino acids or by naturally occurring amino acid derivatives of the twenty standard amino acids (e.g. 4-hydroxyproline, 3-methylhistidine, ornithine, homoserine, 5-hydroxylysine). Amino acid mutations can be generated using genetic or chemical methods well known in the art. Genetic methods may include site-directed mutagenesis, PCR, gene synthesis and the like. It is contemplated that methods of altering the side chain group of an amino acid by methods other than genetic engineering, such as chemical modification, may also be useful. Various designations may be used herein to indicate the same amino acid mutation. For example, a substitution from proline at position 329 of the Fc domain to glycine can be indicated as 329G, G329, G329, P329G, or Pro329Gly.

As used herein, term “polypeptide” refers to a molecule composed of monomers (amino acids) linearly linked by amide bonds (also known as peptide bonds). The term “polypeptide” refers to any chain of two or more amino acids, and does not refer to a specific length of the product. Thus, peptides, dipeptides, tripeptides, oligopeptides, “protein,” “amino acid chain,” or any other term used to refer to a chain of two or more amino acids, are included within the definition of “polypeptide,” and the term “polypeptide” may be used instead of, or interchangeably with any of these terms. The term “polypeptide” is also intended to refer to the products of post-expression modifications of the polypeptide, including without limitation glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, or modification by non-naturally occurring amino acids. A polypeptide may be derived from a natural biological source or produced by recombinant technology, but is not necessarily translated from a designated nucleic acid sequence. It may be generated in any manner, including by chemical synthesis. A polypeptide of the invention may be of a size of about 3 or more, 5 or more, 10 or more, 20 or more, 25 or more, 50 or more, 75 or more, 100 or more, 200 or more, 500 or more, 1,000 or more, or 2,000 or more amino acids. Polypeptides may have a defined three-dimensional structure, although they do not necessarily have such structure. Polypeptides with a defined three-dimensional structure are referred to as folded, and polypeptides which do not possess a defined three-dimensional structure, but rather can adopt a large number of different conformations, and are referred to as unfolded.

By an “isolated” polypeptide or a variant, or derivative thereof is intended a polypeptide that is not in its natural milieu. No particular level of purification is required. For example, an isolated polypeptide can be removed from its native or natural environment. Recombinantly produced polypeptides and proteins expressed in host cells are considered isolated for the purpose of the invention, as are native or recombinant polypeptides which have been separated, fractionated, or partially or substantially purified by any suitable technique.

“Percent (%) amino acid sequence identity” with respect to a reference polypeptide sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the reference polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. For purposes herein, however, % amino acid sequence identity values are generated using the sequence comparison computer program ALIGN-2. The ALIGN-2 sequence comparison computer program was authored by Genentech, Inc., and the source code has been filed with user documentation in the U.S. Copyright Office, Washington D.C., 20559, where it is registered under U.S. Copyright Registration No. TXU510087. The ALIGN-2 program is publicly available from Genentech, Inc., South San Francisco, Calif., or may be compiled from the source code. The ALIGN-2 program should be compiled for use on a UNIX operating system, including digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary. In situations where ALIGN-2 is employed for amino acid sequence comparisons, the % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given amino acid sequence A that has or comprises a certain % amino acid sequence identity to, with, or against a given amino acid sequence B) is calculated as follows:


100 times the fraction X/Y

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

The term “polynucleotide” refers to an isolated nucleic acid molecule or construct, e.g. messenger RNA (mRNA), virally-derived RNA, or plasmid DNA (pDNA). A polynucleotide may comprise a conventional phosphodiester bond or a non-conventional bond (e.g. an amide bond, such as found in peptide nucleic acids (PNA). The term “nucleic acid molecule” refers to any one or more nucleic acid segments, e.g. DNA or RNA fragments, present in a polynucleotide.

By “isolated” nucleic acid molecule or polynucleotide is intended a nucleic acid molecule, DNA or RNA, which has been removed from its native environment. For example, a recombinant polynucleotide encoding a polypeptide contained in a vector is considered isolated for the purposes of the present invention. Further examples of an isolated polynucleotide include recombinant polynucleotides maintained in heterologous host cells or purified (partially or substantially) polynucleotides in solution. An isolated polynucleotide includes a polynucleotide molecule contained in cells that ordinarily contain the polynucleotide molecule, but the polynucleotide molecule is present extrachromosomally or at a chromosomal location that is different from its natural chromosomal location. Isolated RNA molecules include in vivo or in vitro RNA transcripts of the present invention, as well as positive and negative strand forms, and double-stranded forms. Isolated polynucleotides or nucleic acids according to the present invention further include such molecules produced synthetically. In addition, a polynucleotide or a nucleic acid may be or may include a regulatory element such as a promoter, ribosome binding site, or a transcription terminator.

By a nucleic acid or polynucleotide having a nucleotide sequence at least, for example, 95% “identical” to a reference nucleotide sequence of the present invention, it is intended that the nucleotide sequence of the polynucleotide is identical to the reference sequence except that the polynucleotide sequence may include up to five point mutations per each 100 nucleotides of the reference nucleotide sequence. In other words, to obtain a polynucleotide having a nucleotide sequence at least 95% identical to a reference nucleotide sequence, up to 5% of the nucleotides in the reference sequence may be deleted or substituted with another nucleotide, or a number of nucleotides up to 5% of the total nucleotides in the reference sequence may be inserted into the reference sequence. These alterations of the reference sequence may occur at the 5′ or 3′ terminal positions of the reference nucleotide sequence or anywhere between those terminal positions, interspersed either individually among residues in the reference sequence or in one or more contiguous groups within the reference sequence. As a practical matter, whether any particular polynucleotide sequence is at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to a nucleotide sequence of the present invention can be determined conventionally using known computer programs, such as the ones discussed above for polypeptides (e.g. ALIGN-2).

The term “expression cassette” refers to a polynucleotide generated recombinantly or synthetically, with a series of specified nucleic acid elements that permit transcription of a particular nucleic acid in a target cell. The recombinant expression cassette can be incorporated into a plasmid, chromosome, mitochondrial DNA, plastid DNA, virus, or nucleic acid fragment. Typically, the recombinant expression cassette portion of an expression vector includes, among other sequences, a nucleic acid sequence to be transcribed and a promoter. In certain embodiments, the expression cassette of the invention comprises polynucleotide sequences that encode bispecific antigen binding molecules of the invention or fragments thereof.

The term “vector” or “expression vector” is synonymous with “expression construct” and refers to a DNA molecule that is used to introduce and direct the expression of a specific gene to which it is operably associated in a target cell. The term includes the vector as a self-replicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced. The expression vector of the present invention comprises an expression cassette. Expression vectors allow transcription of large amounts of stable mRNA. Once the expression vector is inside the target cell, the ribonucleic acid molecule or protein that is encoded by the gene is produced by the cellular transcription and/or translation machinery. In one embodiment, the expression vector of the invention comprises an expression cassette that comprises polynucleotide sequences that encode bispecific antigen binding molecules of the invention or fragments thereof.

The terms “host cell”, “host cell line,” and “host cell culture” are used interchangeably and refer to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells. Host cells include “transformants” and “transformed cells,” which include the primary transformed cell and progeny derived therefrom without regard to the number of passages. Progeny may not be completely identical in nucleic acid content to a parent cell, but may contain mutations. Mutant progeny that have the same function or biological activity as screened or selected for in the originally transformed cell are included herein. A host cell is any type of cellular system that can be used to generate the bispecific antigen binding molecules of the present invention. Host cells include cultured cells, e.g. mammalian cultured cells, such as CHO cells, BHK cells, NSO cells, SP2/0 cells, YO myeloma cells, P3X63 mouse myeloma cells, PER cells, PER.C6 cells or hybridoma cells, yeast cells, insect cells, and plant cells, to name only a few, but also cells comprised within a transgenic animal, transgenic plant or cultured plant or animal tissue.

An “activating Fc receptor” is an Fc receptor that following engagement by an Fc domain of an antibody elicits signaling events that stimulate the receptor-bearing cell to perform effector functions. Human activating Fc receptors include FcγRIIIa (CD16a), FcγRI (CD64), FcγRIIa (CD32), and FcαRI (CD89).

Antibody-dependent cell-mediated cytotoxicity (ADCC) is an immune mechanism leading to the lysis of antibody-coated target cells by immune effector cells. The target cells are cells to which antibodies or derivatives thereof comprising an Fc region specifically bind, generally via the protein part that is N-terminal to the Fc region. As used herein, the term “reduced ADCC” is defined as either a reduction in the number of target cells that are lysed in a given time, at a given concentration of antibody in the medium surrounding the target cells, by the mechanism of ADCC defined above, and/or an increase in the concentration of antibody in the medium surrounding the target cells, required to achieve the lysis of a given number of target cells in a given time, by the mechanism of ADCC. The reduction in ADCC is relative to the ADCC mediated by the same antibody produced by the same type of host cells, using the same standard production, purification, formulation and storage methods (which are known to those skilled in the art), but that has not been engineered. For example the reduction in ADCC mediated by an antibody comprising in its Fc domain an amino acid substitution that reduces ADCC, is relative to the ADCC mediated by the same antibody without this amino acid substitution in the Fc domain. Suitable assays to measure ADCC are well known in the art (see e.g. PCT publication no. WO 2006/082515 or PCT patent application no. PCT/EP2012/055393).

An “effective amount” of an agent refers to the amount that is necessary to result in a physiological change in the cell or tissue to which it is administered.

A “therapeutically effective amount” of an agent, e.g. a pharmaceutical composition, refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result. A therapeutically effective amount of an agent for example eliminates, decreases, delays, minimizes or prevents adverse effects of a disease.

An “individual” or “subject” is a mammal. Mammals include, but are not limited to, domesticated animals (e.g. cows, sheep, cats, dogs, and horses), primates (e.g. humans and non-human primates such as monkeys), rabbits, and rodents (e.g. mice and rats). Particularly, the individual or subject is a human.

The term “pharmaceutical composition” refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.

A “pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical composition, other than an active ingredient, which is nontoxic to a subject. A pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.

As used herein, “treatment” (and grammatical variations thereof such as “treat” or “treating”) refers to clinical intervention in an attempt to alter the natural course of a disease in the individual being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include, but are not limited to, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis. In some embodiments, T cell activating bispecific antigen binding molecules of the invention are used to delay development of a disease or to slow the progression of a disease.

The term “package insert” is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, combination therapy, contraindications and/or warnings concerning the use of such therapeutic products.

DETAILED DESCRIPTION OF THE EMBODIMENTS

The present invention provides bispecific antigen binding molecules designed for T cell activation and re-direction that combine good efficacy and produceability and methods of making and using the same. In particular this invention relates to bispecific molecules wherein at least two binding moieties have identical light chains and, in some embodiments, corresponding remodeled heavy chains that confer the specific binding to a T cell activating bispecific antigen and a target cell antigen, respectively. The use of this so-called ‘common light chain’ principle, i.e. combining two binders that share one light chain but still have separate specificities, prevents light chain mispairing. Thus, there are less side products during production, facilitating the homogenous preparation of T cell activating bispecific antigen binding molecules.

The present invention particularly pertains to a predefined rare light chain which contributes significantly to antigen binding and heteromultimeric pairing with different binding partners, e.g., heavy chains and fragments thereof. This common light chain is, thus, suitable as for use in a library from which new bi- or multispecific antigen binding molecules can be prepared. Advantageously, the common light chain used in connection with antigen binding molecules and methods disclosed herein can be used to form an antigen binding molecule useful for T cell activation. One of skill in the art can recognize the advantageous efficiency of having such light chain that can function in both, T cell activating antigen binding moieties and target antigen binding moieties. This allows for efficient production of T cell activating bispecific antigen binding molecules that comprise the T cell activation component and a target antigen binding component. In a particular embodiment, the common light chain a lambda constant light chain domain. In a particular embodiment, the common light chain is a human or humanized lambda light chain. In a particular embodiment, the common light chain is of the rare human lambda 7 family of light chain. Using a light chain of the lambda, particularly, the rare lambda 7 family was an uncommon approach and would have been expected to decrease the likelihood of identifying suitable heavy chain binding partners to create antigen binding molecules specific for a variety of targets. Thus, prior to the inventor's work disclosed herein, it was not known that a lambda 7 light chain with suitable properties could be developed for a variety of different unrelated target antigens such as, e.g., FolR1, MUC1, and BCMA. It was further not known that a lambda light chain could be developed to generate stable, functional, high affinity binders to improve production of T cell activating bispecific antigen binding molecules with CD3 specificity and a target antigen specificity, where the target antigens are unrelated, e.g., FolR1, MUC1, and BCMA. In one embodiment, such common light chain can be used to construct a common light chain (CLC) library which is based on a specific CD3 binder, not a germline antibody, to generate specifically CD3 binders contributing to the binder, as described below. The advantage of this approach is that it allows for maintaining the previously identified and validated CD3 binder such that merely a new target antigen binder for the target antigen binding moiety of a T cell activating bispecific antigen binding molecules has to be identified based on the heavy chain. This allows for a module approach to generating different T cell activating bispecific antigen binding molecules with identical or highly homologous chains. While the light chain is identical within a given T cell activating antigen binding molecule, the light chains of different T cell activating bispecific antigen binding molecules might be identical or highly homologous. By “highly homologous” is meant that the light chains of different T cell activating bispecific antigen binding molecules produced by this module approach comprise an amino acid sequence that is at least about 95%, 96%, 97%, 98%, or 99% identical. Preferably, highly homologous light chains of the invention have identical variable light chain regions and differ only on their constant region. For example, in some embodiments, the amino acid variance is confined to the linker region. In some embodiment, the common light chain comprises a kappa constant light chain domain.

In addition to the foregoing advantages, yield of correctly paired heteromultimeric molecules using this approach is enhanced, as explained above, because the light chain used within the antigen binding moieties of a T cell activating bispecific antigen binding molecule is identical. Stated another way, using a common light chain the production of these molecules is facilitated as any mispairing of a light chain to the incorrect heavy chain is abolished. Thus, the isolation of a highly pure T cell activating bispecific antigen binding molecule species is facilitated. In a particular embodiment, the T cell activating bispecific antigen binding molecule uses Fabs as building blocks. Compared to other formats the use of Fab fragments as building blocks as opposed to, e.g., the use of scFv fragments results in higher thermal stability and the lack of scFv aggregation and intermolecular scFv formation.

Prior to the inventors' work described herein, it was not known that a common light chain could be generated that can not only serve as common light chain in a bi- or multispecific molecule but also support the functional property of T cell activation of a T cell activating bispecific antigen binding molecule. Furthermore, prior to the inventors' work described herein, it was not known that a common light chain could be generated that significantly contributes to antigen binding properties of the antigen binding moiety within a T cell activating bispecific antigen binding molecule. A well-established strategy is to identify heavy chains or fragments thereof that contribute most of the binding properties, such as strength and specificity. According to the present invention, the common light chain significantly contributes to the binding properties.

Accordingly, in a first aspect the invention provides a T cell activating bispecific antigen binding molecule comprising a first and a second antigen binding moiety, wherein the first antigen binding moiety comprises a first light chain and wherein the first antigen binding moiety is capable of specific binding to an activating T cell antigen, wherein the second antigen binding moiety comprises a second light chain and wherein the second antigen binding moiety is capable of specific binding to a target cell antigen, wherein the amino acid sequence of the first and the second light chain is identical. In one embodiment, the first antigen binding moiety is a Fab. In one embodiment, the second antigen binding moiety is a Fab. In one aspect, the invention provides a T cell activating bispecific antigen binding molecule comprising a first and a second antigen binding moiety, one of which is a Fab molecule capable of specific binding to an activating T cell antigen and the other one of which is a Fab molecule capable of specific binding to a target cell antigen, wherein the first and the second Fab molecule have identical light chains (variable light chain and constant light chain region, VLCL). In one embodiment, the light chain (VLCL) comprises the light chain CDRs of SEQ ID NO: 32, SEQ ID NO: 33 and SEQ ID NO: 34. In one embodiment said identical light chain (VLCL) comprises SEQ ID NO. 35.

T Cell Activating Bispecific Antigen Binding Molecule Formats

The components of the T cell activating bispecific antigen binding molecule can be fused to each other in a variety of configurations. Exemplary configurations include but are not limited to those depicted in FIGS. 1A-D.

In some embodiments, said T cell activating bispecific antigen binding molecule further comprises an Fc domain composed of a first and a second subunit capable of stable association. Below exemplary embodiments of T cell activating bispecific antigen binding molecule comprising an Fc domain are described. All these T cell activating bispecific antigen binding molecules comprise at least two Fab fragments having identical light chains (VLCL) and having different heavy chains (VHCL) which confer the specifities to two different antigens, i.e. one Fab fragment is capable of specific binding to a T cell activating antigen and the other Fab fragment is capable of specific binding to a target cell antigen.

In some embodiments, the first and the second antigen binding moiety of the T cell activating bispecific antigen binding molecule are fused to each other, optionally via a peptide linker. In one such embodiment, the second antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety. In another such embodiment, the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding moiety. In yet another such embodiment, the second antigen binding moiety is fused at the C-terminus of the Fab light chain to the N-terminus of the Fab light chain of the first antigen binding moiety. In yet another such embodiment, the first antigen binding moiety is fused at the C-terminus of the Fab light chain to the N-terminus of the Fab light chain of the second antigen binding moiety.

In one embodiment the second antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or the second subunit of the Fc domain.

In a particular such embodiment, the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding moiety. In a specific such embodiment, the T cell activating bispecific antigen binding molecule essentially consists of a first and a second antigen binding moiety which comprise identical (VLCL) light chains, an Fc domain composed of a first and a second subunit, and optionally one or more peptide linkers, wherein the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding moiety, and the second antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or the second subunit of the Fc domain.

In an alternative such embodiment, the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or second subunit of the Fc domain. In a specific such embodiment, the T cell activating bispecific antigen binding molecule essentially consists of a first and a second antigen binding moiety which comprise identical (VLCL) light chains, an Fc domain composed of a first and a second subunit, and optionally one or more peptide linkers, wherein the first and the second antigen binding moiety are each fused at the C-terminus of the Fab heavy chain to the N-terminus of one of the subunits of the Fc domain.

In yet another such embodiment, the second antigen binding moiety is fused at the C-terminus of the Fab light chain to the N-terminus of the Fab light chain of the first antigen binding moiety. In a specific such embodiment, the T cell activating bispecific antigen binding molecule essentially consists of a first and a second antigen binding moiety which comprise identical (VLCL) light chains, an Fc domain composed of a first and a second subunit, and optionally one or more peptide linkers, wherein the first antigen binding moiety is fused at the N-terminus of the Fab light chain to the C-terminus of the Fab light chain of the second antigen binding moiety, and the second antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or the second subunit of the Fc domain.

In other embodiments, the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or second subunit of the Fc domain.

In a particular such embodiment, the second antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety. In a specific such embodiment, the T cell activating bispecific antigen binding molecule essentially consists of a first and a second antigen binding moiety which comprise identical (VLCL) light chains, an Fc domain composed of a first and a second subunit, and optionally one or more peptide linkers, wherein the second antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety, and the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or the second subunit of the Fc domain.

In particular of these embodiments, the first antigen binding moiety is capable of specific binding to an activating T cell antigen. In other embodiments, the first antigen binding moiety is capable of specific binding to a target cell antigen.

The antigen binding moieties may be fused to the Fc domain or to each other directly or through a peptide linker, comprising one or more amino acids, typically about 2-20 amino acids. Peptide linkers are known in the art and are described herein. Suitable, non-immunogenic peptide linkers include, for example, (G4S)n (SEQ ID NO: 41), (SG4)n (SEQ ID NO: 42), (G4S)n (SEQ ID NO: 41) or G4(SG4)n (SEQ ID NO: 43) peptide linkers. “n” is generally a number between 1 and 10, typically between 2 and 4. A particularly suitable peptide linker for fusing the Fab light chains of the first and the second antigen binding moiety to each other is (G45)2 (SEQ ID NO: 44). Additionally, linkers may comprise (a portion of) an immunoglobulin hinge region. Particularly where an antigen binding moiety is fused to the N-terminus of an Fc domain subunit, it may be fused via an immunoglobulin hinge region or a portion thereof, with or without an additional peptide linker.

A T cell activating bispecific antigen binding molecule with a single antigen binding moiety capable of specific binding to a target cell antigen is useful, particularly in cases where internalization of the target cell antigen is to be expected following binding of a high affinity antigen binding moiety. In such cases, the presence of more than one antigen binding moiety specific for the target cell antigen may enhance internalization of the target cell antigen, thereby reducing its availablity.

In many other cases, however, it will be advantageous to have a T cell activating bispecific antigen binding molecule comprising two or more antigen binding moieties specific for a target cell antigen, for example to optimize targeting to the target site or to allow crosslinking of target cell antigens.

Accordingly, in certain embodiments, the T cell activating bispecific antigen binding molecule of the invention further comprises a third antigen binding moiety which is a Fab molecule capable of specific binding to a target cell antigen. In one embodiment, the third antigen binding moiety is capable of specific binding to the same target cell antigen as the first or second antigen binding moiety. In a particular embodiment, the first antigen binding moiety is capable of specific binding to an activating T cell antigen, and the second and third antigen binding moieties are capable of specific binding to a target cell antigen. In a preferred embodiment, the first, second and third antigen binding moeity comprise identical (VLCL) light chains.

In one embodiment, the third antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or second subunit of the Fc domain.

In one embodiment, the first and the third antigen binding moiety are each fused at the C-terminus of the Fab heavy chain to the N-terminus of one of the subunits of the Fc domain, and the second antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety. In a specific such embodiment, the T cell activating bispecific antigen binding molecule essentially consists of a first, a second and a third antigen binding moiety (Fab fragment), an Fc domain composed of a first and a second subunit, and optionally one or more peptide linkers, wherein the second antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety, and the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first subunit of the Fc domain, and wherein the third antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the second subunit of the Fc domain. Preferably in said embodiment the first antigen binding moiety is capable of specific binding to an activating T cell antigen, and the second and third antigen binding moieties are capable of specific binding to a target cell antigen, wherein the first, second and third antigen binding moeities are Fab fragments which comprise identical (VLCL) light chains.

In one embodiment, the second and the third antigen binding moiety are each fused at the C-terminus of the Fab heavy chain to the N-terminus of one of the subunits of the Fc domain, and the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding moiety.

In one embodiment, the T cell activating bispecific antigen binding molecule essentially consists of an immunoglobulin molecule capable of specific binding to a target cell antigen, and a Fab molecule capable of specific binding to an activating T cell antigen, fused to the N-terminus of one of the immunoglobulin heavy chains, optionally via a peptide linker. Preferably in said embodiment the immunoglobulin molecule capable of specific binding to a target cell antigen and the Fab molecule capable of specific binding to an activating T cell antigen comprise identical (VLCL) light chains.

The first and the third antigen binding moiety (or the second and the third antigen binding moiety, respectively) may be fused to the Fc domain directly or through a peptide linker. In a particular embodiment first and the third antigen binding moiety (or the second and the third antigen binding moiety, respectively) are each fused to the Fc domain through an immunoglobulin hinge region.

In a specific embodiment, the immunoglobulin hinge region is a human IgG1 hinge region. In one embodiment first and the third antigen binding moiety (or the second and the third antigen binding moiety, respectively) and the Fc domain are part of an immunoglobulin molecule. In a particular embodiment the immunoglobulin molecule is an IgG class immunoglobulin. In an even more particular embodiment the immunoglobulin is an IgG1 subclass immunoglobulin. In another embodiment the immunoglobulin is an IgG4 subclass immunoglobulin. In a further particular embodiment the immunoglobulin is a human immunoglobulin. In other embodiments the immunoglobulin is a chimeric immunoglobulin or a humanized immunoglobulin.

Fc Domain

The Fc domain of the T cell activating bispecific antigen binding molecule consists of a pair of polypeptide chains comprising heavy chain domains of an immunoglobulin molecule. For example, the Fc domain of an immunoglobulin G (IgG) molecule is a dimer, each subunit of which comprises the CH2 and CH3 IgG heavy chain constant domains. The two subunits of the Fc domain are capable of stable association with each other. In one embodiment the T cell activating bispecific antigen binding molecule of the invention comprises not more than one Fc domain.

In one embodiment according the invention the Fc domain of the T cell activating bispecific antigen binding molecule is an IgG Fc domain. In a particular embodiment the Fc domain is an IgG1 Fc domain. In another embodiment the Fc domain is an IgG4 Fc domain. In a more specific embodiment, the Fc domain is an IgG4 Fc domain comprising an amino acid substitution at position S228 (Kabat numbering), particularly the amino acid substitution S228P. This amino acid substitution reduces in vivo Fab arm exchange of IgG4 antibodies (see Stubenrauch et al., Drug Metabolism and Disposition 38, 84-91 (2010)). In a further particular embodiment the Fc domain is human.

Fc Domain Modifications Promoting Heterodimerization

T cell activating bispecific antigen binding molecules according to the invention comprise different antigen binding moieties, fused to one or the other of the two subunits of the Fc domain, thus the two subunits of the Fc domain are typically comprised in two non-identical polypeptide chains. Recombinant co-expression of these polypeptides and subsequent dimerization leads to several possible combinations of the two polypeptides. To improve the yield and purity of T cell activating bispecific antigen binding molecules in recombinant production, it will thus be advantageous to introduce in the Fc domain of the T cell activating bispecific antigen binding molecule a modification promoting the association of the desired polypeptides.

Accordingly, in particular embodiments the Fc domain of the T cell activating bispecific antigen binding molecule according to the invention comprises a modification promoting the association of the first and the second subunit of the Fc domain. The site of most extensive protein-protein interaction between the two subunits of a human IgG Fc domain is in the CH3 domain of the Fc domain. Thus, in one embodiment said modification is in the CH3 domain of the Fc domain.

In a specific embodiment said modification is a so-called “knob-into-hole” modification, comprising a “knob” modification in one of the two subunits of the Fc domain and a “hole” modification in the other one of the two subunits of the Fc domain.

The knob-into-hole technology is described e.g. in U.S. Pat. No. 5,731,168; U.S. Pat. No. 7,695,936; Ridgway et al., Prot Eng 9, 617-621 (1996) and Carter, J Immunol Meth 248, 7-15 (2001). Generally, the method involves introducing a protuberance (“knob”) at the interface of a first polypeptide and a corresponding cavity (“hole”) in the interface of a second polypeptide, such that the protuberance can be positioned in the cavity so as to promote heterodimer formation and hinder homodimer formation. Protuberances are constructed by replacing small amino acid side chains from the interface of the first polypeptide with larger side chains (e.g. tyrosine or tryptophan). Compensatory cavities of identical or similar size to the protuberances are created in the interface of the second polypeptide by replacing large amino acid side chains with smaller ones (e.g. alanine or threonine).

Accordingly, in a particular embodiment, in the CH3 domain of the first subunit of the Fc domain of the T cell activating bispecific antigen binding molecule an amino acid residue is replaced with an amino acid residue having a larger side chain volume, thereby generating a protuberance within the CH3 domain of the first subunit which is positionable in a cavity within the CH3 domain of the second subunit, and in the CH3 domain of the second subunit of the Fc domain an amino acid residue is replaced with an amino acid residue having a smaller side chain volume, thereby generating a cavity within the CH3 domain of the second subunit within which the protuberance within the CH3 domain of the first subunit is positionable.

The protuberance and cavity can be made by altering the nucleic acid encoding the polypeptides, e.g. by site-specific mutagenesis, or by peptide synthesis.

In a specific embodiment, in the CH3 domain of the first subunit of the Fc domain the threonine residue at position 366 is replaced with a tryptophan residue (T366W), and in the CH3 domain of the second subunit of the Fc domain the tyrosine residue at position 407 is replaced with a valine residue (Y407V). In one embodiment, in the second subunit of the Fc domain additionally the threonine residue at position 366 is replaced with a serine residue (T366S) and the leucine residue at position 368 is replaced with an alanine residue (L368A).

In yet a further embodiment, in the first subunit of the Fc domain additionally the serine residue at position 354 is replaced with a cysteine residue (S354C), and in the second subunit of the Fc domain additionally the tyrosine residue at position 349 is replaced by a cysteine residue (Y349C). Introduction of these two cysteine residues results in formation of a disulfide bridge between the two subunits of the Fc domain, further stabilizing the dimer (Carter, J Immunol Methods 248, 7-15 (2001)).

In a particular embodiment the antigen binding moiety capable of binding to an activating T cell antigen is fused (optionally via the antigen binding moiety capable of binding to a target cell antigen) to the first subunit of the Fc domain (comprising the “knob” modification). Without wishing to be bound by theory, fusion of the antigen binding moiety capable of binding to an activating T cell antigen to the knob-containing subunit of the Fc domain will (further) minimize the generation of antigen binding molecules comprising two antigen binding moieties capable of binding to an activating T cell antigen (steric clash of two knob-containing polypeptides).

In an alternative embodiment a modification promoting association of the first and the second subunit of the Fc domain comprises a modification mediating electrostatic steering effects, e.g. as described in PCT publication WO 2009/089004. Generally, this method involves replacement of one or more amino acid residues at the interface of the two Fc domain subunits by charged amino acid residues so that homodimer formation becomes electrostatically unfavorable but heterodimerization electrostatically favorable.

Fc Domain Modifications Reducing Fc Receptor Binding and/or Effector Function

The Fc domain confers to the T cell activating bispecific antigen binding molecule favorable pharmacokinetic properties, including a long serum half-life which contributes to good accumulation in the target tissue and a favorable tissue-blood distribution ratio. At the same time it may, however, lead to undesirable targeting of the T cell activating bispecific antigen binding molecule to cells expressing Fc receptors rather than to the preferred antigen-bearing cells. Moreover, the co-activation of Fc receptor signaling pathways may lead to cytokine release which, in combination with the T cell activating properties and the long half-life of the antigen binding molecule, results in excessive activation of cytokine receptors and severe side effects upon systemic administration. Activation of (Fc receptor-bearing) immune cells other than T cells may even reduce efficacy of the T cell activating bispecific antigen binding molecule due to the potential destruction of T cells e.g. by NK cells.

Accordingly, in particular embodiments the Fc domain of the T cell activating bispecific antigen binding molecules according to the invention exhibits reduced binding affinity to an Fc receptor and/or reduced effector function, as compared to a native IgG1 Fc domain. In one such embodiment the Fc domain (or the T cell activating bispecific antigen binding molecule comprising said Fc domain) exhibits less than 50%, preferably less than 20%, more preferably less than 10% and most preferably less than 5% of the binding affinity to an Fc receptor, as compared to a native IgG1 Fc domain (or a T cell activating bispecific antigen binding molecule comprising a native IgG1 Fc domain), and/or less than 50%, preferably less than 20%, more preferably less than 10% and most preferably less than 5% of the effector function, as compared to a native IgG1 Fc domain domain (or a T cell activating bispecific antigen binding molecule comprising a native IgG1 Fc domain). In one embodiment, the Fc domain domain (or the T cell activating bispecific antigen binding molecule comprising said Fc domain) does not substantially bind to an Fc receptor and/or induce effector function. In a particular embodiment the Fc receptor is an Fcγ receptor. In one embodiment the Fc receptor is a human Fc receptor. In one embodiment the Fc receptor is an activating Fc receptor. In a specific embodiment the Fc receptor is an activating human Fcγ receptor, more specifically human FcγRIIIa, FcγRI or FcγRIIa, most specifically human FcγRIIIa. In one embodiment the effector function is one or more selected from the group of CDC, ADCC, ADCP, and cytokine secretion. In a particular embodiment the effector function is ADCC. In one embodiment the Fc domain domain exhibits substantially similar binding affinity to neonatal Fc receptor (FcRn), as compared to a native IgG1 Fc domain domain. Substantially similar binding to FcRn is achieved when the Fc domain (or the T cell activating bispecific antigen binding molecule comprising said Fc domain) exhibits greater than about 70%, particularly greater than about 80%, more particularly greater than about 90% of the binding affinity of a native IgG1 Fc domain (or the T cell activating bispecific antigen binding molecule comprising a native IgG1 Fc domain) to FcRn.

In certain embodiments the Fc domain is engineered to have reduced binding affinity to an Fc receptor and/or reduced effector function, as compared to a non-engineered Fc domain. In particular embodiments, the Fc domain of the T cell activating bispecific antigen binding molecule comprises one or more amino acid mutation that reduces the binding affinity of the Fc domain to an Fc receptor and/or effector function. Typically, the same one or more amino acid mutation is present in each of the two subunits of the Fc domain. In one embodiment the amino acid mutation reduces the binding affinity of the Fc domain to an Fc receptor. In one embodiment the amino acid mutation reduces the binding affinity of the Fc domain to an Fc receptor by at least 2-fold, at least 5-fold, or at least 10-fold. In embodiments where there is more than one amino acid mutation that reduces the binding affinity of the Fc domain to the Fc receptor, the combination of these amino acid mutations may reduce the binding affinity of the Fc domain to an Fc receptor by at least 10-fold, at least 20-fold, or even at least 50-fold. In one embodiment the T cell activating bispecific antigen binding molecule comprising an engineered Fc domain exhibits less than 20%, particularly less than 10%, more particularly less than 5% of the binding affinity to an Fc receptor as compared to a T cell activating bispecific antigen binding molecule comprising a non-engineered Fc domain. In a particular embodiment the Fc receptor is an Fcγ receptor. In some embodiments the Fc receptor is a human Fc receptor. In some embodiments the Fc receptor is an activating Fc receptor. In a specific embodiment the Fc receptor is an activating human Fcγ receptor, more specifically human FcγRIIIa, FcγRI or FcγRIIa, most specifically human FcγRIIIa. Preferably, binding to each of these receptors is reduced. In some embodiments binding affinity to a complement component, specifically binding affinity to C1q, is also reduced. In one embodiment binding affinity to neonatal Fc receptor (FcRn) is not reduced. Substantially similar binding to FcRn, i.e. preservation of the binding affinity of the Fc domain to said receptor, is achieved when the Fc domain (or the T cell activating bispecific antigen binding molecule comprising said Fc domain) exhibits greater than about 70% of the binding affinity of a non-engineered form of the Fc domain (or the T cell activating bispecific antigen binding molecule comprising said non-engineered form of the Fc domain) to FcRn. The Fc domain, or T cell activating bispecific antigen binding molecules of the invention comprising said Fc domain, may exhibit greater than about 80% and even greater than about 90% of such affinity. In certain embodiments the Fc domain of the T cell activating bispecific antigen binding molecule is engineered to have reduced effector function, as compared to a non-engineered Fc domain. The reduced effector function can include, but is not limited to, one or more of the following: reduced complement dependent cytotoxicity (CDC), reduced antibody-dependent cell-mediated cytotoxicity (ADCC), reduced antibody-dependent cellular phagocytosis (ADCP), reduced cytokine secretion, reduced immune complex-mediated antigen uptake by antigen-presenting cells, reduced binding to NK cells, reduced binding to macrophages, reduced binding to monocytes, reduced binding to polymorphonuclear cells, reduced direct signaling inducing apoptosis, reduced crosslinking of target-bound antibodies, reduced dendritic cell maturation, or reduced T cell priming. In one embodiment the reduced effector function is one or more selected from the group of reduced CDC, reduced ADCC, reduced ADCP, and reduced cytokine secretion. In a particular embodiment the reduced effector function is reduced ADCC. In one embodiment the reduced ADCC is less than 20% of the ADCC induced by a non-engineered Fc domain (or a T cell activating bispecific antigen binding molecule comprising a non-engineered Fc domain).

In one embodiment the amino acid mutation that reduces the binding affinity of the Fc domain to an Fc receptor and/or effector function is an amino acid substitution. In one embodiment the Fc domain comprises an amino acid substitution at a position selected from the group of E233, L234, L235, N297, P331 and P329. In a more specific embodiment the Fc domain comprises an amino acid substitution at a position selected from the group of L234, L235 and P329. In some embodiments the Fc domain comprises the amino acid substitutions L234A and L235A. In one such embodiment, the Fc domain is an IgG1 Fc domain, particularly a human IgG1 Fc domain. In one embodiment the Fc domain comprises an amino acid substitution at position P329. In a more specific embodiment the amino acid substitution is P329A or P329G, particularly P329G. In one embodiment the Fc domain comprises an amino acid substitution at position P329 and a further amino acid substitution at a position selected from E233, L234, L235, N297 and P331. In a more specific embodiment the further amino acid substitution is E233P, L234A, L235A, L235E, N297A, N297D or P331S. In particular embodiments the Fc domain comprises amino acid substitutions at positions P329, L234 and L235. In more particular embodiments the Fc domain comprises the amino acid mutations L234A, L235A and P329G (“P329G LALA”). In one such embodiment, the Fc domain is an IgG1 Fc domain, particularly a human IgG1 Fc domain. The “P329G LALA” combination of amino acid substitutions almost completely abolishes Fcγreceptor binding of a human IgG1 Fc domain, as described in PCT patent application no. PCT/EP2012/055393, incorporated herein by reference in its entirety. PCT/EP2012/055393 also describes methods of preparing such mutant Fc domains and methods for determining its properties such as Fc receptor binding or effector functions.

IgG4 antibodies exhibit reduced binding affinity to Fc receptors and reduced effector functions as compared to IgG1 antibodies. Hence, in some embodiments the Fc domain of the T cell activating bispecific antigen binding molecules of the invention is an IgG4 Fc domain, particularly a human IgG4 Fc domain. In one embodiment the IgG4 Fc domain comprises amino acid substitutions at position S228, specifically the amino acid substitution S228P. To further reduce its binding affinity to an Fc receptor and/or its effector function, in one embodiment the IgG4 Fc domain comprises an amino acid substitution at position L235, specifically the amino acid substitution L235E. In another embodiment, the IgG4 Fc domain comprises an amino acid substitution at position P329, specifically the amino acid substitution P329G. In a particular embodiment, the IgG4 Fc domain comprises amino acid substitutions at positions S228, L235 and P329, specifically amino acid substitutions S228P, L235E and P329G. Such IgG4 Fc domain mutants and their Fcγ receptor binding properties are described in PCT patent application no. PCT/EP2012/055393, incorporated herein by reference in its entirety.

In a particular embodiment the Fc domain exhibiting reduced binding affinity to an Fc receptor and/or reduced effector function, as compared to a native IgG1 Fc domain, is a human IgG1 Fc domain comprising the amino acid substitutions L234A, L235A and optionally P329G, or a human IgG4 Fc domain comprising the amino acid substitutions S228P, L235E and optionally P329G.

In certain embodiments N-glycosylation of the Fc domain has been eliminated. In one such embodiment the Fc domain comprises an amino acid mutation at position N297, particularly an amino acid substitution replacing asparagine by alanine (N297A) or aspartic acid (N297D).

In addition to the Fc domains described hereinabove and in PCT patent application no. PCT/EP2012/055393, Fc domains with reduced Fc receptor binding and/or effector function also include those with substitution of one or more of Fc domain residues 238, 265, 269, 270, 297, 327 and 329 (U.S. Pat. No. 6,737,056). Such Fc mutants include Fc mutants with substitutions at two or more of amino acid positions 265, 269, 270, 297 and 327, including the so-called “DANA” Fc mutant with substitution of residues 265 and 297 to alanine (U.S. Pat. No. 7,332,581).

Mutant Fc domains can be prepared by amino acid deletion, substitution, insertion or modification using genetic or chemical methods well known in the art. Genetic methods may include site-specific mutagenesis of the encoding DNA sequence, PCR, gene synthesis, and the like. The correct nucleotide changes can be verified for example by sequencing.

Binding to Fc receptors can be easily determined e.g. by ELISA, or by Surface Plasmon Resonance (SPR) using standard instrumentation such as a BIAcore instrument (GE Healthcare), and Fc receptors such as may be obtained by recombinant expression. A suitable such binding assay is described herein. Alternatively, binding affinity of Fc domains or cell activating bispecific antigen binding molecules comprising an Fc domain for Fc receptors may be evaluated using cell lines known to express particular Fc receptors, such as human NK cells expressing FcγIIIa receptor.

Effector function of an Fc domain, or a T cell activating bispecific antigen binding molecule comprising an Fc domain, can be measured by methods known in the art. A suitable assay for measuring ADCC is described herein. Other examples of in vitro assays to assess ADCC activity of a molecule of interest are described in U.S. Pat. No. 5,500,362; Hellstrom et al. Proc Natl Acad Sci USA 83, 7059-7063 (1986) and Hellstrom et al., Proc Natl Acad Sci USA 82, 1499-1502 (1985); U.S. Pat. No. 5,821,337; Bruggemann et al., J Exp Med 166, 1351-1361 (1987). Alternatively, non-radioactive assays methods may be employed (see, for example, ACTI™ non-radioactive cytotoxicity assay for flow cytometry (CellTechnology, Inc. Mountain View, Calif.); and CytoTox 96® non-radioactive cytotoxicity assay (Promega, Madison, Wis.)). Useful effector cells for such assays include peripheral blood mononuclear cells (PBMC) and Natural Killer (NK) cells. Alternatively, or additionally, ADCC activity of the molecule of interest may be assessed in vivo, e.g. in a animal model such as that disclosed in Clynes et al., Proc Natl Acad Sci USA 95, 652-656 (1998).

In some embodiments, binding of the Fc domain to a complement component, specifically to C1q, is reduced. Accordingly, in some embodiments wherein the Fc domain is engineered to have reduced effector function, said reduced effector function includes reduced CDC. C1q binding assays may be carried out to determine whether the T cell activating bispecific antigen binding molecule is able to bind C1q and hence has CDC activity. See e.g., C1q and C3c binding ELISA in WO 2006/029879 and WO 2005/100402. To assess complement activation, a CDC assay may be performed (see, for example, Gazzano-Santoro et al., J Immunol Methods 202, 163 (1996); Cragg et al., Blood 101, 1045-1052 (2003); and Cragg and Glennie, Blood 103, 2738-2743 (2004)).

Antigen Binding Moieties

The antigen binding molecule of the invention is bispecific, i.e. it comprises at least two antigen binding moieties capable of specific binding to two distinct antigenic determinants. According to one embodiment of the invention, the antigen binding moieties are Fab molecules (i.e. antigen binding domains composed of a heavy and a light chain, each comprising a variable and a constant region), wherein the light chain (VLCL) of the at least two Fab molecules comprises identical sequences. In one embodiment said VLCL light chain of the Fab molecules capable of specific binding to a target cell antigen and a T cell activating antigen, respectively, comprises the light chain CDRs of SEQ ID NO: 32, SEQ ID NO: 33 and SEQ ID NO: 34.

In one embodiment said VLCL light chain of the Fab molecules capable of specific binding to a target cell antigen and a T cell activating antigen, respectively, comprises SEQ ID NO: 31.

In one embodiment said Fab molecules are human. In another embodiment said Fab molecules are humanized. In yet another embodiment said Fab molecules comprise human heavy and light chain constant regions.

In a particular embodiment according to the invention, the T cell activating bispecific antigen binding molecule is capable of simultaneous binding to a target cell antigen, particularly a tumor cell antigen, and an activating T cell antigen. In one embodiment, the T cell activating bispecific antigen binding molecule is capable of crosslinking a T cell and a target cell by simultaneous binding to a target cell antigen and an activating T cell antigen. In an even more particular embodiment, such simultaneous binding results in lysis of the target cell, particularly a tumor cell. In one embodiment, such simultaneous binding results in activation of the T cell. In other embodiments, such simultaneous binding results in a cellular response of a T lymphocyte, particularly a cytotoxic T lymphocyte, selected from the group of: proliferation, differentiation, cytokine secretion, cytotoxic effector molecule release, cytotoxic activity, and expression of activation markers. In one embodiment, binding of the T cell activating bispecific antigen binding molecule to the activating T cell antigen without simultaneous binding to the target cell antigen does not result in T cell activation.

In one embodiment, the T cell activating bispecific antigen binding molecule is capable of re-directing cytotoxic activity of a T cell to a target cell. In a particular embodiment, said re-direction is independent of MHC-mediated peptide antigen presentation by the target cell and and/or specificity of the T cell.

Particularly, a T cell according to any of the embodiments of the invention is a cytotoxic T cell.

In some embodiments the T cell is a CD4+ or a CD8+ T cell, particularly a CD8+ T cell.

Activating T Cell Antigen Binding Moiety

The T cell activating bispecific antigen binding molecule of the invention comprises at least one antigen binding moiety capable of binding to an activating T cell antigen (also referred to herein as an “activating T cell antigen binding moiety”). In a particular embodiment, the T cell activating bispecific antigen binding molecule comprises not more than one antigen binding moiety capable of specific binding to an activating T cell antigen. In one embodiment the T cell activating bispecific antigen binding molecule provides monovalent binding to the activating T cell antigen. The activating T cell antigen binding moiety is a Fab molecule and comprises an identical VLCL light chain as the antigen binding moiety capable of specific binding to a target cell antigen.

In a particular embodiment the activating T cell antigen is CD3, particularly human CD3 or cynomolgus CD3, most particularly human CD3. In a particular embodiment the activating T cell antigen binding moiety is cross-reactive for (i.e. specifically binds to) human and cynomolgus CD3. In some embodiments, the activating T cell antigen is the epsilon subunit of CD3 (SEQ ID NO: 56).

In one embodiment, the activating T cell antigen binding moiety can compete with monoclonal antibody H2C (described in PCT publication no. WO2008/119567) for binding an epitope of CD3. In another embodiment, the activating T cell antigen binding moiety can compete with monoclonal antibody V9 (described in Rodrigues et al., Int J Cancer Suppl 7, 45-50 (1992) and U.S. Pat. No. 6,054,297) for binding an epitope of CD3. In yet another embodiment, the activating T cell antigen binding moiety can compete with monoclonal antibody FN18 (described in Nooij et al., Eur J Immunol 19, 981-984 (1986)) for binding an epitope of CD3. In a particular embodiment, the activating T cell antigen binding moiety can compete with monoclonal antibody SP34 (described in Pessano et al., EMBO J 4, 337-340 (1985)) for binding an epitope of CD3. In one embodiment, the activating T cell antigen binding moiety binds to the same epitope of CD3 as monoclonal antibody SP34.

In one embodiment, the activating T cell antigen binding moiety comprises the heavy chain CDR1 of SEQ ID NO: 14, the heavy chain CDR2 of SEQ ID NO: 15, the heavy chain CDR3 of SEQ ID NO: 16, the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO: 34. In a further embodiment, the activating T cell antigen binding moiety comprises a heavy chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 36, or variants thereof that retain functionality.

In a further embodiment, the activating T cell antigen binding moiety comprises a light chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 31, or variants thereof that retain functionality.

In one embodiment the activating T cell antigen binding moiety comprises a heavy chain comprising the sequence of SEQ ID NO: 36 and a light chain comprising the sequence of SEQ ID NO: 31.

In one embodiment the activating T cell antigen binding moiety comprises a heavy chain of SEQ ID NO: 40 and a light chain comprising of SEQ ID NO: 35.

Target Cell Antigen Binding Moiety

The T cell activating bispecific antigen binding molecule of the invention comprises at least one antigen binding moiety capable of binding to a target cell antigen (also referred to herein as an “target cell antigen binding moiety”). In certain embodiments, the T cell activating bispecific antigen binding molecule comprises two antigen binding moieties capable of binding to a target cell antigen. In a particular such embodiment, each of these antigen binding moieties specifically binds to the same antigenic determinant. In one embodiment, the T cell activating bispecific antigen binding molecule comprises an immunoglobulin molecule capable of specific binding to a target cell antigen. In one embodiment the T cell activating bispecific antigen binding molecule comprises not more than two antigen binding moieties capable of binding to a target cell antigen. The target cell antigen binding moiety is generally a Fab molecule that binds to a specific antigenic determinant and is able to direct the T cell activating bispecific antigen binding molecule to a target site, for example to a specific type of tumor cell that bears the antigenic determinant. Said Fab molecule has an identical VLCL light chain as the Fab molecule capable of specific binding to a T cell activating antigen. In a preferred embodiment said VLCL light chain of the Fab molecule capable of specific binding to a target cell antigen and the Fab molecule capable of specific binding to a T cell activating antigen comprise the light chain CDR1 of SEQ ID NO: 32, the light chain CDR2 of SEQ ID NO: 33, and the light chain CDR3 of SEQ ID NO: 34. In a preferred embodiment said VLCL light chain of the Fab molecule capable of specific binding to a target cell antigen and the Fab molecule capable of specific binding to a T cell activating antigen comprise a VLCL light chain of SEQ ID NO. 31.

In a further embodiment, the target cell antigen binding moiety comprises a light chain variable region sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 31, or variants thereof that retain functionality.

In a further embodiment, the target cell antigen binding moiety comprises a light chain sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 35, or variants thereof that retain functionality.

In certain embodiments the target cell antigen binding moiety is directed to an antigen associated with a pathological condition, such as an antigen presented on a tumor cell or on a virus-infected cell. Suitable antigens are cell surface antigens, for example, but not limited to, cell surface receptors. In particular embodiments the antigen is a human antigen. In a specific embodiment the target cell antigen is selected from the group of Folate Receptor 1 (FolR1), Mucin-1 (MUC1), and B Cell Maturation Antigen (BCMA).

In a specific embodiment the T cell activating bispecific antigen binding molecule comprises a polypeptide sequence encoded by a polynucleotide sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to a sequence selected from the group of SEQ ID NO: 183, SEQ ID NO: 193, SEQ ID NO: 263, SEQ ID NO: 264, SEQ ID NO: 191, SEQ ID NO: 198, SEQ ID NO: 267, and SEQ ID NO: 272, including functional fragments or variants thereof.

Polynucleotides

The invention further provides isolated polynucleotides encoding a T cell activating bispecific antigen binding molecule as described herein or a fragment thereof.

Polynucleotides of the invention include those that are at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequences set forth in SEQ ID NO: 183, SEQ ID NO: 193, SEQ ID NO: 263, SEQ ID NO: 264, SEQ ID NO: 191, SEQ ID NO: 198, SEQ ID NO: 267, and SEQ ID NO: 272, including functional fragments or variants thereof.

The polynucleotides encoding T cell activating bispecific antigen binding molecules of the invention may be expressed as a single polynucleotide that encodes the entire T cell activating bispecific antigen binding molecule or as multiple (e.g., two or more) polynucleotides that are co-expressed. Polypeptides encoded by polynucleotides that are co-expressed may associate through, e.g., disulfide bonds or other means to form a functional T cell activating bispecific antigen binding molecule. For example, the light chain portion of an antigen binding moiety may be encoded by a separate polynucleotide from the portion of the T cell activating bispecific antigen binding molecule comprising the heavy chain portion of the antigen binding moiety, an Fc domain subunit and optionally (part of) another antigen binding moiety. When co-expressed, the heavy chain polypeptides will associate with the light chain polypeptides to form the antigen binding moiety. In another example, the portion of the T cell activating bispecific antigen binding molecule comprising one of the two Fc domain subunits and optionally (part of) one or more antigen binding moieties could be encoded by a separate polynucleotide from the portion of the T cell activating bispecific antigen binding molecule comprising the the other of the two Fc domain subunits and optionally (part of) an antigen binding moiety. When co-expressed, the Fc domain subunits will associate to form the Fc domain.

In certain embodiments the polynucleotide or nucleic acid is DNA. In other embodiments, a polynucleotide of the present invention is RNA, for example, in the form of messenger RNA (mRNA). RNA of the present invention may be single stranded or double stranded.

Recombinant Methods

T cell activating bispecific antigen binding molecules of the invention may be obtained, for example, by solid-state peptide synthesis (e.g. Merrifield solid phase synthesis) or recombinant production. For recombinant production one or more polynucleotide encoding the T cell activating bispecific antigen binding molecule (fragment), e.g., as described above, is isolated and inserted into one or more vectors for further cloning and/or expression in a host cell. Such polynucleotide may be readily isolated and sequenced using conventional procedures. In one embodiment a vector, preferably an expression vector, comprising one or more of the polynucleotides of the invention is provided. Methods which are well known to those skilled in the art can be used to construct expression vectors containing the coding sequence of a T cell activating bispecific antigen binding molecule (fragment) along with appropriate transcriptional/translational control signals. These methods include in vitro recombinant DNA techniques, synthetic techniques and in vivo recombination/genetic recombination. See, for example, the techniques described in Maniatis et al., MOLECULAR CLONING: A LABORATORY MANUAL, Cold Spring Harbor Laboratory, N.Y. (1989); and Ausubel et al., CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, Greene Publishing Associates and Wiley Interscience, N.Y (1989). The expression vector can be part of a plasmid, virus, or may be a nucleic acid fragment. The expression vector includes an expression cassette into which the polynucleotide encoding the T cell activating bispecific antigen binding molecule (fragment) (i.e. the coding region) is cloned in operable association with a promoter and/or other transcription or translation control elements. As used herein, a “coding region” is a portion of nucleic acid which consists of codons translated into amino acids. Although a “stop codon” (TAG, TGA, or TAA) is not translated into an amino acid, it may be considered to be part of a coding region, if present, but any flanking sequences, for example promoters, ribosome binding sites, transcriptional terminators, introns, 5′ and 3′ untranslated regions, and the like, are not part of a coding region. Two or more coding regions can be present in a single polynucleotide construct, e.g. on a single vector, or in separate polynucleotide constructs, e.g. on separate (different) vectors. Furthermore, any vector may contain a single coding region, or may comprise two or more coding regions, e.g. a vector of the present invention may encode one or more polypeptides, which are post- or co-translationally separated into the final proteins via proteolytic cleavage. In addition, a vector, polynucleotide, or nucleic acid of the invention may encode heterologous coding regions, either fused or unfused to a polynucleotide encoding the T cell activating bispecific antigen binding molecule (fragment) of the invention, or variant or derivative thereof. Heterologous coding regions include without limitation specialized elements or motifs, such as a secretory signal peptide or a heterologous functional domain. An operable association is when a coding region for a gene product, e.g. a polypeptide, is associated with one or more regulatory sequences in such a way as to place expression of the gene product under the influence or control of the regulatory sequence(s). Two DNA fragments (such as a polypeptide coding region and a promoter associated therewith) are “operably associated” if induction of promoter function results in the transcription of mRNA encoding the desired gene product and if the nature of the linkage between the two DNA fragments does not interfere with the ability of the expression regulatory sequences to direct the expression of the gene product or interfere with the ability of the DNA template to be transcribed. Thus, a promoter region would be operably associated with a nucleic acid encoding a polypeptide if the promoter was capable of effecting transcription of that nucleic acid. The promoter may be a cell-specific promoter that directs substantial transcription of the DNA only in predetermined cells. Other transcription control elements, besides a promoter, for example enhancers, operators, repressors, and transcription termination signals, can be operably associated with the polynucleotide to direct cell-specific transcription. Suitable promoters and other transcription control regions are disclosed herein. A variety of transcription control regions are known to those skilled in the art. These include, without limitation, transcription control regions, which function in vertebrate cells, such as, but not limited to, promoter and enhancer segments from cytomegaloviruses (e.g. the immediate early promoter, in conjunction with intron-A), simian virus 40 (e.g. the early promoter), and retroviruses (such as, e.g. Rous sarcoma virus). Other transcription control regions include those derived from vertebrate genes such as actin, heat shock protein, bovine growth hormone and rabbit â-globin, as well as other sequences capable of controlling gene expression in eukaryotic cells. Additional suitable transcription control regions include tissue-specific promoters and enhancers as well as inducible promoters (e.g. promoters inducible tetracyclins). Similarly, a variety of translation control elements are known to those of ordinary skill in the art. These include, but are not limited to ribosome binding sites, translation initiation and termination codons, and elements derived from viral systems (particularly an internal ribosome entry site, or IRES, also referred to as a CITE sequence). The expression cassette may also include other features such as an origin of replication, and/or chromosome integration elements such as retroviral long terminal repeats (LTRs), or adeno-associated viral (AAV) inverted terminal repeats (ITRs).

Polynucleotide and nucleic acid coding regions of the present invention may be associated with additional coding regions which encode secretory or signal peptides, which direct the secretion of a polypeptide encoded by a polynucleotide of the present invention. For example, if secretion of the T cell activating bispecific antigen binding molecule is desired, DNA encoding a signal sequence may be placed upstream of the nucleic acid encoding a T cell activating bispecific antigen binding molecule of the invention or a fragment thereof. According to the signal hypothesis, proteins secreted by mammalian cells have a signal peptide or secretory leader sequence which is cleaved from the mature protein once export of the growing protein chain across the rough endoplasmic reticulum has been initiated. Those of ordinary skill in the art are aware that polypeptides secreted by vertebrate cells generally have a signal peptide fused to the N-terminus of the polypeptide, which is cleaved from the translated polypeptide to produce a secreted or “mature” form of the polypeptide. In certain embodiments, the native signal peptide, e.g. an immunoglobulin heavy chain or light chain signal peptide is used, or a functional derivative of that sequence that retains the ability to direct the secretion of the polypeptide that is operably associated with it. Alternatively, a heterologous mammalian signal peptide, or a functional derivative thereof, may be used. For example, the wild-type leader sequence may be substituted with the leader sequence of human tissue plasminogen activator (TPA) or mouse β-glucuroni dase.

DNA encoding a short protein sequence that could be used to facilitate later purification (e.g. a histidine tag) or assist in labeling the T cell activating bispecific antigen binding molecule may be included within or at the ends of the T cell activating bispecific antigen binding molecule (fragment) encoding polynucleotide.

In a further embodiment, a host cell comprising one or more polynucleotides of the invention is provided. In certain embodiments a host cell comprising one or more vectors of the invention is provided. The polynucleotides and vectors may incorporate any of the features, singly or in combination, described herein in relation to polynucleotides and vectors, respectively. In one such embodiment a host cell comprises (e.g. has been transformed or transfected with) a vector comprising a polynucleotide that encodes (part of) a T cell activating bispecific antigen binding molecule of the invention. As used herein, the term “host cell” refers to any kind of cellular system which can be engineered to generate the T cell activating bispecific antigen binding molecules of the invention or fragments thereof. Host cells suitable for replicating and for supporting expression of T cell activating bispecific antigen binding molecules are well known in the art. Such cells may be transfected or transduced as appropriate with the particular expression vector and large quantities of vector containing cells can be grown for seeding large scale fermenters to obtain sufficient quantities of the T cell activating bispecific antigen binding molecule for clinical applications. Suitable host cells include prokaryotic microorganisms, such as E. coli, or various eukaryotic cells, such as Chinese hamster ovary cells (CHO), insect cells, or the like. For example, polypeptides may be produced in bacteria in particular when glycosylation is not needed. After expression, the polypeptide may be isolated from the bacterial cell paste in a soluble fraction and can be further purified. In addition to prokaryotes, eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for polypeptide-encoding vectors, including fungi and yeast strains whose glycosylation pathways have been “humanized”, resulting in the production of a polypeptide with a partially or fully human glycosylation pattern. See Gerngross, Nat Biotech 22, 1409-1414 (2004), and Li et al., Nat Biotech 24, 210-215 (2006). Suitable host cells for the expression of (glycosylated) polypeptides are also derived from multicellular organisms (invertebrates and vertebrates). Examples of invertebrate cells include plant and insect cells. Numerous baculoviral strains have been identified which may be used in conjunction with insect cells, particularly for transfection of Spodoptera frugiperda cells. Plant cell cultures can also be utilized as hosts. See e.g. U.S. Pat. Nos. 5,959,177, 6,040,498, 6,420,548, 7,125,978, and 6,417,429 (describing PLANTIBODIES™ technology for producing antibodies in transgenic plants). Vertebrate cells may also be used as hosts. For example, mammalian cell lines that are adapted to grow in suspension may be useful. Other examples of useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7); human embryonic kidney line (293 or 293T cells as described, e.g., in Graham et al., J Gen Virol 36, 59 (1977)), baby hamster kidney cells (BHK), mouse sertoli cells (TM4 cells as described, e.g., in Mather, Biol Reprod 23, 243-251 (1980)), monkey kidney cells (CV1), African green monkey kidney cells (VERO-76), human cervical carcinoma cells (HELA), canine kidney cells (MDCK), buffalo rat liver cells (BRL 3A), human lung cells (W138), human liver cells (Hep G2), mouse mammary tumor cells (MMT 060562), TRI cells (as described, e.g., in Mather et al., Annals N.Y. Acad Sci 383, 44-68 (1982)), MRC 5 cells, and FS4 cells. Other useful mammalian host cell lines include Chinese hamster ovary (CHO) cells, including dhfr CHO cells (Urlaub et al., Proc Natl Acad Sci USA 77, 4216 (1980)); and myeloma cell lines such as YO, NS0, P3X63 and Sp2/0. For a review of certain mammalian host cell lines suitable for protein production, see, e.g., Yazaki and Wu, Methods in Molecular Biology, Vol. 248 (B.K.C. Lo, ed., Humana Press, Totowa, N.J.), pp. 255-268 (2003). Host cells include cultured cells, e.g., mammalian cultured cells, yeast cells, insect cells, bacterial cells and plant cells, to name only a few, but also cells comprised within a transgenic animal, transgenic plant or cultured plant or animal tissue. In one embodiment, the host cell is a eukaryotic cell, preferably a mammalian cell, such as a Chinese Hamster Ovary (CHO) cell, a human embryonic kidney (HEK) cell or a lymphoid cell (e.g., Y0, NS0, Sp20 cell).

Standard technologies are known in the art to express foreign genes in these systems. Cells expressing a polypeptide comprising either the heavy or the light chain of an antigen binding domain such as an antibody, may be engineered so as to also express the other of the antibody chains such that the expressed product is an antibody that has both a heavy and a light chain.

In one embodiment, a method of producing a T cell activating bispecific antigen binding molecule according to the invention is provided, wherein the method comprises culturing a host cell comprising a polynucleotide encoding the T cell activating bispecific antigen binding molecule, as provided herein, under conditions suitable for expression of the T cell activating bispecific antigen binding molecule, and recovering the T cell activating bispecific antigen binding molecule from the host cell (or host cell culture medium).

The components of the T cell activating bispecific antigen binding molecule are genetically fused to each other. T cell activating bispecific antigen binding molecule can be designed such that its components are fused directly to each other or indirectly through a linker sequence. The composition and length of the linker may be determined in accordance with methods well known in the art and may be tested for efficacy. Examples of linker sequences between different components of T cell activating bispecific antigen binding molecules are found in the sequences provided herein. Additional sequences may also be included to incorporate a cleavage site to separate the individual components of the fusion if desired, for example an endopeptidase recognition sequence.

In certain embodiments the one or more antigen binding moieties of the T cell activating bispecific antigen binding molecules comprise at least an antibody variable region capable of binding an antigenic determinant. Variable regions can form part of and be derived from naturally or non-naturally occurring antibodies and fragments thereof. Methods to produce polyclonal antibodies and monoclonal antibodies are well known in the art (see e.g. Harlow and Lane, “Antibodies, a laboratory manual”, Cold Spring Harbor Laboratory, 1988). Non-naturally occurring antibodies can be constructed using solid phase-peptide synthesis, can be produced recombinantly (e.g. as described in U.S. Pat. No. 4,186,567) or can be obtained, for example, by screening combinatorial libraries comprising variable heavy chains and variable light chains (see e.g. U.S. Pat. No. 5,969,108 to McCafferty).

Any animal species of antibody, antibody fragment, antigen binding domain or variable region can be used in the T cell activating bispecific antigen binding molecules of the invention. Non-limiting antibodies, antibody fragments, antigen binding domains or variable regions useful in the present invention can be of murine, primate, or human origin. If the T cell activating bispecific antigen binding molecule is intended for human use, a chimeric form of antibody may be used wherein the constant regions of the antibody are from a human. A humanized or fully human form of the antibody can also be prepared in accordance with methods well known in the art (see e. g. U.S. Pat. No. 5,565,332 to Winter). Humanization may be achieved by various methods including, but not limited to (a) grafting the non-human (e.g., donor antibody) CDRs onto human (e.g. recipient antibody) framework and constant regions with or without retention of critical framework residues (e.g. those that are important for retaining good antigen binding affinity or antibody functions), (b) grafting only the non-human specificity-determining regions (SDRs or a-CDRs; the residues critical for the antibody-antigen interaction) onto human framework and constant regions, or (c) transplanting the entire non-human variable domains, but “cloaking” them with a human-like section by replacement of surface residues. Humanized antibodies and methods of making them are reviewed, e.g., in Almagro and Fransson, Front Biosci 13, 1619-1633 (2008), and are further described, e.g., in Riechmann et al., Nature 332, 323-329 (1988); Queen et al., Proc Natl Acad Sci USA 86, 10029-10033 (1989); U.S. Pat. Nos. 5,821,337, 7,527,791, 6,982,321, and 7,087,409; Jones et al., Nature 321, 522-525 (1986); Morrison et al., Proc Natl Acad Sci 81, 6851-6855 (1984); Morrison and Oi, Adv Immunol 44, 65-92 (1988); Verhoeyen et al., Science 239, 1534-1536 (1988); Padlan, Molec Immun 31(3), 169-217 (1994); Kashmiri et al., Methods 36, 25-34 (2005) (describing SDR (a-CDR) grafting); Padlan, Mol Immunol 28, 489-498 (1991) (describing “resurfacing”); Dall'Acqua et al., Methods 36, 43-60 (2005) (describing “FR shuffling”); and Osbourn et al., Methods 36, 61-68 (2005) and Klimka et al., Br J Cancer 83, 252-260 (2000) (describing the “guided selection” approach to FR shuffling). Human antibodies and human variable regions can be produced using various techniques known in the art. Human antibodies are described generally in van Dijk and van de Winkel, Curr Opin Pharmacol 5, 368-74 (2001) and Lonberg, Curr Opin Immunol 20, 450-459 (2008). Human variable regions can form part of and be derived from human monoclonal antibodies made by the hybridoma method (see e.g. Monoclonal Antibody Production Techniques and Applications, pp. 51-63 (Marcel Dekker, Inc., New York, 1987)). Human antibodies and human variable regions may also be prepared by administering an immunogen to a transgenic animal that has been modified to produce intact human antibodies or intact antibodies with human variable regions in response to antigenic challenge (see e.g. Lonberg, Nat Biotech 23, 1117-1125 (2005). Human antibodies and human variable regions may also be generated by isolating Fv clone variable region sequences selected from human-derived phage display libraries (see e.g., Hoogenboom et al. in Methods in Molecular Biology 178, 1-37 (O′Brien et al., ed., Human Press, Totowa, N.J., 2001); and McCafferty et al., Nature 348, 552-554; Clackson et al., Nature 352, 624-628 (1991)). Phage typically display antibody fragments, either as single-chain Fv (scFv) fragments or as Fab fragments.

In certain embodiments, the antigen binding moieties useful in the present invention are engineered to have enhanced binding affinity according to, for example, the methods disclosed in U.S. Pat. Appl. Publ. No. 2004/0132066, the entire contents of which are hereby incorporated by reference. The ability of the T cell activating bispecific antigen binding molecule of the invention to bind to a specific antigenic determinant can be measured either through an enzyme-linked immunosorbent assay (ELISA) or other techniques familiar to one of skill in the art, e.g. surface plasmon resonance technique (analyzed on a BIACORE T100 system) (Liljeblad, et al., Glyco J 17, 323-329 (2000)), and traditional binding assays (Heeley, Endocr Res 28, 217-229 (2002)). Competition assays may be used to identify an antibody, antibody fragment, antigen binding domain or variable domain that competes with a reference antibody for binding to a particular antigen, e.g. an antibody that competes with the V9 antibody for binding to CD3. In certain embodiments, such a competing antibody binds to the same epitope (e.g. a linear or a conformational epitope) that is bound by the reference antibody. Detailed exemplary methods for mapping an epitope to which an antibody binds are provided in Morris (1996) “Epitope Mapping Protocols,” in Methods in Molecular Biology vol. 66 (Humana Press, Totowa, N.J.). In an exemplary competition assay, immobilized antigen (e.g. CD3) is incubated in a solution comprising a first labeled antibody that binds to the antigen (e.g. V9 antibody) and a second unlabeled antibody that is being tested for its ability to compete with the first antibody for binding to the antigen. The second antibody may be present in a hybridoma supernatant. As a control, immobilized antigen is incubated in a solution comprising the first labeled antibody but not the second unlabeled antibody. After incubation under conditions permissive for binding of the first antibody to the antigen, excess unbound antibody is removed, and the amount of label associated with immobilized antigen is measured. If the amount of label associated with immobilized antigen is substantially reduced in the test sample relative to the control sample, then that indicates that the second antibody is competing with the first antibody for binding to the antigen. See Harlow and Lane (1988) Antibodies: A Laboratory Manual ch.14 (Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y.).

T cell activating bispecific antigen binding molecules prepared as described herein may be purified by art-known techniques such as high performance liquid chromatography, ion exchange chromatography, gel electrophoresis, affinity chromatography, size exclusion chromatography, and the like. The actual conditions used to purify a particular protein will depend, in part, on factors such as net charge, hydrophobicity, hydrophilicity etc., and will be apparent to those having skill in the art. For affinity chromatography purification an antibody, ligand, receptor or antigen can be used to which the T cell activating bispecific antigen binding molecule binds. For example, for affinity chromatography purification of T cell activating bispecific antigen binding molecules of the invention, a matrix with protein A or protein G may be used. Sequential Protein A or G affinity chromatography and size exclusion chromatography can be used to isolate a T cell activating bispecific antigen binding molecule essentially as described in the Examples. The purity of the T cell activating bispecific antigen binding molecule can be determined by any of a variety of well known analytical methods including gel electrophoresis, high pressure liquid chromatography, and the like. For example, the heavy chain fusion proteins expressed as described in the Examples were shown to be intact and properly assembled as demonstrated by reducing SDS-PAGE. Three bands were resolved at approximately Mr 25,000, Mr 50,000 and Mr 75,000, corresponding to the predicted molecular weights of the T cell activating bispecific antigen binding molecule light chain, heavy chain and heavy chain/light chain fusion protein.

Assays

T cell activating bispecific antigen binding molecules provided herein may be identified, screened for, or characterized for their physical/chemical properties and/or biological activities by various assays known in the art.

Affinity Assays

The affinity of the T cell activating bispecific antigen binding molecule for an Fc receptor or a target antigen can be determined in accordance with the methods set forth in the Examples by surface plasmon resonance (SPR), using standard instrumentation such as a BIAcore instrument (GE Healthcare), and receptors or target proteins such as may be obtained by recombinant expression. Alternatively, binding of T cell activating bispecific antigen binding molecules for different receptors or target antigens may be evaluated using cell lines expressing the particular receptor or target antigen, for example by flow cytometry (FACS). A specific illustrative and exemplary embodiment for measuring binding affinity is described in the following and in the Examples below.

According to one embodiment, KD is measured by surface plasmon resonance using a BIACORE® T100 machine (GE Healthcare) at 25° C.

To analyze the interaction between the Fc-portion and Fc receptors, His-tagged recombinant Fc-receptor is captured by an anti-Penta His antibody (Qiagen) (“Penta His” disclosed as SEQ ID NO: 45) immobilized on CM5 chips and the bispecific constructs are used as analytes. Briefly, carboxymethylated dextran biosensor chips (CM5, GE Healthcare) are activated with N-ethyl-N′-(3-dimethylaminopropyl)-carbodiimide hydrochloride (EDC) and N-hydroxysuccinimide (NETS) according to the supplier's instructions. Anti Penta-His antibody (“Penta His” disclosed as SEQ ID NO: 45) is diluted with 10 mM sodium acetate, pH 5.0, to 40 μg/ml before injection at a flow rate of 5 μl/min to achieve approximately 6500 response units (RU) of coupled protein. Following the injection of the ligand, 1 M ethanolamine is injected to block unreacted groups. Subsequently the Fc-receptor is captured for 60 s at 4 or 10 nM. For kinetic measurements, four-fold serial dilutions of the bispecific construct (range between 500 nM and 4000 nM) are injected in HBS-EP (GE Healthcare, 10 mM HEPES, 150 mM NaCl, 3 mM EDTA, 0.05% Surfactant P20, pH 7.4) at 25° C. at a flow rate of 30 μl/min for 120 s.

To determine the affinity to the target antigen, bispecific constructs are captured by an anti human Fab specific antibody (GE Healthcare) that is immobilized on an activated CM5-sensor chip surface as described for the anti Penta-His antibody (“Penta His” disclosed as SEQ ID NO: 45). The final amount of coupled protein is is approximately 12000 RU. The bispecific constructs are captured for 90 s at 300 nM. The target antigens are passed through the flow cells for 180 s at a concentration range from 250 to 1000 nM with a flowrate of 30 μl/min. The dissociation is monitored for 180 s.

Bulk refractive index differences are corrected for by subtracting the response obtained on reference flow cell. The steady state response was used to derive the dissociation constant KD by non-linear curve fitting of the Langmuir binding isotherm. Association rates (kon) and dissociation rates (koff) are calculated using a simple one-to-one Langmuir binding model (BIACORE® T100 Evaluation Software version 1.1.1) by simultaneously fitting the association and dissociation sensorgrams. The equilibrium dissociation constant (KD) is calculated as the ratio koff/kon. See, e.g., Chen et al., J Mol Biol 293, 865-881 (1999).

Activity Assays

Biological activity of the T cell activating bispecific antigen binding molecules of the invention can be measured by various assays as described in the Examples. Biological activities may for example include the induction of proliferation of T cells, the induction of signaling in T cells, the induction of expression of activation markers in T cells, the induction of cytokine secretion by T cells, the induction of lysis of target cells such as tumor cells, and the induction of tumor regression and/or the improvement of survival.

Compositions, Formulations, and Routes of Administration

In a further aspect, the invention provides pharmaceutical compositions comprising any of the T cell activating bispecific antigen binding molecules provided herein, e.g., for use in any of the below therapeutic methods. In one embodiment, a pharmaceutical composition comprises any of the T cell activating bispecific antigen binding molecules provided herein and a pharmaceutically acceptable carrier. In another embodiment, a pharmaceutical composition comprises any of the T cell activating bispecific antigen binding molecules provided herein and at least one additional therapeutic agent, e.g., as described below.

Further provided is a method of producing a T cell activating bispecific antigen binding molecule of the invention in a form suitable for administration in vivo, the method comprising (a) obtaining a T cell activating bispecific antigen binding molecule according to the invention, and (b) formulating the T cell activating bispecific antigen binding molecule with at least one pharmaceutically acceptable carrier, whereby a preparation of T cell activating bispecific antigen binding molecule is formulated for administration in vivo.

Pharmaceutical compositions of the present invention comprise a therapeutically effective amount of one or more T cell activating bispecific antigen binding molecule dissolved or dispersed in a pharmaceutically acceptable carrier. The phrases “pharmaceutical or pharmacologically acceptable” refers to molecular entities and compositions that are generally non-toxic to recipients at the dosages and concentrations employed, i.e. do not produce an adverse, allergic or other untoward reaction when administered to an animal, such as, for example, a human, as appropriate. The preparation of a pharmaceutical composition that contains at least one T cell activating bispecific antigen binding molecule and optionally an additional active ingredient will be known to those of skill in the art in light of the present disclosure, as exemplified by Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, incorporated herein by reference. Moreover, for animal (e.g., human) administration, it will be understood that preparations should meet sterility, pyrogenicity, general safety and purity standards as required by FDA Office of Biological Standards or corresponding authorities in other countries. Preferred compositions are lyophilized formulations or aqueous solutions. As used herein, “pharmaceutically acceptable carrier” includes any and all solvents, buffers, dispersion media, coatings, surfactants, antioxidants, preservatives (e.g. antibacterial agents, antifungal agents), isotonic agents, absorption delaying agents, salts, preservatives, antioxidants, proteins, drugs, drug stabilizers, polymers, gels, binders, excipients, disintegration agents, lubricants, sweetening agents, flavoring agents, dyes, such like materials and combinations thereof, as would be known to one of ordinary skill in the art (see, for example, Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, pp. 1289-1329, incorporated herein by reference). Except insofar as any conventional carrier is incompatible with the active ingredient, its use in the therapeutic or pharmaceutical compositions is contemplated.

The composition may comprise different types of carriers depending on whether it is to be administered in solid, liquid or aerosol form, and whether it need to be sterile for such routes of administration as injection. T cell activating bispecific antigen binding molecules of the present invention (and any additional therapeutic agent) can be administered intravenously, intradermally, intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intraprostatically, intrasplenically, intrarenally, intrapleurally, intratracheally, intranasally, intravitreally, intravaginally, intrarectally, intratumorally, intramuscularly, intraperitoneally, subcutaneously, subconjunctivally, intravesicularlly, mucosally, intrapericardially, intraumbilically, intraocularally, orally, topically, locally, by inhalation (e.g. aerosol inhalation), injection, infusion, continuous infusion, localized perfusion bathing target cells directly, via a catheter, via a lavage, in cremes, in lipid compositions (e.g. liposomes), or by other method or any combination of the forgoing as would be known to one of ordinary skill in the art (see, for example, Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, incorporated herein by reference). Parenteral administration, in particular intravenous injection, is most commonly used for administering polypeptide molecules such as the T cell activating bispecific antigen binding molecules of the invention.

Parenteral compositions include those designed for administration by injection, e.g. subcutaneous, intradermal, intralesional, intravenous, intraarterial intramuscular, intrathecal or intraperitoneal injection. For injection, the T cell activating bispecific antigen binding molecules of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiological saline buffer. The solution may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the T cell activating bispecific antigen binding molecules may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use. Sterile injectable solutions are prepared by incorporating the T cell activating bispecific antigen binding molecules of the invention in the required amount in the appropriate solvent with various of the other ingredients enumerated below, as required. Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and/or the other ingredients. In the case of sterile powders for the preparation of sterile injectable solutions, suspensions or emulsion, the preferred methods of preparation are vacuum-drying or freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered liquid medium thereof. The liquid medium should be suitably buffered if necessary and the liquid diluent first rendered isotonic prior to injection with sufficient saline or glucose. The composition must be stable under the conditions of manufacture and storage, and preserved against the contaminating action of microorganisms, such as bacteria and fungi. It will be appreciated that endotoxin contamination should be kept minimally at a safe level, for example, less that 0.5 ng/mg protein. Suitable pharmaceutically acceptable carriers include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g. Zn-protein complexes); and/or non-ionic surfactants such as polyethylene glycol (PEG). Aqueous injection suspensions may contain compounds which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, dextran, or the like. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl cleats or triglycerides, or liposomes.

Active ingredients may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical Sciences (18th Ed. Mack Printing Company, 1990). Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the polypeptide, which matrices are in the form of shaped articles, e.g. films, or microcapsules. In particular embodiments, prolonged absorption of an injectable composition can be brought about by the use in the compositions of agents delaying absorption, such as, for example, aluminum monostearate, gelatin or combinations thereof.

In addition to the compositions described previously, the T cell activating bispecific antigen binding molecules may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the T cell activating bispecific antigen binding molecules may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.

Pharmaceutical compositions comprising the T cell activating bispecific antigen binding molecules of the invention may be manufactured by means of conventional mixing, dissolving, emulsifying, encapsulating, entrapping or lyophilizing processes. Pharmaceutical compositions may be formulated in conventional manner using one or more physiologically acceptable carriers, diluents, excipients or auxiliaries which facilitate processing of the proteins into preparations that can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.

The T cell activating bispecific antigen binding molecules may be formulated into a composition in a free acid or base, neutral or salt form. Pharmaceutically acceptable salts are salts that substantially retain the biological activity of the free acid or base. These include the acid addition salts, e.g., those formed with the free amino groups of a proteinaceous composition, or which are formed with inorganic acids such as for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric or mandelic acid. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as for example, sodium, potassium, ammonium, calcium or ferric hydroxides; or such organic bases as isopropylamine, trimethylamine, histidine or procaine. Pharmaceutical salts tend to be more soluble in aqueous and other protic solvents than are the corresponding free base forms.

Therapeutic Methods and Compositions

Any of the T cell activating bispecific antigen binding molecules provided herein may be used in therapeutic methods. T cell activating bispecific antigen binding molecules of the invention can be used as immunotherapeutic agents, for example in the treatment of cancers.

For use in therapeutic methods, T cell activating bispecific antigen binding molecules of the invention would be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.

In one aspect, T cell activating bispecific antigen binding molecules of the invention for use as a medicament are provided. In further aspects, T cell activating bispecific antigen binding molecules of the invention for use in treating a disease are provided. In certain embodiments, T cell activating bispecific antigen binding molecules of the invention for use in a method of treatment are provided. In one embodiment, the invention provides a T cell activating bispecific antigen binding molecule as described herein for use in the treatment of a disease in an individual in need thereof. In certain embodiments, the invention provides a T cell activating bispecific antigen binding molecule for use in a method of treating an individual having a disease comprising administering to the individual a therapeutically effective amount of the T cell activating bispecific antigen binding molecule. In certain embodiments the disease to be treated is a proliferative disorder. In a particular embodiment the disease is cancer. In certain embodiments the method further comprises administering to the individual a therapeutically effective amount of at least one additional therapeutic agent, e.g., an anti-cancer agent if the disease to be treated is cancer. In further embodiments, the invention provides a T cell activating bispecific antigen binding molecule as described herein for use in inducing lysis of a target cell, particularly a tumor cell. In certain embodiments, the invention provides a T cell activating bispecific antigen binding molecule for use in a method of inducing lysis of a target cell, particularly a tumor cell, in an individual comprising administering to the individual an effective amount of the T cell activating bispecific antigen binding molecule to induce lysis of a target cell. An “individual” according to any of the above embodiments is a mammal, preferably a human.

In a further aspect, the invention provides for the use of a T cell activating bispecific antigen binding molecule of the invention in the manufacture or preparation of a medicament. In one embodiment the medicament is for the treatment of a disease in an individual in need thereof. In a further embodiment, the medicament is for use in a method of treating a disease comprising administering to an individual having the disease a therapeutically effective amount of the medicament. In certain embodiments the disease to be treated is a proliferative disorder. In a particular embodiment the disease is cancer. In one embodiment, the method further comprises administering to the individual a therapeutically effective amount of at least one additional therapeutic agent, e.g., an anti-cancer agent if the disease to be treated is cancer. In a further embodiment, the medicament is for inducing lysis of a target cell, particularly a tumor cell. In still a further embodiment, the medicament is for use in a method of inducing lysis of a target cell, particularly a tumor cell, in an individual comprising administering to the individual an effective amount of the medicament to induce lysis of a target cell. An “individual” according to any of the above embodiments may be a mammal, preferably a human.

In a further aspect, the invention provides a method for treating a disease. In one embodiment, the method comprises administering to an individual having such disease a therapeutically effective amount of a T cell activating bispecific antigen binding molecule of the invention. In one embodiment a composition is administered to said individual, comprising the T cell activating bispecific antigen binding molecule of the invention in a pharmaceutically acceptable form. In certain embodiments the disease to be treated is a proliferative disorder. In a particular embodiment the disease is cancer. In certain embodiments the method further comprises administering to the individual a therapeutically effective amount of at least one additional therapeutic agent, e.g., an anti-cancer agent if the disease to be treated is cancer. An “individual” according to any of the above embodiments may be a mammal, preferably a human.

In a further aspect, the invention provides a method for inducing lysis of a target cell, particularly a tumor cell. In one embodiment the method comprises contacting a target cell with a T cell activating bispecific antigen binding molecule of the invention in the presence of a T cell, particularly a cytotoxic T cell. In a further aspect, a method for inducing lysis of a target cell, particularly a tumor cell, in an individual is provided. In one such embodiment, the method comprises administering to the individual an effective amount of a T cell activating bispecific antigen binding molecule to induce lysis of a target cell. In one embodiment, an “individual” is a human.

In certain embodiments the disease to be treated is a proliferative disorder, particularly cancer. Non-limiting examples of cancers include bladder cancer, brain cancer, head and neck cancer, pancreatic cancer, lung cancer, breast cancer, ovarian cancer, uterine cancer, cervical cancer, endometrial cancer, esophageal cancer, colon cancer, colorectal cancer, rectal cancer, gastric cancer, prostate cancer, blood cancer, skin cancer, squamous cell carcinoma, bone cancer, and kidney cancer. Other cell proliferation disorders that can be treated using a T cell activating bispecific antigen binding molecule of the present invention include, but are not limited to neoplasms located in the: abdomen, bone, breast, digestive system, liver, pancreas, peritoneum, endocrine glands (adrenal, parathyroid, pituitary, testicles, ovary, thymus, thyroid), eye, head and neck, nervous system (central and peripheral), lymphatic system, pelvic, skin, soft tissue, spleen, thoracic region, and urogenital system. Also included are pre-cancerous conditions or lesions and cancer metastases. In certain embodiments the cancer is chosen from the group consisting of renal cell cancer, skin cancer, lung cancer, colorectal cancer, breast cancer, brain cancer, head and neck cancer. A skilled artisan readily recognizes that in many cases the T cell activating bispecific antigen binding molecule may not provide a cure but may only provide partial benefit. In some embodiments, a physiological change having some benefit is also considered therapeutically beneficial. Thus, in some embodiments, an amount of T cell activating bispecific antigen binding molecule that provides a physiological change is considered an “effective amount” or a “therapeutically effective amount”. The subject, patient, or individual in need of treatment is typically a mammal, more specifically a human.

In some embodiments, an effective amount of a T cell activating bispecific antigen binding molecule of the invention is administered to a cell. In other embodiments, a therapeutically effective amount of a T cell activating bispecific antigen binding molecule of the invention is administered to an individual for the treatment of disease.

For the prevention or treatment of disease, the appropriate dosage of a T cell activating bispecific antigen binding molecule of the invention (when used alone or in combination with one or more other additional therapeutic agents) will depend on the type of disease to be treated, the route of administration, the body weight of the patient, the type of T cell activating bispecific antigen binding molecule, the severity and course of the disease, whether the T cell activating bispecific antigen binding molecule is administered for preventive or therapeutic purposes, previous or concurrent therapeutic interventions, the patient's clinical history and response to the T cell activating bispecific antigen binding molecule, and the discretion of the attending physician. The practitioner responsible for administration will, in any event, determine the concentration of active ingredient(s) in a composition and appropriate dose(s) for the individual subject. Various dosing schedules including but not limited to single or multiple administrations over various time-points, bolus administration, and pulse infusion are contemplated herein.

The T cell activating bispecific antigen binding molecule is suitably administered to the patient at one time or over a series of treatments. Depending on the type and severity of the disease, about 1 μg/kg to 15 mg/kg (e.g. 0.1 mg/kg-10 mg/kg) of T cell activating bispecific antigen binding molecule can be an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion. One typical daily dosage might range from about 1 μg/kg to 100 mg/kg or more, depending on the factors mentioned above. For repeated administrations over several days or longer, depending on the condition, the treatment would generally be sustained until a desired suppression of disease symptoms occurs. One exemplary dosage of the T cell activating bispecific antigen binding molecule would be in the range from about 0.005 mg/kg to about 10 mg/kg. In other non-limiting examples, a dose may also comprise from about 1 microgram/kg body weight, about 5 microgram/kg body weight, about 10 microgram/kg body weight, about 50 microgram/kg body weight, about 100 microgram/kg body weight, about 200 microgram/kg body weight, about 350 microgram/kg body weight, about 500 microgram/kg body weight, about 1 milligram/kg body weight, about 5 milligram/kg body weight, about 10 milligram/kg body weight, about 50 milligram/kg body weight, about 100 milligram/kg body weight, about 200 milligram/kg body weight, about 350 milligram/kg body weight, about 500 milligram/kg body weight, to about 1000 mg/kg body weight or more per administration, and any range derivable therein. In non-limiting examples of a derivable range from the numbers listed herein, a range of about 5 mg/kg body weight to about 100 mg/kg body weight, about 5 microgram/kg body weight to about 500 milligram/kg body weight, etc., can be administered, based on the numbers described above. Thus, one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 5.0 mg/kg or 10 mg/kg (or any combination thereof) may be administered to the patient. Such doses may be administered intermittently, e.g. every week or every three weeks (e.g. such that the patient receives from about two to about twenty, or e.g. about six doses of the T cell activating bispecific antigen binding molecule). An initial higher loading dose, followed by one or more lower doses may be administered. However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.

The T cell activating bispecific antigen binding molecules of the invention will generally be used in an amount effective to achieve the intended purpose. For use to treat or prevent a disease condition, the T cell activating bispecific antigen binding molecules of the invention, or pharmaceutical compositions thereof, are administered or applied in a therapeutically effective amount. Determination of a therapeutically effective amount is well within the capabilities of those skilled in the art, especially in light of the detailed disclosure provided herein.

For systemic administration, a therapeutically effective dose can be estimated initially from in vitro assays, such as cell culture assays. A dose can then be formulated in animal models to achieve a circulating concentration range that includes the IC50 as determined in cell culture. Such information can be used to more accurately determine useful doses in humans.

Initial dosages can also be estimated from in vivo data, e.g., animal models, using techniques that are well known in the art. One having ordinary skill in the art could readily optimize administration to humans based on animal data.

Dosage amount and interval may be adjusted individually to provide plasma levels of the T cell activating bispecific antigen binding molecules which are sufficient to maintain therapeutic effect. Usual patient dosages for administration by injection range from about 0.1 to 50 mg/kg/day, typically from about 0.5 to 1 mg/kg/day. Therapeutically effective plasma levels may be achieved by administering multiple doses each day. Levels in plasma may be measured, for example, by HPLC.

In cases of local administration or selective uptake, the effective local concentration of the T cell activating bispecific antigen binding molecules may not be related to plasma concentration. One having skill in the art will be able to optimize therapeutically effective local dosages without undue experimentation.

A therapeutically effective dose of the T cell activating bispecific antigen binding molecules described herein will generally provide therapeutic benefit without causing substantial toxicity. Toxicity and therapeutic efficacy of a T cell activating bispecific antigen binding molecule can be determined by standard pharmaceutical procedures in cell culture or experimental animals. Cell culture assays and animal studies can be used to determine the LD50 (the dose lethal to 50% of a population) and the ED50 (the dose therapeutically effective in 50% of a population). The dose ratio between toxic and therapeutic effects is the therapeutic index, which can be expressed as the ratio LD50/ED50. T cell activating bispecific antigen binding molecules that exhibit large therapeutic indices are preferred. In one embodiment, the T cell activating bispecific antigen binding molecule according to the present invention exhibits a high therapeutic index. The data obtained from cell culture assays and animal studies can be used in formulating a range of dosages suitable for use in humans. The dosage lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon a variety of factors, e.g., the dosage form employed, the route of administration utilized, the condition of the subject, and the like. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition (see, e.g., Fingl et al., 1975, in: The Pharmacological Basis of Therapeutics, Ch. 1, p. 1, incorporated herein by reference in its entirety).

The attending physician for patients treated with T cell activating bispecific antigen binding molecules of the invention would know how and when to terminate, interrupt, or adjust administration due to toxicity, organ dysfunction, and the like. Conversely, the attending physician would also know to adjust treatment to higher levels if the clinical response were not adequate (precluding toxicity). The magnitude of an administered dose in the management of the disorder of interest will vary with the severity of the condition to be treated, with the route of administration, and the like. The severity of the condition may, for example, be evaluated, in part, by standard prognostic evaluation methods. Further, the dose and perhaps dose frequency will also vary according to the age, body weight, and response of the individual patient.

Other Agents and Treatments

The T cell activating bispecific antigen binding molecules of the invention may be administered in combination with one or more other agents in therapy. For instance, a T cell activating bispecific antigen binding molecule of the invention may be co-administered with at least one additional therapeutic agent. The term “therapeutic agent” encompasses any agent administered to treat a symptom or disease in an individual in need of such treatment. Such additional therapeutic agent may comprise any active ingredients suitable for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other. In certain embodiments, an additional therapeutic agent is an immunomodulatory agent, a cytostatic agent, an inhibitor of cell adhesion, a cytotoxic agent, an activator of cell apoptosis, or an agent that increases the sensitivity of cells to apoptotic inducers. In a particular embodiment, the additional therapeutic agent is an anti-cancer agent, for example a microtubule disruptor, an antimetabolite, a topoisomerase inhibitor, a DNA intercalator, an alkylating agent, a hormonal therapy, a kinase inhibitor, a receptor antagonist, an activator of tumor cell apoptosis, or an antiangiogenic agent.

Such other agents are suitably present in combination in amounts that are effective for the purpose intended. The effective amount of such other agents depends on the amount of T cell activating bispecific antigen binding molecule used, the type of disorder or treatment, and other factors discussed above. The T cell activating bispecific antigen binding molecules are generally used in the same dosages and with administration routes as described herein, or about from 1 to 99% of the dosages described herein, or in any dosage and by any route that is empirically/clinically determined to be appropriate.

Such combination therapies noted above encompass combined administration (where two or more therapeutic agents are included in the same or separate compositions), and separate administration, in which case, administration of the T cell activating bispecific antigen binding molecule of the invention can occur prior to, simultaneously, and/or following, administration of the additional therapeutic agent and/or adjuvant. T cell activating bispecific antigen binding molecules of the invention can also be used in combination with radiation therapy.

Articles of Manufacture

In another aspect of the invention, an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of the disorders described above is provided. The article of manufacture comprises a container and a label or package insert on or associated with the container. Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc. The containers may be formed from a variety of materials such as glass or plastic. The container holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). At least one active agent in the composition is a T cell activating bispecific antigen binding molecule of the invention. The label or package insert indicates that the composition is used for treating the condition of choice. Moreover, the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises a T cell activating bispecific antigen binding molecule of the invention; and (b) a second container with a composition contained therein, wherein the composition comprises a further cytotoxic or otherwise therapeutic agent. The article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the compositions can be used to treat a particular condition. Alternatively, or additionally, the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.

Screening Methods

Described herein is the advantageous efficiency of using a single light chain or highly homologous variants thereof for methods of identifying appropriate heavy chain variable regions to construct a T cell activating bispecific antigen binding molecules. To construct these binders, the light chain not only has to be able to pair with various heavy chains to produce binding moieties of different specificity but also retain the ability to form a binding moiety that can activate the T cells to which it binds.

The light chain described herein can be used as a common light chain (CLC) for identifying appropriate heavy chain variable regions, e.g., by screeining a library of heavy chan variable regions. This allows for maintaining the previously identified and validated CD3 binder such that merely a new target antigen binder for the target antigen binding moiety of a T cell activating bispecific antigen binding molecules has to be identified.

Accordingly, in another aspect, the invention provides for a method for identifying a variable heavy chain for use in a bispecific antigen binding molecule specific for a T cell activation antigen and a target cell antigen, comprising the step of screening a combinatorial library comprising variable heavy chains with a light chain comprising the amino acid sequence of SEQ ID NO: 32, SEQ ID NO: 33 and SEQ ID NO: 34. In one embodiment, the light chain comprises the amino acid sequence of SEQ ID NO: 31. In one embodiment, the light chain comprises the amino acid sequence of SEQ ID NO: 35. This method can be used to develop stable, functional, high affinity binders to improve production of T cell activating bispecific antigen binding molecules with, e.g., CD3 specificity and a target antigen specificity, where the target antigens are unrelated, e.g., FolR1, MUC1, and BCMA.

Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, the descriptions and examples should not be construed as limiting the scope of the invention. The disclosures of all patent and scientific literature cited herein are expressly incorporated in their entirety by reference.

EXAMPLES

General Methods

Recombinant DNA Techniques

Standard methods were used to manipulate DNA as described in Sambrook et al., Molecular cloning: A laboratory manual; Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1989. The molecular biological reagents were used according to the manufacturers' instructions. General information regarding the nucleotide sequences of human immunoglobulins light and heavy chains is given in: Kabat, E. A. et al., (1991) Sequences of Proteins of Immunological Interest, 5th ed., NIH Publication No. 91-3242.

DNA Sequencing

DNA sequences were determined by standard double strand sequencing at Synergene (Schlieren).

Gene Synthesis

Desired gene segments where required were either generated by PCR using appropriate templates or were synthesized by Geneart AG (Regensburg, Germany) from synthetic oligonucleotides and PCR products by automated gene synthesis. In cases where no exact gene sequence was available, oligonucleotide primers were designed based on sequences from closest homologues and the genes were isolated by RT-PCR from RNA originating from the appropriate tissue. The gene segments flanked by singular restriction endonuclease cleavage sites were cloned into standard cloning/sequencing vectors. The plasmid DNA was purified from transformed bacteria and concentration determined by UV spectroscopy. The DNA sequence of the subcloned gene fragments was confirmed by DNA sequencing. Gene segments were designed with suitable restriction sites to allow sub-cloning into the respective expression vectors. All constructs were designed with a 5′-end DNA sequence coding for a leader peptide which targets proteins for secretion in eukaryotic cells.

Isolation of Primary Human Pan T Cells from PBMCs

Peripheral blood mononuclear cells (PBMCs) were prepared by Histopaque density centrifugation from enriched lymphocyte preparations (buffy coats) obtained from local blood banks or from fresh blood from healthy human donors. Briefly, blood was diluted with sterile PBS and carefully layered over a Histopaque gradient (Sigma, H8889). After centrifugation for 30 minutes at 450×g at room temperature (brake switched off), part of the plasma above the PBMC containing interphase was discarded. The PBMCs were transferred into new 50 ml Falcon tubes and tubes were filled up with PBS to a total volume of 50 ml. The mixture was centrifuged at room temperature for 10 minutes at 400×g (brake switched on). The supernatant was discarded and the PBMC pellet washed twice with sterile PBS (centrifugation steps at 4° C. for 10 minutes at 350×g). The resulting PBMC population was counted automatically (ViCell) and stored in RPMI1640 medium, containing 10% FCS and 1% L-alanyl-L-glutamine (Biochrom, K0302) at 37° C., 5% CO2 in the incubator until assay start.

T cell enrichment from PBMCs was performed using the Pan T Cell Isolation Kit II (Miltenyi Biotec #130-091-156), according to the manufacturer's instructions. Briefly, the cell pellets were diluted in 40 μl cold buffer per 10 million cells (PBS with 0.5% BSA, 2 mM EDTA, sterile filtered) and incubated with 10 μl Biotin-Antibody Cocktail per 10 million cells for 10 min at 4° C. 30 μl cold buffer and 20 μl Anti-Biotin magnetic beads per 10 million cells were added, and the mixture incubated for another 15 min at 4° C. Cells were washed by adding 10-20× the current volume and a subsequent centrifugation step at 300×g for 10 min. Up to 100 million cells were resuspended in 500 μl buffer. Magnetic separation of unlabeled human pan T cells was performed using LS columns (Miltenyi Biotec #130-042-401) according to the manufacturer's instructions. The resulting T cell population was counted automatically (ViCell) and stored in AIM-V medium at 37° C., 5% CO2 in the incubator until assay start (not longer than 24 h).

Isolation of Primary Human Naive T Cells from PBMCs

Peripheral blood mononuclar cells (PBMCs) were prepared by Histopaque density centrifugation from enriched lymphocyte preparations (buffy coats) obtained from local blood banks or from fresh blood from healthy human donors. T-cell enrichment from PBMCs was performed using the Naive CD8+ T cell isolation Kit from Miltenyi Biotec (#130-093-244), according to the manufacturer's instructions, but skipping the last isolation step of CD8+ T cells (also see description for the isolation of primary human pan T cells).

Isolation of Murine Pan T Cells from Splenocytes

Spleens were isolated from C57BL/6 mice, transferred into a GentleMACS C-tube (Miltenyi Biotech #130-093-237) containing MACS buffer (PBS+0.5% BSA+2 mM EDTA) and dissociated with the GentleMACS Dissociator to obtain single-cell suspensions according to the manufacturer's instructions. The cell suspension was passed through a pre-separation filter to remove remaining undissociated tissue particles. After centrifugation at 400×g for 4 min at 4° C., ACK Lysis Buffer was added to lyse red blood cells (incubation for 5 min at room temperature). The remaining cells were washed with MACS buffer twice, counted and used for the isolation of murine pan T cells. The negative (magnetic) selection was performed using the Pan T Cell Isolation Kit from Miltenyi Biotec (#130-090-861), following the manufacturer's instructions. The resulting T cell population was automatically counted (ViCell) and immediately used for further assays.

Isolation of Primary Cynomolgus PBMCs from Heparinized Blood

Peripheral blood mononuclar cells (PBMCs) were prepared by density centrifugation from fresh blood from healthy cynomolgus donors, as follows: Heparinized blood was diluted 1:3 with sterile PBS, and Lymphoprep medium (Axon Lab #1114545) was diluted to 90% with sterile PBS. Two volumes of the diluted blood were layered over one volume of the diluted density gradient and the PBMC fraction was separated by centrifugation for 30 min at 520×g, without brake, at room temperature. The PBMC band was transferred into a fresh 50 ml Falcon tube and washed with sterile PBS by centrifugation for 10 min at 400×g at 4° C. One low-speed centrifugation was performed to remove the platelets (15 min at 150×g, 4° C.), and the resulting PBMC population was automatically counted (ViCell) and immediately used for further assays.

Exemplary Antigen Generation

The antigen is expressed in two different versions. For a non-Fc containing construct the extracellular domain is fused to an avi-His tag attached to its C-terminus. For an Fc containing antigen an Fc fusion using the knob-into-hole technology to the N-terminus of a heterodimeric Fc part is applied (Merchant et al.) to have only one molecule of the protein of interest per Fc dimer. This is done to avoid the formation of any artificial dimeric structures of the protein of interest. The avi-tag is here attached to the C-terminus of the Fc. Those proteins can easily be transiently expressed in mammalian cells like HEK or CHO, purified via ProteinA chromatography, biotinylted and can be attached to streptavidin beads for phage selections via the biotinylated avi-tag according to standard methods.

Affinity Maturation

Affinity maturation of the Fab fragments coming out of a CLC-library has to be limited to the heavy chain only, in order to retain the light chain in an unchanged manner to have the CD3e binding affinity retained. Since our initial library is randomized in the CDR3 of the heavy chain only, we focus for the maturation step on the CDRs 1 and 2. For this we design PCR primers for each framework that will introduce randomization in to CDR1 and CDR2 separately. Each primer is designed to bind to one of the six heavy chain frameworks and can be used in a generic way for each antibody clone coming from that particular phage library. The process of maturation is carried out as described by Knappik, or by Steidl (Knappik et al., J. Mol. Biol. (2000) 296, 57-86). S. Steidl et al.; Molecular Immunology 46 (2008) 135-144). Phage panning is carried out as described above, with the difference that the concentrations of the soluble antigen is used at 2×10̂−8M and is decreased to a final concentration to 2×10̂−10M.

Cloning, Production, Purification and Biochemical Characterization of CLC TCB

The resulting variable region of heavy and light chain DNA sequences are subcloned in frame with either the constant heavy chain or the constant light chain pre-inserted into the respective recipient mammalian expression vector. The antibody expression is driven by an MPSV promoter and carries a synthetic polyA signal sequence at the 3′ end of the CDS. In addition each vector contains an EBV OriP sequence for transient expression in HEK293-EBNA cells. As antibody isotypes IgG1 P329G LALA or IgG4 SPLE PG are used.

The CLC TCB is produced by co-transfecting HEK293-EBNA cells with the mammalian expression vectors using polyethylenimine. The cells are transfected with the corresponding expression vectors in a 1:1:3 ratio (“vector heavy chain Fc(hole)”: “vector heavy chain Fc(knob)-FabCrossfab”): “vector common light chain”.

For transfection HEK293 EBNA cells are cultivated in suspension serum free in CD CHO culture medium. For the production in 500 ml shake flask 400 million HEK293 EBNA cells are seeded 24 hours before transfection. For transfection cells are centrifuged for 5 min by 210×g, supernatant is replaced by pre-warmed 20 ml CD CHO medium. Expression vectors are mixed in 20 ml CD CHO medium to a final amount of 200 g DNA. After addition of 540 μl PEI solution is vortexed for 15 s and subsequently incubated for 10 min at room temperature. Afterwards cells are mixed with the DNA/PEI solution, transferred to a 500 ml shake flask and incubated for 3 hours by 37° C. in an incubator with a 5% CO2 atmosphere. After incubation time 160 ml F17 medium is added and cell are cultivated for 24 hours. One day after transfection 1 mM valporic acid and 7% Feed 1 is added. After 7 days cultivation supernatant is collected for purification by centrifugation for 15 min at 210×g, the solution is sterile filtered (0.22 μm filter) and sodium azide in a final concentration of 0.01% w/v is added, and kept at 4° C.

The secreted protein is purified from cell culture supernatants by affinity chromatography using ProteinA. Supernatant is loaded on a HiTrap ProteinA HP column (CV=5 mL, GE Healthcare) equilibrated with 40 ml 20 mM sodium phosphate, 20 mM sodium citrate, 0.5 M sodium chloride, pH 7.5. Unbound protein is removed by washing with at least 10 column volume 20 mM sodium phosphate, 20 mM sodium citrate, 0.5 M sodium chloride, pH 7.5. Target protein is eluted during a gradient over 20 column volume from 20 mM sodium citrate, 0.5 M sodium chloride, pH 7.5 to 20 mM sodium citrate, 0.5 M sodium chloride, pH 2.5. Protein solution is neutralized by adding 1/10 of 0.5 M sodium phosphate, pH 8. Target protein is concentrated and filtrated prior loading on a HiLoad Superdex 200 column (GE Healthcare) equilibrated with 20 mM Histidine, 140 mM sodium chloride solution of pH 6.0.

The protein concentration of purified protein samples is determined by measuring the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence.

Purity and molecular weight of molecules are analyzed by CE-SDS analyses in the presence and absence of a reducing agent. The Caliper LabChip GXII system (Caliper lifescience) is used according to the manufacturer's instruction. 2ug sample is used for analyses.

The aggregate content of antibody samples is analyzed using a TSKgel G3000 SW XL analytical size-exclusion column (Tosoh) in 25 mM K2HPO4, 125 mM NaCl, 200 mM L-Arginine Monohydrocloride, 0.02% (w/v) NaN3, pH 6.7 running buffer at 25° C.

CLC TCB Characterization in Cell-Based Assays

Binding of CLC TCB to ECD Tumor Antigen- and CD3-Expressing Cells

The binding of CLC TCB is tested using tumor cells expressing the antigen X of interest and a CD3e-expressing immortalized T lymphocyte line (Jurkat). Briefly, cells are harvested, counted, checked for viability and resuspended at 2×106 cells/ml in FACS buffer (100 μl PBS 0.1% BSA). 100 μl of cell suspension (containing 0.2×106 cells) is incubated in round-bottom 96-well plate for 30 min at 4° C. with increasing concentrations of the CLC TCB (3 pM-200 nM), washed twice with cold PBS 0.1% BSA, re-incubated for further 30 min at 4° C. with the PE-conjugated AffiniPure F(ab′)2 Fragment goat anti-human IgG Fcg Fragment Specific secondary antibody (Jackson Immuno Research Lab PE #109-116-170), washed twice with cold PBS 0.1% BSA and immediately analyzed by FACS using a FACS CantoII (Software FACS Diva) by gating live, DAPI-negative, cells. Binding curves are obtained using GraphPadPrism5.

Example 1 Purification of Biotinylated Folate Receptor-Fc Fusions

To generate new antibodies against human FolR1 the following antigens and screening tools were generated as monovalent Fc fusion proteins (the extracellular domain of the antigen linked to the hinge region of Fc-knob which is co-expressed with an Fc-hole molecule). The antigen genes were synthesized (Geneart, Regensburg, Germany) based on sequences obtained from GenBank or SwissProt and inserted into expression vectors to generate fusion proteins with Fc-knob with a C-terminal Avi-tag for in vivo or in vitro biotinylation. In vivo biotinylation was achieved by co-expression of the bacterial birA gene encoding a bacterial biotin ligase during production. Expression of all genes was under control of a chimeric MPSV promoter on a plasmid containing an oriP element for stable maintenance of the plasmids in EBNA containing cell lines.

For preparation of the biotinylated monomeric antigen/Fc fusion molecules, exponentially growing suspension HEK293 EBNA cells were co-transfected with three vectors encoding the two components of fusion protein (knob and hole chains) as well as BirA, an enzyme necessary for the biotinylation reaction. The corresponding vectors were used at a 9.5:9.5:1 ratio (“antigen ECD-Fc knob-avi tag”: “Fc hole”: “BirA”).

For protein production in 500 ml shake flasks, 400 million HEK293 EBNA cells were seeded 24 hours before transfection. For transfection cells were centrifuged for 5 minutes at 210 g, and supernatant was replaced by pre-warmed CD CHO medium. Expression vectors were resuspended in 20 mL of CD CHO medium containing 200 μg of vector DNA. After addition of 540 μL of polyethylenimine (PEI), the solution was mixed for 15 seconds and incubated for 10 minutes at room temperature. Afterwards, cells were mixed with the DNA/PEI solution, transferred to a 500 mL shake flask and incubated for 3 hours at 37° C. in an incubator with a 5% CO2 atmosphere. After the incubation, 160 mL of F17 medium was added and cells were cultured for 24 hours. One day after transfection, 1 mM valproic acid and 7% Feed 1 (Lonza) were added to the culture. The production medium was also supplemented with 100 μM biotin. After 7 days of culturing, the cell supernatant was collected by spinning down cells for 15 min at 210 g. The solution was sterile filtered (0.22 μm filter), supplemented with sodium azide to a final concentration of 0.01% (w/v), and kept at 4° C.

Secreted proteins were purified from cell culture supernatants by affinity chromatography using Protein A, followed by size exclusion chromatography. For affinity chromatography, the supernatant was loaded on a HiTrap ProteinA HP column (CV=5 mL, GE Healthcare) equilibrated with 40 mL 20 mM sodium phosphate, 20 mM sodium citrate pH 7.5. Unbound protein was removed by washing with at least 10 column volumes of 20 mM sodium phosphate, 20 mM sodium citrate pH 7.5. The bound protein was eluted using a linear pH-gradient created over 20 column volumes of 20 mM sodium citrate, 100 mM sodium chloride, 100 mM glycine, pH 3.0. The column was then washed with 10 column volumes of 20 mM sodium citrate, 100 mM sodium chloride, 100 mM glycine, pH 3.0.

pH of collected fractions was adjusted by adding 1/10 (v/v) of 0.5 M sodium phosphate, pH 8.0. The protein was concentrated and filtered prior to loading on a HiLoad Superdex 200 column (GE Healthcare) equilibrated with 20 mM histidine, 140 mM sodium chloride, pH 6.0.

The protein concentration was determined by measuring the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence. Purity and molecular weight of the FolR1-Fc-fusion was analyzed by SDS capillary electrophoresis in the presence and absence of a reducing agent following the manufacturer instructions (instrument Caliper LabChipGX, Perkin Elmer). The aggregate content of samples was analyzed using a TSKgel G3000 SW XL analytical size-exclusion column (Tosoh) equilibrated in 25 mM K2HPO4, 125 mM NaCl, 200 mM L-arginine monohydrochloride, 0.02% (w/v) NaN3, pH 6.7 running buffer at 25° C.

Purified antigen-Fc-fusion proteins were analyzed by surface plasmon resonance assays using commercially available antibodies to confirm correct and natural conformation of the antigens (data not shown).

TABLE 1 Antigens produced for isolation, selection and counter selection of human FolR1 antibodies ECD Accession Seq ID Antigen (aa) number Sequence No human 25-234 P15328 RIAWARTELLNVCMNAKHHKEKPGPEDKLHEQCRPWR 227 FolR1 KNACCSTNTSQEAHKDVSYLYRFNWNHCGEMAPACKR HFIQDTCLYECSPNLGPWIQQVDQSWRKERVLNVPLC KEDCEQWWEDCRTSYTCKSNWHKGWNWTSGFNKCAVG AACQPFHFYFPTPTVLCNEIWTHSYKVSNYSRGSGRC IQMWFDPAQGNPNEEVARFYAAAM human 17-230 P14207 TMCSAQDRTDLLNVCMDAKHHKTKPGPEDKLHDQCSP 228 FolR2 WKKNACCTASTSQELHKDTSRLYNFNWDHCGKMEPAC KRHFIQDTCLYECSPNLGPWIQQVNQSWRKERFLDVP LCKEDCQRWWEDCHTSHTCKSNWHRGWDWTSGVNKCP AGALCRTFESYFPTPAALCEGLWSHSYKVSNYSRGSG RCIQMWFDSAQGNPNEEVARFYAAAMHVN human 24-243 P41439 SARARTDLLNVCMNAKHHKTQPSPEDELYGQCSPWKK 229 FolR3 NACCTASTSQELHKDTSRLYNFNWDHCGKMEPTCKRH FIQDSCLYECSPNLGPWIRQVNQSWRKERILNVPLCK EDCERWWEDCRTSYTCKSNWHKGWNWTSGINECPAGA LCSTFESYFPTPAALCEGLWSHSFKVSNYSRGSGRCI QMWFDSAQGNPNEEVAKFYAAAMNAGAPSRGIIDS murine 25-232 P35846 TRARTELLNVCMDAKHHKEKPGPEDNLHDQCSPWKTN 230 FolR1 SCCSTNTSQEAHKDISYLYRFNWNHCGTMTSECKRHF IQDTCLYECSPNLGPWIQQVDQSWRKERILDVPLCKE DCQQWWEDCQSSFTCKSNWHKGWNWSSGHNECPVGAS CHPFTFYFPTSAALCEEIWSHSYKLSNYSRGSGRCIQ MWFDPAQGNPNEEVARFYAEAMS cynomolgus 25-234 G7PR14 EAQTRTARARTELLNVCMNAKHHKEKPGPEDKLHEQC 231 FolR1 RPWKKNACCSTNTSQEAHKDVSYLYRFNWNHCGEMAP ACKRHFIQDTCLYECSPNLGPWIQQVDQSWRKERVLN VPLCKEDCERWWEDCRTSYCKSNWHKGWNWTSGFNKC PVGAACQPFHFYFPTPTVLCNEIWTYSYKVSNYSRGS GRCIQMWFDPAQGNPNEEVARFYAAAMS

TABLE 2 Summary of the yield and final monomer content of the FolR-Fc-fusions. Monomer [%] Antigen (SEC) Yield huFolR1 100 30 mg/L cyFolR1 100 32 mg/L muFolR1 100 31 mg/L huFolR2 100 16 mg/L huFolR3 95 38 mg/L

Example 2 Generation of Common Light Chain with CD3* Specificity

The T cell activating bispecific molecules described herein comprise at least one CD3 binding moiety. This moiety can be generated by immunizing laboratory animals, screening phage library or using known anti-CD3 antibodies. The common light chain with CD3ε specificity was generated by humanizing the light chain of a murine parental anti-CD3ε antibody (CH2527). For humanization of an antibody of non-human origin, the CDR residues from the non-human antibody (donor) have to be transplanted onto the framework of a human (acceptor) antibody. Generally, acceptor framework sequences are selected by aligning the sequence of the donor to a collection of potential acceptor sequences and choosing one that has either reasonable homology to the donor, or shows similar amino acids at some positions critical for structure and activity. In the present case, the search for the antibody acceptor framework was performed by aligning the mouse VL-domain sequence of the parental antibody to a collection of human germline sequences and choosing the human sequence that showed high sequence identity. Surprisingly, a good match in terms of framework sequence homology was found in a rather infrequent human light chain belonging to the V-domain family 7 of the lambda type, more precisely, hVL_7_46 (IMGT nomenclature, GenBank Acc No. Z73674). This infrequent human light chain was subsequently chosen as acceptor framework for humanization of the light chain of CH2527. The three complementarity determining regions (CDRs) of the mouse light chain variable domain were grafted onto this acceptor framework. Since the framework 4 region is not part of the variable region of the germline V-gene, the alignment for this region (J-element) was done individually. Hence the IGLJ3-02 sequence was chosen for humanization of this light chain. Thirteen humanized variants were generated (CH2527-VL7_46-1 to VL7_46-10, VL7_46-12 to VL7_46-14). These differ in framework residues (and combinations thereof) that were back-mutated to the murine V-domain sequence or in CDR-residues (Kabat definition) that could be kept identical to the human germline sequence. The following framework residues outside the CDRs were back-mutated to the murine residues in the final humanized VL-domain variant VL7_46-13 (murine residues listed): V36, E38, F44, G46, G49, and G57, respectively. The human J-element IGLJ3-02 was 100% identical to the J-element of the murine parental antibody.

Example 3 SPR Assessment of Humanized Variants with CD3* Specificity

Humanized VL variants were assessed as chimera in a 2+1 classical format (FIG. 1D), i.e. humanized light chain V-domains were paired with murine heavy chain V-domains. SPR assessment was carried out on a ProteOn XPR36 instrument (Bio-Rad). More precisely, the variants were captured directly from the culture supernatant on an anti-Fab derivatized GLM sensorchip (Goat Anti-Human IgG, F(ab′)2 Fragment Specific, Jackson ImmunoResearch) in vertical orientation. The following analytes were subsequently injected horizontally as single concentrations to assess binding to human and cynomolgus CD3ε: 3 μM hu CD3ε(−1-26)-Fc(knob)-avi (ID807) and 2.5 μM cy CD3ε-(−1-26)-Fc(knob)-Avi-Fc(hole) (ID873), respectively. Binding responses were qualitatively compared to binding of the murine control construct and graded + (comparable binding observed), +/− (reduced binding observed) and—(no binding observed). The capture antibody was regenerated after each cycle of ligand capture and analyte binding and the murine construct was re-injected at the end of the study to confirm the activity of the capture surface. The results are summarized in Table 3.

TABLE 3 Qualitative binding assessment based on SPR for the humanized light chain variants combined with the murine heavy chain of CH2527. Only the humanized light chain variant that was finally chosen, CH2527-VL7_46-13, highlighted in bold letters, exhibited comparable binding to human and cynomolgus CD3ε. humanized VL variant binding to CD3ε murine_CH2527-VL + CH12527-VL7_46-1 CH12527-VL7_46-2 CH12527-VL7_46-3 CH12527-VL7_46-4 CH12527-VL7_46-5 CH12527-VL7_46-6 CH12527-VL7_46-7 CH12527-VL7_46-8 CH12527-VL7_46-9 CH12527-VL7_46-10 CH12527-VL7_46-12 +/- CH252741-VL7_46-13 + CH12527-VL7_46-14

Example 4 Properties of Humanized Common Light Chain with CD3* Specificity

The light chain V-domain variant that was chosen for the humanized lead molecule is VL7_46-13. The degree of humanness, i.e. the sequence homology of the humanized V-domain to the human germline V-domain sequence was determined. For VL7_46-13, the overall sequence identity with the closest human germline homolog is 65% before humanization and 80% afterwards. Omitting the CDR regions, the sequence identity is 92% to the closest human germline homolog. As can be seen from Table 3, VL7_46-13 is the only humanized VL variant out of a panel of 13 variants that showed comparable binding to the parental murine antibody and also retained its cross-reactivity to cynomolgus CD3ε. This result indicates that it was not trivial to humanize the murine VL-domain without losing binding affinity to CD3ε which required several back-mutations to murine framework residues (in particular G46) while retaining G24 in CDR1. In addition, this result shows that the VL-domain plays a crucial role in target recognition. Importantly, the humanized VL-domain VL7_46-13 based on an infrequent human germline belonging to the V-domain family 7 of the lambda type and retaining affinity and specificity for CD3ε, is also suitable to be used as a common light chain in phage-displayed antibody libraries of the Fab-format and enables successful selection for novel specificities which greatly facilitates the generation and production of bispecific molecules binding to CD3ε and e.g. a tumor target and sharing the same ‘common’ light chain.

Example 5 Generation of a Phage Displayed Antibody Library Using a Human Germ-Line Common Light Chain Derived from HVK1-39

Several approaches to generate bispecific antibodies that resemble full length human IgG utilize modifications in the Fc region that induce heterodimerization of two distinct heavy chains. Such examples include knobs-into-holes (Merchant et al., Nat Biotechnol. 1998 July; 16(7):677-81) SEED (Davis et al., Protein Eng Des Sel. 2010 April; 23(4):195-202) and electrostatic steering technologies (Gunasekaran et al., J Biol Chem. 2010 Jun. 18; 285(25):19637-46). Although these approaches enable effective heterodimerization of two distinct heavy chains, appropriate pairing of cognate light and heavy chains remains a problem. Usage of a common light chain (LC) can solve this issue (Merchant, et al. Nat Biotech 16, 677-681 (1998)).

Here, we describe the generation of an antibody library for the display on a M13 phage.

Essentially, we designed a multi framework library for the heavy chain with one constant (or “common”) light chain. This library is designed for generating multispecific antibodies without the need to use sophisticated technologies to avoid light chain mispairing.

By using a common light chain the production of these molecules can be facilitated as no mispairing occurs any longer and the isolation of a highly pure bispecific antibody is facilitated. As compared to other formats the use of Fab fragments as building blocks as opposed to e.g. the use of scFv fragments results in higher thermal stability and the lack of scFv aggregation and intermolecular scFv formation.

Library Generation

In the following the generation of an antibody library for the display on M13 phage is described. Essentially, we designed a multi framework library for the heavy chain with one constant (or “common”) light chain.

We used these heavy chains in the library (GenBank Accession Numbers in brackets):

IGHV1-46*01 (X92343) (SEQ ID NO:104),

IGHV1-69*06 (L22583), (SEQ ID NO:105)

IGHV3-15*01 (X92216), (SEQ ID NO:106)

IGHV3-23*01 (M99660), (SEQ ID NO:107)

IGHV4-59*01 (AB019438), (SEQ ID NO:108)

IGHV5-51*01 (M99686), (SEQ ID NO:109)

All heavy chains use the IGHJ2 as J-element, except the IGHV1-69*06 which uses IGHJ6 sequence. The design of the randomization included the CDR-H1, CDR-H2, and CDR-H3. For CDR-H1 and CDR-H2 a “soft” randomization strategy was chosen, and the randomization oligonucleotides were such that the codon for the amino acid of the germ-line sequence was present at 50%. All other amino acids, except cysteine, were summing up for the remaining 50%. In CDR-H3, where no germ-line amino acid is present due to the presence of the genetic D-element, oligonucleotides were designed that allow for the usage of randomized inserts between the V-element and the J-element of 4 to 9 amino acids in length. Those oligonucleotides contained in their randomized part e.g. The three amino acids G/Y/S are present to 15% each, those amino acids A/D/T/R/P/L/V/N/W/F/I/E are present to 4,6% each.

Exemplary methods for generation of antibody libraries are described in Hoogenboom et al., Nucleic Acids Res. 1991, 19, 4133-413; Lee et., al J. Mol. Biol. (2004) 340, 1073-1093.

The light chain is derived from the human sequence hVK1-39, and is used in an unmodified and non-randomized fashion. This will ensure that the same light chain can be used for other projects without additional modifications.

Exemplary Library Selection:

Selections with all affinity maturation libraries are carried out in solution according to the following procedure using a monomeric and biotinylated extracellular domain of a target antigen X.

1. 10̂12 phagemid particles of each library are bound to 100 nM biotinylated soluble antigen for 0.5 h in a total volume of 1 ml. 2. Biotinylated antigen is captured and specifically bound phage particles are isolated by addition of ˜5×10̂7 streptavidin-coated magnetic beads for 10 min. 3. Beads are washed using 5-10× 1 ml PBS/Tween20 and 5-10× 1 ml PBS. 4. Elution of phage particles is done by addition of 1 ml 100 mM TEA (triethylamine) for 10 min and neutralization by addition of 500 ul 1M Tris/HCl pH 7.4 and 5. Re-infection of exponentially growing E. coli TG1 bacteria, infection with helper phage VCSM13 and subsequent PEG/NaCl precipitation of phagemid particles is applied in subsequent selection rounds. Selections are carried out over 3-5 rounds using either constant or decreasing (from 10̂−7M to 2×10̂−9M) antigen concentrations. In round 2, capture of antigen/phage complexes is performed using neutravidin plates instead of streptavidin beads. All binding reactions are supplemented either with 100 nM bovine serum albumin, or with non-fat milk powder in order to compete for unwanted clones arising from mere sticky binding of the antibodies to the plastic support.

Selections are being carried out over three or four rounds using decreasing antigen concentrations of the antigen starting from 100 nM and going down to 5 nM in the final selection round. Specific binders are defined as signals ca. 5× higher than background and are identified by ELISA. Specific binders are identified by ELISA as follows: 100 μl of 10 nM biotinylated antigen per well are coated on neutravidin plates. Fab-containing bacterial supernatants are added and binding Fabs are detected via their Flag-tags by using an anti-Flag/HRP secondary antibody. ELISA-positive clones are bacterially expressed as soluble Fab fragments in 96-well format and supernatants are subjected to a kinetic screening experiment by SPR-analysis using ProteOn XPR36 (BioRad). Clones expressing Fabs with the highest affinity constants are identified and the corresponding phagemids are sequenced. For further characterization, the Fab sequences are amplified via PCR from the phagemid and cloned via appropriate restriction sites into human IgG1 expression vectors for mammalian production.

Generation of a Phage Displayed Antibody Library Using a Humanized CD3ε Specific Common Light Chain

Here, the generation of an antibody library for the display on M13 phage is described.

Essentially, we designed a multi framework library for the heavy chain with one constant (or “common”) light chain. This library was designed for the generation of Fc-containing, but FcgR binding inactive T cell bispecific antibodies of IgG1 P329G LALA or IgG4 SPLE PG isotype in which one or two Fab recognize a tumor surface antigen expressed on a tumor cell whereas the remaining Fab arm of the antibody recognizes CD3e on a T cell.

Library Generation

In the following the generation of an antibody library for the display on M13 phage is described. Essentially, we designed a multi framework library for the heavy chain with one constant (or “common”) light chain. This library is designed solely for the generation of Fc-containing, but FcgR binding inactive T cell bispecific antibodies of IgG1 P329G LALA or IgG4 SPLE PG isotype.

Diversity was introduced via randomization oligonucleotides only in the CDR3 of the different heavy chains. Methods for generation of antibody libraries are well known in the art and are described in (Hoogenboom et al., Nucleic Acids Res. 1991, 19, 4133-413; or in: Lee et., al J. Mol. Biol. (2004) 340, 1073-1093).

We used these heavy chains in the library:

IGHV1-46*01 (X92343), (SEQ ID NO:104)

IGHV1-69*06 (L22583), (SEQ ID NO:105)

IGHV3-15*01 (X92216), (SEQ ID NO:106)

IGHV3-23*01 (M99660), (SEQ ID NO:107)

IGHV4-59*01 (AB019438), (SEQ ID NO:108)

IGHV5-51*01 (M99686), (SEQ ID NO:109)

We used the light chain derived from the humanized human and Cynomolgus CD3ε specific antibody CH2527 in the library: (VL7_46-13; SEQ ID NO:112). This light chain was not randomized and used without any further modifications in order to ensure compatibility with different bispecific binders.

All heavy chains use the IGHJ2 as J-element, except the IGHV1-69*06 which uses IGHJ6 sequence. The design of the randomization focused on the CDR-H3 only, and PCR oligonucleotides were designed that allow for the usage of randomized inserts between the V-element and the J-element of 4 to 9 amino acids in length.

Example 6 Selection of Antibody Fragments from Common Light Chain Libraries (Comprising Light Chain with CD3* Specificity) to FolR1

The antibodies 16A3, 15A1, 18D3, 19E5, 19A4, 15H7, 15B6, 16D5, 15E12, 21D1, 16F12, 21A5, 21G8, 19H3, 20G6, and 20H7 comprising the common light chain VL7_46-13 with CD3ε specificity were obtained by phage display selections against different species (human, cynomolgus and murine) of FolR1. Clones 16A3, 15A1, 18D3, 19E5, 19A4, 15H7, 15B6, 21D1, 16F12, 19H3, 20G6, and 20H7 were selected from a sub-library in which the common light chain was paired with a heavy chain repertoire based on the human germline VH1_46. In this sub-library, CDR3 of VH1_46 has been randomized based on 6 different CDR3 lengths. Clones 16D5, 15E12, 21A5, and 21G8 were selected from a sub-library in which the common light chain was paired with a heavy chain repertoire based on the human germline VH3_15. In this sub-library, CDR3 of VH3_15 has been randomized based on 6 different CDR3 lengths. In order to obtain species cross-reactive (or murine FolR1-reactive) antibodies, the different species of FolR1 were alternated (or kept constant) in different ways over 3 rounds of biopanning: 16A3 and 15A1 (human-cynomolgus-human FolR1); 18D3 (cynomolgus-human-murine FolR1); 19E5 and 19A4 (3 rounds against murine FolR1); 15H7, 15B6, 16D5, 15E12, 21D1, 16F12, 21A5, 21G8 (human-cynomolgus-human FolR1); 19H3, 20G6, and 20H7 (3 rounds against murine FolR1).

Human, murine and cynomolgus FolR1 as antigens for the phage display selections as well as ELISA- and SPR-based screenings were transiently expressed as N-terminal monomeric Fc-fusion in HEK EBNA cells and in vivo site-specifically biotinylated via co-expression of BirA biotin ligase at the avi-tag recognition sequence located at the C-terminus of the Fc portion carrying the receptor chain (Fc knob chain). In order to assess the specificity to FolR1, two related receptors, human FolR2 and FolR3 were generated in the same way.

Selection rounds (biopanning) were performed in solution according to the following pattern:

1. Pre-clearing of ˜1012 phagemid particles on maxisorp plates coated with 10 ug/ml of an unrelated human IgG to deplete the libraries of antibodies recognizing the Fc-portion of the antigen.

2. Incubating the non-Fc-binding phagemid particles with 100 nM biotinylated human, cynomolgus, or murine FolR1 for 0.5 h in the presence of 100 nM unrelated non-biotinylated Fc knob-into-hole construct for further depletion of Fc-binders in a total volume of 1 ml.

3. Capturing the biotinylated FolR1 and attached specifically binding phage by transfer to 4 wells of a neutravidin pre-coated microtiter plate for 10 min (in rounds 1 & 3).

4. Washing the respective wells using 5× PBS/Tween20 and 5× PBS.

5. Eluting the phage particles by addition of 250 ul 100 mM TEA (triethylamine) per well for 10 min and neutralization by addition of 500 ul 1 M Tris/HCl pH 7.4 to the pooled eluates from 4 wells.

6. Post-clearing of neutralized eluates by incubation on neutravidin pre-coated microtiter plate with 100 nM biotin-captured FolR2 or FolR3 for final removal of Fc- and unspecific binders.

7. Re-infection of log-phase E. coli TG1 cells with the supernatant of eluted phage particles, infection with helperphage VCSM13, incubation on a shaker at 30° C. over night and subsequent PEG/NaCl precipitation of phagemid particles to be used in the next selection round.

Selections were carried out over 3 rounds using constant antigen concentrations of 100 nM. In round 2, in order to avoid enrichment of binders to neutravidin, capture of antigen: phage complexes was performed by addition of 5.4×107 streptavidin-coated magnetic beads. Specific binders were identified by ELISA as follows: 100 ul of 25 nM biotinylated human, cynomolgus, or murine FolR1 and 10 ug/ml of human IgG were coated on neutravidin plates and maxisorp plates, respectively. Fab-containing bacterial supernatants were added and binding Fabs were detected via their Flag-tags using an anti-Flag/HRP secondary antibody. Clones exhibiting signals on human FolR1 and being negative on human IgG were short-listed for further analyses and were also tested in a similar fashion against the remaining two species of FolR1. They were bacterially expressed in a 0.5 liter culture volume, affinity purified and further characterized by SPR-analysis using BioRad's ProteOn XPR36 biosensor.

Affinities (KD) of selected clones were measured by surface plasmon resonance (SPR) using a ProteOn XPR36 instrument (Biorad) at 25° C. with biotinylated human, cynomolgus, and murine FolR1 as well as human FolR2 and FolR3 (negative controls) immobilized on NLC chips by neutravidin capture. Immobilization of antigens (ligand): Recombinant antigens were diluted with PBST (10 mM phosphate, 150 mM sodium chloride pH 7.4, 0.005% Tween 20) to 10 μg/ml, then injected at 30 μl/minute in vertical orientation. Injection of analytes: For ‘one-shot kinetics’ measurements, injection direction was changed to horizontal orientation, two-fold dilution series of purified Fab (varying concentration ranges) were injected simultaneously along separate channels 1-5, with association times of 200 s, and dissociation times of 600 s. Buffer (PBST) was injected along the sixth channel to provide an “in-line” blank for referencing. Association rate constants (kon) and dissociation rate constants (koff) were calculated using a simple one-to-one Langmuir binding model in ProteOn Manager v3.1 software by simultaneously fitting the association and dissociation sensorgrams. The equilibrium dissociation constant (KD) was calculated as the ratio koff/kon. Table 4 lists the equilibrium dissociation constants (KD) of the selected clones specific for FolR1.

TABLE 4 Equilibrium dissociation constants (KD) for anti-FolR1 antibodies (Fab-format) selected by phage display from common light chain sub-libraries comprising VL7_46-13, a humanized light chain specific for CD3ε KD in nM. huFolR1 cyFolR1 muFolR1 huFolR2 huFolR3 Clone [nm] [nm] [nm] [nm] [nm] 16A3 21.7 18 very weak no binding no binding 15A1 30.9 17.3 very weak no binding no binding 18D3 93.6 40.2 very weak no binding no binding 19E5 522 276 19.4 no binding no binding 19A4 2050 4250 43.1 no binding no binding 15H7 13.4 72.5 no binding no binding no binding 15B6 19.1 13.9 no binding no binding no binding 16D5 39.5 114 no binding no binding no binding 15E12 55.7 137 no binding no binding no binding 21D1 62.6 32.1 no binding no binding no binding 16F12 68 90.9 no binding no binding no binding 21A5 68.8 131 no binding no binding no binding 21G8 130 261 no binding no binding no binding 19H3 no binding no binding 89.7 no binding no binding 20G6 no binding no binding 78.5 no binding no binding

Example 7 Selection of Antibody Fragments from Generic Multi-Framework Libraries to FolR1

The antibodies 11F8, 36F2, 9D11, 5D9, 6B6, and 14E4 were obtained by phage display selections based on generic multi-framework sub-libraries against different species (human, cynomolgus and murine) of FolR1. In these multi-framework sub-libraries, different VL-domains with randomized CDR3 (3 different lengths) are paired with different VH-domains with randomized CDR3 (6 different lengths). The selected clones are of the following VL/VH pairings: 11F8 (Vk_1_5/VH_1_69), 36F2 (Vk_3_20/VH_1_46), 9D11 (Vk2D_28/VH1_46), 5D9 (Vk3_20/VH1_46), 6B6 (Vk3_20/VH1_46), and 14E4 (Vk3_20/VH3_23). In order to obtain species cross-reactive (or murine FolR1-reactive) antibodies, the different species of FolR1 were alternated (or kept constant) in different ways over 3 or 4 rounds of biopanning: 11F8 (cynomolgus-murine-human FolR1); 36F2 (human-murine-cynomolgus-murine FolR1); 9D11 (cynomolgus-human-cynomolgus FolR1); 5D9 (human-cynomolgus-human FolR1); 6B6 (human-cynomolgus-human FolR1) and 14E4 (3 rounds against murine FolR1).

Human, murine and cynomolgus FolR1 as antigens for the phage display selections as well as ELISA- and SPR-based screenings were transiently expressed as N-terminal monomeric Fc-fusion in HEK EBNA cells and in vivo site-specifically biotinylated via co-expression of BirA biotin ligase at the avi-tag recognition sequence located at the C-terminus of the Fc portion carrying the receptor chain (Fc knob chain). In order to assess the specificity to FolR1, two related receptors, human FolR2 and FolR3 were generated in the same way.

Selection rounds (biopanning) were performed in solution according to the following pattern:

1. Pre-clearing of ˜1012 phagemid particles on maxisorp plates coated with 10 ug/ml of an unrelated human IgG to deplete the libraries of antibodies recognizing the Fc-portion of the antigen.

2. Incubating the non-Fc-binding phagemid particles with 100 nM biotinylated human, cynomolgus, or murine FolR1 for 0.5 h in the presence of 100 nM unrelated non-biotinylated Fc knob-into-hole construct for further depletion of Fc-binders in a total volume of 1 ml.

3. Capturing the biotinylated FolR1 and attached specifically binding phage by transfer to 4 wells of a neutravidin pre-coated microtiter plate for 10 min (in rounds 1 & 3).

4. Washing the respective wells using 5× PBS/Tween20 and 5× PBS.

5. Eluting the phage particles by addition of 250 ul 100 mM TEA (triethylamine) per well for 10 min and neutralization by addition of 500 ul 1 M Tris/HCl pH 7.4 to the pooled eluates from 4 wells.

6. Post-clearing of neutralized eluates by incubation on neutravidin pre-coated microtiter plate with 100 nM biotin-captured FolR2 or FolR3 for final removal of Fc- and unspecific binders.

7. Re-infection of log-phase E. coli TG1 cells with the supernatant of eluted phage particles, infection with helperphage VCSM13, incubation on a shaker at 30° C. over night and subsequent PEG/NaCl precipitation of phagemid particles to be used in the next selection round.

Selections were carried out over 3 rounds using constant antigen concentrations of 100 nM. In round 2 and 4, in order to avoid enrichment of binders to neutravidin, capture of antigen: phage complexes was performed by addition of 5.4×107 streptavidin-coated magnetic beads. Specific binders were identified by ELISA as follows: 100 ul of 25 nM biotinylated human, cynomolgus, or murine FolR1 and 10 ug/ml of human IgG were coated on neutravidin plates and maxisorp plates, respectively. Fab-containing bacterial supernatants were added and binding Fabs were detected via their Flag-tags using an anti-Flag/HRP secondary antibody. Clones exhibiting signals on human FolR1 and being negative on human IgG were short-listed for further analyses and were also tested in a similar fashion against the remaining two species of FolR1. They were bacterially expressed in a 0.5 liter culture volume, affinity purified and further characterized by SPR-analysis using BioRad's ProteOn XPR36 biosensor.

Affinities (KD) of selected clones were measured by surface plasmon resonance (SPR) using a ProteOn XPR36 instrument (Biorad) at 25° C. with biotinylated human, cynomolgus, and murine FolR1 as well as human FolR2 and FolR3 (negative controls) immobilized on NLC chips by neutravidin capture. Immobilization of antigens (ligand): Recombinant antigens were diluted with PBST (10 mM phosphate, 150 mM sodium chloride pH 7.4, 0.005% Tween 20) to 10 μg/ml, then injected at 30 μl/minute in vertical orientation. Injection of analytes: For ‘one-shot kinetics’ measurements, injection direction was changed to horizontal orientation, two-fold dilution series of purified Fab (varying concentration ranges) were injected simultaneously along separate channels 1-5, with association times of 150 or 200 s, and dissociation times of 200 or 600 s, respectively. Buffer (PBST) was injected along the sixth channel to provide an “in-line” blank for referencing. Association rate constants (kon) and dissociation rate constants (koff) were calculated using a simple one-to-one Langmuir binding model in ProteOn Manager v3.1 software by simultaneously fitting the association and dissociation sensorgrams. The equilibrium dissociation constant (KD) was calculated as the ratio koff/kon. Table 5 lists the equilibrium dissociation constants (KD) of the selected clones specific for FolR1.

TABLE 5 Equilibrium dissociation constants (KD) for anti-FolR1 antibodies (Fab-format) selected by phage display from generic multi-framework sub-libraries KD in nM. KD (nM) Clone huFolR1 cyFolR1 muFolR1 huFolR2 huFolR3 11F8 632 794 1200 no binding no binding 36F2 1810 1640 737 no binding no binding 9D11 8.64 5.29 no binding no binding no binding 5D9 8.6 5.9 no binding no binding no binding 6B6 14.5 9.4 no binding no binding no binding 14E4 no binding no binding 6.09 no binding no binding

Example 8 Production and Purification of Novel FolR1 Binders in IgG and T-Cell Bispecific Formats

To identify FolR1 binders which are able to induce T-cell dependent killing of selected target cells the antibodies isolated from a common light chain- or Fab-library were converted into the corresponding human IgG1 format. In brief, the variable heavy and variable light chains of unique FolR1 binders from phage display were amplified by standard PCR reactions using the Fab clones as the template. The PCR products were purified and inserted (either by restriction endonuclease and ligase based cloning, or by ‘recombineering’ using the InFusion kit from Invitrogen) into suitable expression vectors in which they are fused to the appropriate human constant heavy or human constant light chain. The expression cassettes in these vectors consist of a chimeric MPSV promoter and a synthetic polyadenylation site. In addition, the plasmids contain the oriP region from the Epstein Barr virus for the stable maintenance of the plasmids in HEK293 cells harboring the EBV nuclear antigen (EBNA). After PEI mediated transfection the antibodies were transiently produced in HEK293 EBNA cells and purified by standard ProteinA affinity chromatography followed by size exclusion chromatography as described:

Transient Transfection and Production

All (bispecific) antibodies (if not obtained from a commercial source) used herein were transiently produced in HEK293 EBNA cells using a PEI mediated transfection procedure for the required vectors as described below. HEK293 EBNA cells are cultivated in suspension serum free in CD CHO culture medium. For the production in 500 ml shake flask 400 million HEK293 EBNA cells are seeded 24 hours before transfection (for alternative scales all amounts were adjusted accordingly). For transfection cells are centrifuged for 5 min by 210×g, supernatant is replaced by pre-warmed 20 ml CD CHO medium. Expression vectors are mixed in 20 ml CD CHO medium to a final amount of 200 μg DNA. After addition of 540 μl PEI solution is vortexed for 15 s and subsequently incubated for 10 min at room temperature. Afterwards cells are mixed with the DNA/PEI solution, transferred to a 500 ml shake flask and incubated for 3 hours by 37° C. in an incubator with a 5% CO2 atmosphere. After incubation time 160 ml F17 medium is added and cell are cultivated for 24 hours. One day after transfection 1 mM valporic acid and 7% Feed 1 is added. After 7 days cultivation supernatant is collected for purification by centrifugation for 15 min at 210×g, the solution is sterile filtered (0.22 μm filter) and sodium azide in a final concentration of 0.01% w/v is added, and kept at 4° C. After production the supernatants were harvested and the antibody containing supernatants were filtered through 0.22 μm sterile filters and stored at 4° C. until purification.

Antibody Purification

All molecules were purified in two steps using standard procedures, such as protein A affinity purification (Äkta Explorer) and size exclusion chromatography. The supernatant obtained from transient production was adjusted to pH 8.0 (using 2 M TRIS pH 8.0) and applied to HiTrap PA FF (GE Healthcare, column volume (cv)=5 ml) equilibrated with 8 column volumes (cv) buffer A (20 mM sodium phosphate, 20 mM sodium citrate, pH 7.5). After washing with 10 cv of buffer A, the protein was eluted using a pH gradient to buffer B (20 mM sodium citrate pH 3, 100 mM NaCl, 100 mM glycine) over 12 cv. Fractions containing the protein of interest were pooled and the pH of the solution was gently adjusted to pH 6.0 (using 0.5 M Na2HPO4 pH 8.0). Samples were concentrated to 2 ml using ultra-concentrators (Vivaspin 15R 30.000 MWCO HY, Sartorius) and subsequently applied to a HiLoad™ 16/60 Superdex™ 200 preparative grade (GE Healthcare) equilibrated with 20 mM Histidine, pH 6.0, 140 mM NaCl, 0.01% Tween-20. The aggregate content of eluted fractions was analyzed by analytical size exclusion chromatography. Therefore, 30 μl of each fraction was applied to a TSKgel G3000 SW XL analytical size-exclusion column (Tosoh) equilibrated in 25 mM K2HPO4, 125 mM NaCl, 200 mM L-arginine monohydrochloride, 0.02% (w/v) NaN3, pH 6.7 running buffer at 25° C. Fractions containing less than 2% oligomers were pooled and concentrated to final concentration of 1-1.5 mg/ml using ultra concentrators (Vivaspin 15R 30.000 MWCO HY, Sartorius). The protein concentration was determined by measuring the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence. Purity and molecular weight of the constructs were analyzed by SDS capillary electrophoresis in the presence and absence of a reducing agent following the manufacturer instructions (instrument Caliper LabChipGX, Perkin Elmer). Purified proteins were frozen in liquid N2 and stored at −80° C.

Based on in vitro characterization results selected binders were converted into a T-cell bispecific format. In these molecules the FolR1:CD3 binding moieties are arranged in a 2:1 order with the FolR1 Fabs being located at the N-terminus. For clones isolated from the standard Fab library the CD3 binding part was generated as a CrossFab (CH1Cκ crossing) while for the clones from the common light chain library no crossing was necessary. These bispecific molecules were produced and purified analogously to the IgGs.

TABLE 6 Yield and monomer content of novel FolR1 binders in IgG and TCB format, respectively. IgG TCB Yield Monomer Monomer # Clone Library [mg/L] [%] Yield [mg/L} [%] 1 11F8 Fab 8.03 96.26 2 14E4 Fab 8.90 98.12 3 15B6 CLC 7.72 100.00 4 15E12 CLC 6.19 100.00 5 15H7 CLC 8.94 100.00 6 16A3 CLC 0.60 n.d. 7 16D5 CLC 36.50 96.96 4.36 97.19 8 16F12 CLC 5.73 97.17 9 18D3 CLC 0.90 n.d. 10 19A4 CLC 38.32 100.00 37.50 100.00 11 19E5 CLC 46.09 100.00 12 19H3 CLC 7.64 100.00 13 20G6 CLC 24.00 100.00 14 20H7 CLC 45.39 100.00 15 21A5 CLC 1.38 98.56 47.31 95.08 16 21D1 CLC 5.47 100.00 17 21G8 CLC 6.14 97.28 9.27 100.00 18 36F2 Fab 11.22 100.00 18.00 100.00 19 5D9 Fab 20.50 100.00 0.93 97.32 20 6B6 Fab 3.83 100.00 4.17 91.53 21 9D11 Fab 14.61 100.00 2.63 100.00 CLC: Common light chain

Example 9 2+1 and 1+1 T-Cell Bispecific Formats

Four different T-cell bispecific formats were prepared for one common light chain binder (16D5) and three formats for one binder from the Fab library (9D11) to compare their killing properties in vitro.

The standard format is the 2+1 inverted format as already described (FolR1:CD3 binding moieties arranged in a 2:1 order with the FolR1 Fabs located at the N-terminus). In the 2+1 classical format the FolR1:CD3 binding moieties are arranged in a 2:1 order with the CD3 Fab being located at the N-terminus. Two monovalent formats were also prepared. The 1+1 head-to-tail has the FolR1:CD3 binding moieties arranged in a 1:1 order on the same arm of the molecule with the FolR1 Fab located at the N-terminus. In the 1+1 classical format the FolR1:CD3 binding moieties are present once, each on one arm of the molecule. For the 9D11 clone isolated from the standard Fab library the CD3 binding part was generated as a CrossFab (CH1Cκ crossing) while for the 16D5 from the common light chain library no crossing was necessary. These bispecific molecules were produced and purified analogously to the standard inverted T-cell bispecific format.

TABLE 7 Summary of the yield and final monomer content of the different T-cell bispecific formats. Monomer [%] Construct (SEC) Yield 16D5 FolR1 TCB 2 + 1 (inverted)  96% 5.4 mg/L 16D5 FolR1 TCB 2 + 1 (classical)  90% 4.6 mg/L 16D5 FolR1 TCB 1 + 1 100% 5.4 mg/L (head-to-tail) 16D5 FolR1 TCB 1 + 1 (classical) 100% 0.7 mg/L 9D11 FolR1 TCB 2 + 1 (inverted) 100% 2.6 mg/L 9D11 FolR1 TCB 1 + 1 100% 6.1 mg/L (head-to-tail) 9D11 FolR1 TCB 1 + 1 (classical)  96% 1.3 mg/L Mov19 FolR1 TCB 2 + 1 (inverted)  98% 3 mg/L Mov19 FolR1 TCB 1 + 1 100% 5.2 mg/L (head-to-tail)

Example 10 Biochemical Characterization of FolR1 binders by Surface Plasmon Resonance

Binding of FolR1 binders as IgG or in the T-cell bispecific format to different recombinant folate receptors (human FolR1, 2 and 3, murine FolR1 and cynomolgus FolR1; all as Fc fusions) was assessed by surface plasmon resonance (SPR). All SPR experiments were performed on a Biacore T200 at 25° C. with HBS-EP as running buffer (0.01 M HEPES pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Surfactant P20, Biacore, Freiburg/Germany).

Single Injections

First the anti-FolR1 IgGs were analyzed by single injections (Table 1) to characterize their crossreactivity (to human, murine and cyno FolR1) and specificity (to human FolR1, human FolR2, human FolR3). Recombinant biotinylated monomeric Fc fusions of human, cynomolgus and murine Folate Receptor 1 (FolR1-Fc) or human Folate Receptor 2 and 3 (FolR2-Fc, FolR3-Fc) were directly coupled on a SA chip using the standard coupling instruction (Biacore, Freiburg/Germany). The immobilization level was about 300-400 RU. The IgGs were injected for 60 seconds at a concentration of 500 nM. IgGs binding to huFolR2 and huFolR3 were rejected for lack of specificity. Most of the binders are only crossreactive between human and cyno FolR1, additional crossreactivity to murine FolR1 went most of the time hand in hand with loss of specificity.

TABLE 8 Crossreactivity and specificity of 25 new folate receptor 1 binders (as IgGs) as well as of two control IgGs (Mov19 and Farletuzumab). Clone Binding to Binding to Binding to Binding to Binding to name huFolR1 cyFolR1 muFolR1 huFolR2 huFolR3 Mov19 + + Farletuzumab + + 16A3 + + +/− 18D3 + + 19E5 + + + + + 19A4 + + + 15H7 + + + 15B6 + + 16D5 + + 15E12 + + +/− + + 21D1 + + +/− 16F12 + + 21A5 + + +/− 21G8 + + + + 19H3 + 20G6 + 20H7 + 9D11 + + 5D9 + + + + 6B6 + + + + 11F8 + + + + + 36F2 + + + 14E4 + + means binding, − means no binding, +/- means weak binding.

Avidity to Folate Receptor 1

The avidity of the interaction between the anti-FolR1 IgGs or T cell bispecifics and the recombinant folate receptors was determined as described below (Table 9).

Recombinant biotinylated monomeric Fc fusions of human, cynomolgus and murine Folate Receptor 1 (FolR1-Fc) were directly coupled on a SA chip using the standard coupling instruction (Biacore, Freiburg/Germany). The immobilization level was about 300-400 RU. The anti-FolR1 IgGs or T cell bispecifics were passed at a concentration range from 2.1 to 500 nM with a flow of 30 μL/minutes through the flow cells over 180 seconds. The dissociation was monitored for 600 seconds. Bulk refractive index differences were corrected for by subtracting the response obtained on reference flow cell immobilized with recombinant biotinylated IL2 receptor Fc fusion. For the analysis of the interaction of 19H3 IgG and murine folate receptor 1, folate (Sigma F7876) was added in the HBS-EP running buffer at a concentration of 2.3 μM. The binding curves resulting from the bivalent binding of the IgGs or T cell bispecifics were approximated to a 1:1 Langmuir binding and fitted with that model (which is not correct, but gives an idea of the avidity). The apparent avidity constants for the interactions were derived from the rate constants of the fitting using the Bia Evaluation software (GE Healthcare).

TABLE 9 Bivalent binding (avidity with apparent KD) of selected FolR1 binders as IgGs or as T-cell bispecifics (TCB) on human and cyno FolR1. Apparent Analyte Ligand ka (1/Ms) kd (1/s) KD (M) 16D5 TCB huFolR1 8.31E+04 3.53E−04 4.24E−09 cyFolR1 1.07E+05 3.70E−04 3.45E−09 9D11 TCB huFolR1 1.83E+05 9.83E−05 5.36E−10 cyFolR1 2.90E+05 6.80E−05 2.35E−10 21A5 TCB huFolR1 2.43E+05 2.64E−04 1.09E−09 cyFolR1 2.96E+05 2.76E−04 9.32E−10 36F2 IgG huFolR1 2.62E+06 1.51E−02 5.74E−9 cyFolR1 3.02E+06 1.60E−02 5.31E−9 muFolR1  3.7E+05 6.03E−04 1.63E−9 Mov19 IgG huFolR1 8.61E+05 1.21E−04  1.4E−10 cyFolR1 1.29E+06 1.39E−04 1.08E−10 Farletuzumab huFolR1 1.23E+06    9E−04  7.3E−10 cyFolR1 1.33E+06 8.68E−04  6.5E−10 19H3 IgG muFolR1  7.1E+05  1.1E−03 1.55E−09

1. Affinity to Folate Receptor 1

The affinity of the interaction between the anti-FolR1 IgGs or the T cell bispecifics and the recombinant folate receptors was determined as described below (Table 10).

For affinity measurement, direct coupling of around 6000-7000 resonance units (RU) of the anti-human Fab specific antibody (Fab capture kit, GE Healthcare) was performed on a CM5 chip at pH 5.0 using the standard amine coupling kit (GE Healthcare). Anti-FolR1 IgGs or T cell bispecifics were captured at 20 nM with a flow rate of 10 μl/min for 20 or 40 sec, the reference flow cell was left without capture. Dilution series (6.17 to 500 nM or 12.35 to 3000 nM) of human or cyno Folate Receptor 1 Fc fusion were passed on all flow cells at 30 μl/min for 120 or 240 sec to record the association phase. The dissociation phase was monitored for 240 s and triggered by switching from the sample solution to HBS-EP. The chip surface was regenerated after every cycle using a double injection of 60 sec 10 mM Glycine-HCl pH 2.1 or pH 1.5. Bulk refractive index differences were corrected for by subtracting the response obtained on the reference flow cell 1. The affinity constants for the interactions were derived from the rate constants by fitting to a 1:1 Langmuir binding using the Bia Evaluation software (GE Healthcare).

TABLE 10 Monovalent binding (affinity) of selected FolR1 binders as IgGs or as T-cell bispecifics (TCB) on human and cyno FolR1. ka kd KD Ligand Analyte (1/Ms) (1/s) (M) 16D5 TCB huFolR1 1.53E+04 6.88E−04 4.49E−08 cyFolR1 1.32E+04 1.59E−03 1.21E−07 9D11 TCB huFolR1 3.69E+04 3.00E−04 8.13E−09 cyFolR1 3.54E+04 2.06E−04 5.82E−09 21A5 TCB huFolR1 1.79E+04  1.1E−03 6.16E−08 cyFolR1 1.48E+04 2.06E−03  1.4E−07 Mov19 IgG huFolR1 2.89E+05 1.59E−04  5.5E−10 cyFolR1 2.97E+05 1.93E−04  6.5E−10 Farletuzumab huFolR1 4.17E+05 2.30E−02 5.53E−08 cyFolR1 5.53E+05 3.73E−02 6.73E−08

2. Affinity to CD3

The affinity of the interaction between the anti-FolR1 T cell bispecifics and the recombinant human CD3εδ-Fc was determined as described below (Table 11).

For affinity measurement, direct coupling of around 9000 resonance units (RU) of the anti-human Fab specific antibody (Fab capture kit, GE Healthcare) was performed on a CM5 chip at pH 5.0 using the standard amine coupling kit (GE Healthcare). Anti-FolR1 T cell bispecifics were captured at 20 nM with a flow rate of 10 μl/min for 40 sec, the reference flow cell was left without capture. Dilution series (6.17 to 500 nM) of human CD3εδ-Fc fusion were passed on all flow cells at 30 μl/min for 240 sec to record the association phase. The dissociation phase was monitored for 240 s and triggered by switching from the sample solution to HBS-EP. The chip surface was regenerated after every cycle using a double injection of 60 sec 10 mM Glycine-HCl pH 2.1. Bulk refractive index differences were corrected for by subtracting the response obtained on the reference flow cell 1. The affinity constants for the interactions were derived from the rate constants by fitting to a 1:1 Langmuir binding using the Bia Evaluation software (GE Healthcare).

TABLE 11 Monovalent binding (affinity) of selected FolR1 T-cell bispecifics (TCB) on human CD3-Fc. Ligand Analyte ka (1/Ms) kd (1/s) KD (M) 16D5 TCB huCD3 4.25E+04 3.46E−03 8.14E−08 21A5 TCB huCD3 3.72E+04 3.29E−03  8.8E−08 The CD3 binding part is identical for all constructs and the affinity is similar for the measured T cell bispecifics (KD range between 60 and 90 nM).

Example 11 Simultaneous Binding T Cell Bispecifics on Folate Receptor 1 and CD3

Simultaneous binding of the anti-FolR1 T cell bispecifics on recombinant Folate Receptor 1 and recombinant human CD3εδ-Fc was determined by surface plasmon resonance as described below. Recombinant biotinylated monomeric Fc fusions of human, cynomolgus and murine Folate Receptor 1 (FolR1-Fc) were directly coupled on a SA chip using the standard coupling instruction (Biacore, Freiburg/Germany). The immobilization level was about 300-400 RU. The anti-FolR1 T cell bispecifics were injected for 60 s at 500 nM with a flow of 30 μL/minutes through the flow cells, followed by an injection of hu CDεδ-Fc for 60 s at 500 nM. Bulk refractive index differences were corrected for by subtracting the response obtained on reference flow cell immobilized with recombinant biotinylated IL2 receptor Fc fusion. The four T cell bispecifics tested (16D5 TCB, 21A5 TCB, 51C7 TCB and 45D2 TCB) were able to bind simultaneously to Folate Receptor 1 and human CD3 as expected.

Example 12 Epitope Binning

For epitope binning, the anti-FolR1 IgGs or T cell bispecifics were directly immobilized on a CM5 chip at pH 5.0 using the standard amine coupling kit (GE Healthcare), with a final response around 700 RU. 500 nM huFolR1-Fc was then captured for 60 s, followed by 500 nM of the different binders for 30 s. The surface was regenerated with two injections of 10 mM glycine pH 2 for 30 s each. It is assessed if the different binders can bind to huFolR1 captured on immobilized binders (Table 12).

TABLE 12 Epitope characterization of selected FolR1 binders as IgGs or as T-cell bispecifics (TCB) on human FolR1. Analytes in solution On 16D5 21A5 9D11 36F2 Mov19 huFolR1 TCB TCB TCB IgG IgG Farletuzumab Im- 16D5 + + + mobi- TCB lized 21A5 + + + TCB 9D11 No additional binding on FolR1 possible once TCB captured on 9D11 36F2 IgG Measure not possible, huFolR1 dissociates too rapidly Mov19 + + +/− IgG + means binding, − means no binding, +/− means weak binding

Based on these results and additional data with simultaneous binding on immobilized huFolR1, the binders were separated in three groups. It is not clear if 9D11 has a separate epitope because it displaces all the other binders. 16D5 and 21A5 seem to be in the same group and Mov19, Farletuzumab (Coney et al., Cancer Res. 1991 Nov. 15; 51(22):6125-32; Kalli et al., Curr Opin Investig Drugs. 2007 December; 8(12):1067-73) and 36F2 in another (Table 13). However, 36F2 binds to a different epitope than Mov 19 and Farletuzumab as it binds to human, cynomous and murine FolR1.

TABLE 13 Epitope grouping of selected FolR1 binders as IgGs or as T-cell bispecifics (TCB) on human FolR1 Epitope 1 Epitope 2 Epitope 3 16D5 9D11 Mov19 21A5 Farletuzumab 36F2

Example 13 Selection of Binders

FolR1 binders in the IgG formats were screened by surface plasmon resonance (SPR) and by in vitro assay on cells to select the best candidates.

The anti-FolR1 IgGs were analyzed by SPR to characterize their crossreactivity (to human, murine and cynomolgus FolR1) and specificity (to human FolR1, human FolR2, human FolR3). Unspecific binding to human FolR2 and 3 was considered an exclusion factor. Binding and specificity to human FolR1 was confirmed on cells. Some binders did not bind on cells expressing FolR1 even though they recognized the recombinant human FolR1 in SPR. Aggregation temperature was determined but was not an exclusion factor because the selected binders were all stable. Selected binders were tested in a polyreactivity ELISA to check for unspecific binding, which led to the exclusion of four binders. This process resulted in an initial selection of three binders: 36F2 (Fab library), 9D11 (Fab library) and 16D5 (common light chain). 36F2 dissociated rapidly from huFolR1 and was, therefore, initially not favored.

Example 14 Specific Binding of Newly Generated FolR1 Binders to Human FolR1 Positive Tumor Cells

New FolR1 binders were generated via Phage Display using either a Fab library or a common light chain library using the CD3 light chain. The identified binders were converted into a human IgG1 format and binding to FolR1 high expressing HeLa cells was addressed. As reference molecule the human FolR1 binder Mov19 was included. Most of the binders tested in this assay showed intermediate to good binding to FolR1 with some clones binding equally well as Mov19 (see FIG. 2). The clones 16A3, 18D3, 15H7, 15B6, 21D1, 14E4 and 16F12 were excluded because binding to FolR1 on cells could not be confirmed by flow cytometry. In a next step the selected clones were tested for specificity to human FolR1 by excluding binding to the closely related human FolR2. HEK cells were transiently transfected with either human FolR1 or human FolR2 to address specificity. The clones 36F2 and 9D11 derived from the Fab library and the clones 16D5 and 21A5 derived from the CLC library bind specifically to human FolR1 and not to human FolR2 (see FIGS. 3A-B). All the other tested clones showed at least some binding to human FolR2 (see FIGS. 3A-B). Therefore these clones were excluded from further characterization. In parallel cross-reactivity of the FolR1 clones to cyno FolR1 was addressed by performing binding studies to HEK cells transiently transfected with cyno FolR1. All tested clones were able to bind cyno FolR1 and the four selected human FoLR1 specific clones 36F2, 9D11, 16D5 and 21A5 bind comparably well human and cyno FoLR1 (FIG. 4). Subsequently three human FolR1 specific cyno cross-reactive binders were converted into TCB format and tested for induction of T cell killing and T cell activation. These clones were 9D11 from the Fab library and 16D5 and 21A5 from the CLC library. As reference molecule Mov19 FolR1 TCB was included in all studies. These FolR1 TCBs were then used to compare induction of internalization after binding to FolR1 on HeLa cells. All three tested clones are internalized upon binding to FolR1 comparable to internalization upon binding of Mov19 FoLR1 TCB (FIG. 5). 21A5 FolR1 TCB was discontinued due to signs of polyreactivity.

Example 15 T Cell-Mediated Killing of FolR1-Expressing Tumor Target Cells Induced by FolR1 TCB Antibodies

The FolR1 TCBs were used to determine T cell mediated killing of tumor cells expressing FoLR1. A panel of potential target cell lines was used to determine FoLR1 binding sites by Qifikit analysis.

The used panel of tumor cells contains FolR1 high, intermediate and low expressing tumor cells and a FolR1 negative cell line.

TABLE 14 FolR1 binding sites on tumor cells FolR1 Cell line Origin binding sites Hela Cervix adenocarcinoma 2′240′716 Skov3 Ovarian adenocarcinoma 91′510 OVCAR5 Ovarian adenocarcinoma 22′077 HT29 Colorectal adenocarcinoma 10′135 MKN45 Gastric adenocarcinoma 54

Binding of the three different FoLR1 TCBs (containing 9D11, 16D5 and Mov19 binders) to this panel of tumor cell lines was determined showing that the FolR1 TCBs bind specifically to FolR1 expressing tumor cells and not to a FoLR1 negative tumor cell line. The amount of bound construct is proportional to the FolR1 expression level and there is still good binding of the constructs to the FolR1 low cell line HT-29 detectable. In addition there is no binding of the negative control DP47 TCB to any of the used cell lines (FIGS. 6A-E).

The intermediate expressing cell line SKOV3 and the low expressing cell line HT-29 were further on used to test T cell mediated killing and T cell activation using 16D5 TCB and 9D11 TCB; DP47 TCB was included as negative control. Both cell lines were killed in the presence of already very low levels of 16D5 TCB and 9D11 TCB and there was no difference in activity between both TCBs even though 9D11 TCB binds stronger to FolR1 than 16D5 TCB. Overall killing of SKOV3 cells was higher compared to HT-29 which reflects the higher expression levels of FolR1 on SKOV3 cells (FIGS. 7A-D). In line with this, a strong upregulation of the activation marker CD25 and CD69 on CD4+ T cells and CD8+ T cells was detected. Activation of T cells was very similar in the presence of SKOV3 cells and HT-29 cells. The negative control DP47 TCB does not induce any killing at the used concentrations and there was no significant upregulation of CD25 and CD69 on T cells.

TABLE 15 EC50 values of tumor cell killing and T cell activation with SKOV3 cells CD4+ CD4+ CD8+ CD8+ Killing Killing CD69+ CD25+ CD69+ CD25+ Construct 24 h (pM) 48 h (pM) (%) (%) (%) (%) 9D11 1.1 0.03 0.51 0.46 0.019 0.03 FolR1 TCB 16D5 0.7 0.04 0.34 0.33 0.025 0.031 FolR1 TCB

TABLE 16 EC50 values of tumor cell killing and T cell activation with HT-29 cells CD4+ CD4+ CD8+ CD8+ Killing Killing CD69+ CD25+ CD69+ CD25+ Construct 24 h (pM) 48 h (pM) (%) (%) (%) (%) 9D11 2.3 0.1 1.22 1.11 0.071 0.084 FolR1 TCB 16D5 2.8 0.1 0.69 0.62 0.021 0.028 FolR1 TCB

Example 16 Binding to Erythrocytes and T Cell Activation in whole Blood

To prove that there is no spontaneous activation in the absence of FoLR1 expressing tumor cells we tested if there is binding of the FolR1 clones to erythrocytes which might potentially express FolR1. We could not observe any specific binding of 9D11 IgG, 16D5 IgG and Mov19 IgG to erythrocytes, as negative control DP47 IgG was included (FIG. 8).

To exclude any further unspecific binding to blood cells or unspecific activation via FoLR1 TCB, 9D11 TCB, 16D5 TCB and Mov19 TCB were added into whole blood and upregulation of CD25 and CD69 on CD4+ T cells and CD8+ T cells was analyzed by flow cytometry. DP47 TCB was included as negative control. No activation of T cells with any of the tested constructs could be observed by analyzing upregulation of CD25 and CD69 on CD4+ T cells and CD8+ T cells (FIG. 9).

Example 17 Removal of the N-Glycosylation Site in 9D11 Light Chain

During analysis of the different FolR1 binders to identify potential sequence hot spots, at the end of CDR L3 of the clone 9D11 a putative N-glycosylation site was identified. Usually the consensus motif for N-glycosylation is defined as N-X-S/T-X (where X is not P). The sequence of CDR L3 (MQASIMNRT (SEQ ID NO: 46)) perfectly matches this consensus motif having the sequence N-R-T. Since glycosylation might not be completely reproducible among different production batches this could have an impact on FolR1 binding, if the glycosylation in CDR L3 contributes to antigen binding. To evaluate if this N-glycosylation site is important for FolR1 binding, or could be replaced without impairing binding, different variants of the 9D11 light chain were generated in which the N-glycosylation site was exchanged by site specific mutagenesis.

1. Transient Rransfection and Production

The four T cell bispecifics were transiently produced in HEK293 EBNA cells using a PEI mediated transfection procedure for the required vectors as described below. HEK293 EBNA cells were cultivated in suspension serum free in CD CHO culture medium. For the production in 500 ml shake flask 400 million HEK293 EBNA cells were seeded 24 hours before transfection (for alternative scales all amounts were adjusted accordingly). For transfection cells were centrifuged for 5 min by 210×g, supernatant was replaced by pre-warmed 20 ml CD CHO medium. Expression vectors were mixed in 20 ml CD CHO medium to a final amount of 200 μg DNA. After addition of 540 μl PEI solution was vortexed for 15 s and subsequently incubated for 10 min at room temperature. Afterwards cells were mixed with the DNA/PEI solution, transferred to a 500 ml shake flask and incubated for 3 hours by 37° C. in an incubator with a 5% CO2 atmosphere. After incubation time 160 ml F17 medium was added and cell were cultivated for 24 hours. One day after transfection 1 mM valporic acid and 7% Feed 1 was added. After 7 days cultivation supernatant was collected for purification by centrifugation for 15 min at 210×g, the solution is sterile filtered (0.22 μm filter) and sodium azide in a final concentration of 0.01% w/v was added, and kept at 4° C. After production the supernatants were harvested and the antibody containing supernatants were filtered through 0.22 μm sterile filters and stored at 4° C. until purification.

2. Antibody Purification

All molecules were purified in two steps using standard procedures, such as protein A affinity purification (Äkta Explorer) and size exclusion chromatography. The supernatant obtained from transient production was adjusted to pH 8.0 (using 2 M TRIS pH 8.0) and applied to HiTrap PA HP (GE Healthcare, column volume (cv)=5 ml) equilibrated with 8 column volumes (cv) buffer A (20 mM sodium phosphate, 20 mM sodium citrate, 0.5 M NaCl, 0.01% Tween-20, pH 7.5). After washing with 10 cv of buffer A, the protein was eluted using a pH gradient to buffer B (20 mM sodium citrate pH 2.5, 0.5 M NaCl, 0.01% Tween-20) over 20 cv. Fractions containing the protein of interest were pooled and the pH of the solution was gently adjusted to pH 6.0 (using 2 M Tris pH 8.0). Samples were concentrated to 1 ml using ultra-concentrators (Vivaspin 15R 30.000 MWCO HY, Sartorius) and subsequently applied to a Superdex™ 200 10/300 GL (GE Healthcare) equilibrated with 20 mM Histidine, pH 6.0, 140 mM NaCl, 0.01% Tween-20. The aggregate content of eluted fractions was analyzed by analytical size exclusion chromatography. Therefore, 30 μl of each fraction was applied to a TSKgel G3000 SW XL analytical size-exclusion column (Tosoh) equilibrated in 25 mM K2HPO4, 125 mM NaCl, 200 mM L-arginine monohydrochloride, 0.02% (w/v) NaN3, pH 6.7 running buffer at 25° C. Fractions containing less than 2% oligomers were pooled and concentrated to final concentration of 1-1.5 mg/ml using ultra concentrators (Vivaspin 15R 30.000 MWCO HY, Sartorius). The protein concentration was determined by measuring the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence. Purity and molecular weight of the constructs were analyzed by SDS capillary electrophoresis in the presence and absence of a reducing agent following the manufacturer instructions (instrument Caliper LabChipGX, Perkin Elmer). Purified proteins were frozen in liquid N2 and stored at -80° C.

3. Aggregation Temperature

Stability of the four constructs was tested on an Optim1000 (Avacta, PALL Corporation) by a gradient heating from 25° to 80° at 0.1° C./min. The temperature at onset of aggregation is recorded.

TABLE 34 Yield, monomer content and aggregation temperature of four N-glycosylation site knock-out mutant of the 9D11 binder in the 2 + 1 inverted T-cell bispecific format. All four mutants behaved similarly to the wild-type 9D11 binder Yield Mononomer Aggregation Clone Mutation [mg/L} [%] temperature 9D11 T102N 1.34 97 56° 9D11 T102A 1.29 100 56° 9D11 N100Q 2.5 100 56° 9D11 N100S 2.05 100 56° 9D11 2.6 100 57°

The following variants were generated: N100S (N95S); N100Q (N95Q), T102A (T97A) and T102N (T97N) (Kabat numbering indicated in parenthesis) and converted into the T-cell bispecific format. After transient production in HEK293 EBNA cells and purification the different variants were analyzed for target binding and cell killing activity in comparison to the original 9D11 clone.

TABLE 17 primers used for removal of N-glycosylation site in CDR L3 of 9D11 (sequences see below) Amino acid Mutagenesis # exchange primer 1 N95S GAB-7735 2 N95Q GAB-7734 3 T97A GAB7736 4 T97N GAB-7737

Example 18 Binding and T Cell Mediated Killing with 9D11 a-Glyco Variants

Due to a glycosylation site in the CDRs four different 9D11 variants were produced with a mutation removing the glycosylation site (Example 17). These four variants were tested in comparison to the original 9D11 for binding to FolR1 on HeLa cells (FIG. 10) and induction of tumor cell killing on SKOV3 and HT-29 (FIG. 11A-B, E-F). None of the variants showed differences in binding or induction of tumor cell killing. In parallel unspecific killing of the FolR1 negative cell lines MKN-45 was addressed (FIGS. 11C-D). Also, no differences between the variants and the original binder could be observed. None of the constructs induced unspecific killing on FoLR1 negative tumor cells.

Example 19 FolR1 Expression on Primary Epithelial Cells

FolR1 is expressed at low levels on primary epithelial cells. Here we wanted to test if these levels are sufficient to induce T cell mediated killing in the presence of the FolR1 TCBs. To test this we used primary human bronchial epithelial cells, primary human choroid plexus epithelial cell, primary human renal cortical epithelial cells and primary human retinal pigment epithelial cells. As positive control either FolR1 positive SKOV3 cells or HT-29 cells were included. First we verified FolR1 expression on the used primary cells and determined the amount of FolR1 binding sites on these cells. Bronchial epithelial cells, renal cortical epithelial cells and retinal pigment epithelial cells express very low but significant levels of FolR1 compared to the levels expressed on tumor cells. The choroid plexus epithelial cells do not express significant levels of FolR1.

TABLE 18 FolR1 binding sites on primary epithelial cells Cell line Binding sites Bronchial epithelium 492 Choroid plexus epithelium 104 Renal cortical epithelium 312 Retinal pigment epithelium 822 Skov3 69′890

The primary epithelial cells that demonstrated FolR1 expression on the surface were used to address the question if these cells can be killed by T cells in the presence of FoLR1 TCBs. No significant levels of killing could be measured but induction of T cell activation in the presence of retinal pigment epithelial cells, bronchial epithelial cells and renal cortical cells resulting in upregulation of CD25 and CD69 was detected. The strongest activation is seen with retinal pigment epithelial cells resulting in upregulation of CD25 and CD69 both on CD4+ T cells and CD8+ T cells. In the presence of bronchial epithelial cells lower activation of T cells is induced with upregulation of CD69 on CD4+ T cells and CD8+ T cells but very low upregulation of CD25 only on CD4+ T cells but not on CD8+ T cells. The lowest activation of T cells is obtained in the presence of renal epithelial cells with no upregulation of CD25 on CD4 T+ cells and CD8+ T cells and CD69 been only upregulated on CD8+ T cells (FIGS. 12A-X).

Example 20 Comparison of Different TCB Formats Containing either 16D5 or 9D11 Binder

To determine if the TCB 2+1 inverted format is the most active format with the selected FolR1 binder, different formats containing either 16D5 or 9D11 were produced and compared in target cell binding, T cell mediated killing and T cell activation. The 16D5 binder was tested in the TCB 2+1 inverted (FIG. 1A), TCB 2+1 classical (FIG. 1D), TCB 1+1 classical (FIG. 1C) and TCB 1+1 head-to-tail (FIG. 1B) format; the 9D11 binder was tested in the TCB 2+1 inverted (FIG. 1A), TCB 1+1 classical (FIG. 1C) and TCB 1+1 head-to-tail (FIG. 1B) format.

All constructs were tested for binding to FolR1 on HeLa cells. The molecules bivalent for binding to FolR1 bind stronger compared to the monovalent constructs due to avidity. The difference between the bivalent vs. monovalent constructs is more pronounced for 16D5. The reason might be that due to the lower affinity of 16D5 the avidity effect for this binder is stronger. Between the two 1+1 TCBs there is no significant difference in binding but there is a difference between the two 2+1 constructs. The inverted 2+1 construct binds stronger to FolR1 than the classical 2+1 construct. This indicates that in the classical 2+1 construct the binding to FoLR1 is influenced by the presence of the CD3 Fab whereas in the inverted construct binding is less influenced.

By testing T cell mediated killing with these constructs we could show that stronger binding of the 2+1 inverted TCB in converted into stronger tumor cell killing and T cell activation compared to the 2+1 classical TCB. The 16D5 FolR1 TCB 2+1 classical is only a little bit more active than the respective 1+1 head-to-tail construct. The 1+1 head-to-tail construct is significantly more active than the 1+1 classical construct. This does not reflect the situation seen in binding and might be due to better crosslinking with the head-to-tail construct. Overall tumor cell killing and T cell activation is comparable with all tested constructs, the differences in potency seen with the differences are only in terms of EC50 values. In general it can be concluded that the FolR1 TCB 2+1 inverted independent of the used binder is the preferred format to induce T cell mediated tumor cell killing and T cell activation (see FIG. 13A-C and FIG. 14A-C).

TABLE 19 EC50 values of target cell binding and T cell mediated killing with different TCB formats Binding Killing Killing Construct EC50 (nM) 24 h (pM) 48 h (pM) 16D5 FolR1 TCB 11.03 1.43 0.18 2 + 1 inverted 16D5 FolR1 TCB 17.07 5.60 2.18 2 + 1 classical 16D5 FolR1 TCB 107.3 n.d. n.d. 1 + 1 classical 16D5 FolR1 TCB 102.6 26.24 6.06 1 + 1 head-to-tail 9D11 FolR1 TCB 17.52 0.74 0.14 2 + 1 inverted 9D11 FolR1 TCB 38.57 20.92 n.d. 1 + 1 classical 9D11 FolR1 TCB 44.20 4.73 n.d. 1 + 1 head-to-tail

TABLE 20 EC50 values of T cell activation in the presence of SKOV3 cells with different TCB formats CD4+ CD4+ CD8+ CD8+ CD25+ CD69+ CD25+ CD69+ Construct (%) (%) (%) (%) 16D5 FolR1 TCB 1.96 0.33 2.10 n.d. 2 + 1 inverted 16D5 FolR1 TCB 13.83 3.67 12.88 4.47 2 + 1 classical 16D5 FolR1 TCB 38.54 n.d. n.d. n.d. 1 + 1 classical 16D5 FolR1 TCB 17.14 7.47 25.15 n.d. 1 + 1 head-to-tail 9D11 FolR1 TCB 1.41 0.27 1.24 0.35 2 + 1 inverted 9D11 FolR1 TCB 34.01 n.d. 34.39 7.40 1 + 1 classical 9D11 FolR1 TCB 3.73 2.47 4.98 2.89 1 + 1 head-to-tail

Example 21 Tumor Cell Lines and Primary Cells

HeLa cells (CCL-2) were obtained from ATCC and cultured in DMEM with 10% FCS and 2 mM Glutamine, SKOV3 (HTB-77) were obtained from ATCC and cultured in RPMI with 10% FCS and 2 mM Glutamine, OVCAR5 were obtained from NCI and cultured in RPMI with 10% FCS and 2 mM Glutamine, HT-29 (ACC-299) were obtained from DSMZ and cultured in McCoy's 5A medium with 10% FCS and 2 mM Glutamine, MKN-45 (ACC-409) were obtained from DSMZ and cultured in RPMI with 10% FCS and 2 mM Glutamine.

All tested primary epithelial cells were obtained from ScienCell Research Laboratories. Human Bronchial Epithelium Cells (HBEpiC, Catalog Number 3210 were cultured in Bronchial Epithelial Cell Medium (BEpiCM, Cat. No. 3211, ScienCell). Human Colonic Epithelial Cells (HCoEpiC), Catalog Number 2950 were cultured in Colonic Epithelial Cell Medium (CoEpiCM, Cat. No. 2951, ScienCell). Human Retinal Pigment Epithelial Cells (HRPEpiC), Catalog Number 6540 were cultured in Epithelial Cell Medium (EpiCM, Cat. No. 4101, ScienCell). Human Renal Cortical Epithelial Cells (HRCEpiC), Catalog Number 4110, were cultured in Epithelial Cell Medium (EpiCM, Cat. No. 4101, ScienCell). Human Choroid Plexus Epithelial Cells (HCPEpiC), Catalog Number 1310 were cultured in Epithelial Cell Medium (EpiCM, Cat. No. 4101, ScienCell).

Example 22 Target Binding by Flow Cytometry

Target cells as indicated were harvested with Cell Dissociation Buffer, washed with PBS and resuspended in FACS buffer. The antibody staining was performed in a 96well round bottom plate. Therefore 200′000 cells were seeded per well. The plate was centrifuged for 4 min at 400 g and the supernatant was removed. The test antibodies were diluted in FACS buffer and 20 μl of the antibody solution were added to the cells for 30 min at 4° C. To remove unbound antibody the cells were washed twice with FACS buffer before addition of the diluted secondary antibody (FITC conjugated AffiniPure F(ab′)2 fragment goat anti-human IgG, Fcg Fragment, Jackson ImmunoResearch #109-096-098 or PE-conjugated AffiniPure F(ab′)2 Fragment goat anti-human IgG Fcg Fragment Specific, Jackson ImmunoResearch #109-116-170. After 30 min incubation on 4° C. unbound secondary antibody was washed away. Before measurement the cells were resuspended in 200 μl FACS buffer and analyzed by flow cytometry using BD Canto II or BD Fortessa.

Example 23 Internalization

The cells were harvested and the viability was determined. The cells were re-suspended in fresh cold medium at 2 Mio cells per ml and the cell suspension was transferred in a 15 ml falcon tube for each antibody. The antibodies that should be tested for internalization were added with a final concentration of 20 μg per ml to the cells. The tubes were incubated for 45 min in the cold room on a shaker. After incubation the cells were washed three times with cold PBS to remove unbound antibodies. 0.2 Mio cells per well were transfer to the FACS plate as time point zero. The labeled cells were re-suspended in warm medium and incubated at 37° C. At the indicated time-points 0.2 Mio cells per well were transferred in cold PBS, washed in plated on the FACS plate. To detect the constructs that remain on the surface the cells were stained with PE-labeled anti-human Fc secondary antibody. Therefore 20 μl of the diluted antibody were added per well and the plate was incubated for 30 min at 4° C. Then the cells were washed twice to remove unbound antibodies and then fixed with 1% PFA to prevent any further internalization. The fluorescence was measured using BD FACS Cantoll.

Example 24 QIFIKIT® Analysis

QIFIKIT® contains a series of beads, 10 μm in diameter and coated with different, but well-defined quantities of mouse Mab molecules (high-affinity anti-human CD5, Clone CRIS-1, isotype IgG2a). The beads mimic cells with different antigen densities which have been labeled with a primary mouse Mab, isotype IgG. Briefly, cells were labeled with primary mouse monoclonal antibody directed against the antigen of interest. In a separate test well, cells were labeled with irrelevant mouse monoclonal antibody (isotype control). Then, cells, Set-Up Beads and Calibration Beads were labeled with a fluorescein-conjugated anti-mouse secondary antibody included in the kit. The primary antibody used for labeling of the cells has to be used at saturating concentration. The primary antibody may be of any mouse IgG isotype. Under these conditions, the number of bound primary antibody molecules corresponds to the number of antigenic sites present on the cell surface. The secondary antibody is also used at saturating concentration. Consequently, the fluorescence is correlated with the number of bound primary antibody molecules on the cells and on the beads.

Example 25 T Cell Mediated Tumor Cell Killing and T Cell Activation

Target cells were harvested with Trypsin/EDTA, counted and viability was checked. The cells were resuspended in their respective medium with a final concentration of 300′000 cells per ml. Then 100 μl of the target cell suspension was transferred into each well of a 96-flat bottom plate. The plate was incubated overnight at 37° C. in the incubator to allow adherence of the cells to the plate. On the next day PBMCs were isolated from whole blood from healthy donors. The blood was diluted 2:1 with PBS and overlayed on 15 ml Histopaque-1077 (# 10771, Sigma-Aldrich) in Leucosep tubes and centrifuged for 30 min at 450 g without break. After centrifugation the band containing the cells was collected with a 10 ml pipette and transferred into 50 ml tubes. The tubes were filled up with PBS until 50 ml and centrifuged (400 g, 10 min, room temperature). The supernatant was removed and the pellet resuspended in PBS. After centrifugation (300 g, 10 min, room temperature), supernatants were discarded, 2 tubes were pooled and the washing step was repeated (this time centrifugation 350×g, 10 min, room temperature). Afterwards the cells were resuspended and the pellets pooled in 50 ml PBS for cell counting. After counting cells were centrifuged (350 g, 10 min, room temperature) and resuspended at 6 Mio cells per ml in RPMI with 2% FCS and 2 nM Glutamine. Medium was removed from plated target cells and the test antibodies diluted in RPMI with 2% FCS and 2 nM Glutamine were added as well as. 300′000 cells of the effector cell solution were transferred to each well resulting in a E:T ratio of 10:1. To determine the maximal release target cells were lysed with Triton X-100. LDH release was determined after 24 h and 48 h using Cytotoxicity Detection Kit (#1644793, Roche Applied Science). Activation marker upregulation on T cells after tumor cell killing was measured by flow cytometry. Briefly PBMCs were harvested, transferred into a 96 well round bottom plate and stained with CD4 PE-Cy7 (#3557852, BD Bioscience), CD8 FITC (#555634, BD Bioscience), CD25 APC (#555434, BD Bioscience), CD69 PE (#310906, BioLegend) antibodies diluted in FACS buffer. After 30 min incubation at 4° C. the cells were washed twice with FACS buffer. Before measuring the fluorescence using BD Canto II the cells were resuspended in 200 μl FACS buffer.

Example 26 T Cell Activation in Whole Blood

280 μl of fresh blood were added into a 96 well conical deep well plate. Then 20 μl of the diluted TCBs were added to the blood and mixed well by shaking the plate. After 24 h incubation at 37° C. in an incubator the blood was mixed and 35 μl were transferred to a 96 well round bottom plate. Then 20 μl of the antibody staining mix were added consisting of CD4 PE-Cy7 (#3557852, BD Bioscience), CD8 FITC (#555634, BD Bioscience), CD25 APC (#555434, BD Bioscience), CD69 PE (#310906, BioLegend) and CD45 V500 (#560777, BD Horizon) and incubated for 15 min in the dark at room temperature. Before measuring 200 μl of the freshly prepared BD FACS lysing solution (#349202, BD FCAS) was added to the blood. After 15 min incubation at room temperature the cells were measured with BD Fortessa.

Example 27 SDPK (Single Dose Pharmacokinetics) Study of Humanized FOLR1 TCB (Clone 16D5) in Immunodeficient NOD/Shi-scid/IL-2Rγnull (NOG) Mice

Female NOD/Shi-scid/IL-2Rγnull (NOG) mice, age 6-7 weeks at start of the experiment (bred at Taconic, Denmark) were maintained under specific-pathogen-free condition with daily cycles of 12 h light/12 h darkness according to committed guidelines (GV-Solas; Felasa; TierschG). The experimental study protocol was reviewed and approved by local government (P 2011/128). After arrival, animals were maintained for one week to get accustomed to the new environment and for observation. Continuous health monitoring was carried out on a regular basis.

Mice were injected i.v. with 10/1/0.1 μg/mouse of the FOLR1 TCB whereas 3 mice were bled per group and time point. All mice were injected with a total volume of 200 μl of the appropriate solution. To obtain the proper amount of the FOLR1 TCB per 200 μl, the stock solutions were diluted with PBS when necessary. Serum samples were collected 5 min, 1 h, 3 h, 8 h, 24 h, 48 h, 72 h, 96 h and 168 h after therapy injection.

FIG. 15 shows that the 16D5 FOLR1 TCB shows typical and dose proportional IgG-like PK properties in NOG mice with slow clearance.

TABLE 21 Formulation Concentration Compound Dose buffer (mg/mL) FOLR1 TCB 10 μg 20 mM Histidine, 5.43 (16D5) (corresponding 140 mM NaCl, (=stock to ca. 0.5 pH 6.0 solution) mg/kg) FOLR1 TCB 1 μg 20 mM Histidine, 5.43 (16D5) (corresponding 140 mM NaCl, (=stock to ca. 0.05 pH 6.0 solution) mg/kg) FOLR1 TCB 0.1 μg 20 mM Histidine, 5.43 (16D5) (corresponding 140 mM NaCl, (=stock to ca. 0.005 pH 6.0 solution) mg/kg)

Example 28 In Vivo Efficacy of FOLR1 TCB (Clone 16D5) after Human PBMC Rransfer in Skov3-Bearing NOG Mice

The FOLR1 TCB was tested in the human ovarian carcinoma cell line Skov3, injected s.c. into PBMC engrafted NOG mice.

The Skov3 ovarian carcinoma cells were obtained from ATCC (HTB-77). The tumor cell line was cultured in RPMI containing 10% FCS (Gibco) at 37° C. in a water-saturated atmosphere at 5% CO2. Passage 35 was used for transplantation, at a viability >95%. 5×106 cells per animal were injected s.c. into the right flank of the animals in a total of 100 μl of RPMI cell culture medium (Gibco).

Female NOD/Shi-scid/IL-2Rγnull (NOG) mice, age 6-7 weeks at start of the experiment (bred at Taconic, Denmark) were maintained under specific-pathogen-free condition with daily cycles of 12 h light/12 h darkness according to committed guidelines (GV-Solas; Felasa; TierschG). The experimental study protocol was reviewed and approved by local government (P 2011/128). After arrival, animals were maintained for one week to get accustomed to the new environment and for observation. Continuous health monitoring was carried out on a regular basis.

According to the protocol (FIG. 16), mice were injected s.c. on study day 0 with 5×106 of the Skov3. At study day 21, human PBMC of a healthy donor were isolated via the Ficoll method and 10×106 cells were injected i.p. into the tumor-bearing mice. Two days after, mice were randomized and equally distributed in five treatment groups (n=12) followed by i.v. injection with either 10/1/0.1 μg/mouse of the FOLR1 TCB or 10 μg/mouse of the DP47 control TCB once weekly for three weeks. All mice were injected i.v. with 200 μl of the appropriate solution. The mice in the vehicle group were injected with PBS. To obtain the proper amount of TCB per 200 μl, the stock solutions were diluted with PBS when necessary. Tumor growth was measured once weekly using a caliper (FIG. 17) and tumor volume was calculated as followed:


Tv:(W2/2)×L(W: Width, L: Length)

The once weekly injection of the FOLR1 TCB resulted in a dose-dependent anti-tumoral effect. Whereas a dose of 10 μg/mouse and 1 μg/mouse induced tumor shrinkage and 0.1 μg/mouse a tumor stasis (FIG. 17, Table 22). Maximal tumor shrinkage was achieved at a dose of 10 μg/mouse as compared to a non-targeted control DP47 TCB.

TABLE 22 Tumor growth Compound Dose inhibition DP47 TCB 10 μg  7% control (corresponding TCB to ca. 0.5 mg/kg) FOLR1 10 μg 90% TCB (corresponding (16D5) to ca. 0.5 mg/kg) FOLR1 1 μg 74% TCB (corresponding (16D5) to ca. 0.05 mg/kg) FOLR1 0.1 μg 56% TCB (corresponding (16D5) to ca. 0.005 mg/kg)

For PD read-outs, three mice per treatment group were sacrificed at study day 32, tumors were removed and single cell suspensions were prepared through an enzymatic digestion with Collagenase V, Dispase II and DNAse for subsequent FACS-analysis (FIGS. 19 and 20). Single cells where either used directly for staining of extracellular antigens and activation markers or were re-stimulated using 5 ng/ml PMA and 500 ng/ml Ionomycin in the presence of a protein transport inhibitor Monensin for 5 h in normal culture medium. After re-stimulation, cells were stained for surface antigens, followed by a fixation and permeabilization step. Fix samples were then stained intracellulary for TNF-α, IFN-γ, IL-10 and IL-2 and analyzed by flow cytometry.

Same procedure was used for the degranulation of cells, but an anti-CD107a antibody was added during the restimulation period and fixed samples were staining for intracellular perforin and granzyme-B contents. The FACS analysis revealed statistically higher number of infiltrating CD4+ and CD8+ T-cells in the tumor tissue upon treatment with FOLR1 TCB compared to vehicle and untargeted control TCB. Furthermore, higher numbers of TNF-□, IFN-□ and IL-2 producing as well as perforin+/granzym-B+ CD4+ and CD8+ T-cells were detected in FOLR1 TCB treated tumors. Tumor infiltrating T-cells treated with FOLR1 TCB also showed higher degranulation rates compared to control groups.

At study termination day 38, all animals were sacrificed; tumors were removed and weight (FIG. 18). The weight of the tumors treated with 10 and 1 μg/mouse of the FOLR1 TCB showed a statistically significant difference compared to the control groups.

TABLE 23 Formulation Concentration Compound Dose buffer (mg/mL) PBS FOLR1 TCB 10 μg 20 mM Histidine, 3.88 (16D5) 140 mM NaCl, (=stock pH 6.0 solution) FOLR1 TCB 1 μg 20 mM Histidine, 3.88 (16D5) 140 mM NaCl, (=stock pH 6.0 solution) FOLR1 TCB 0.1 μg 20 mM Histidine, 3.88 (16D5) 140 mM NaCl, (=stock pH 6.0 solution) DP47 TCB 10 μg 20 mM Histidine, 4.35 140 mM NaCl, (=stock pH 6.0 solution)

Example 29 Generation of a Bispecific FolR1/CD3-Kappa-Lambda Antibody

To generate a bispecific antibody (monovalent for each antigen) that simultaneously can bind to human CD3 and human folate receptor alpha (FolR1) without using any hetero-dimerization approach (e.g. knob-into-hole technology), a combination of a common light chain library with the so-called CrossMab technology was applied: The variable region of the humanized CD3 binder (CH2527_VL7_46/13) was fused to the CH1 domain of a standard human IgG1 antibody to form the VLVH crossed molecule (fused to Fc) which is common for both specificities. To generate the crossed counterparts (VHCL), a CD3 specific variable heavy chain domain (CH2527_VH_23/12) was fused to a constant human λ light chain whereas a variable heavy chain domain specific for human FolR1 (clone 16D5, isolated from common light chain library) was fused to a constant human κ light chain. This enables the purification of the desired bispecific antibody by applying subsequent purification steps with KappaSelect and LambdaFabSelect columns (GE Healthcare) to remove undesired homodimeric antibodies.

All antibody expression vectors were generated using standard recombinant DNA technology as described in Sambrook, J. et al., Molecular cloning: A laboratory manual; Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989. Molecular biological reagents were used according the manufacturer's recommendations. Genes or gene fragments were either amplified by polymerase chain reaction (PCR) or generated from synthetic oligonucleotides at Geneart AG (Regensburg, Germany) by automated gene synthesis. PCR-amplified or subcloned DNA fragments were confirmed by DNA sequencing (Synergene GmbH, Switzerland). Plasmid DNA was transformed into and amplified in suitable E. coli host strains for preparation of transfection-grade plasmid DNA using standard Maxiprep kits (Qiagen). For production of the bispecific molecules HEK293 EBNA cells were transfected with plasmids encoding the respective genes using a standard polyethlenimine (PEI) based method. The used plasmid ratio of the three expression vectors was 1:1:1. Transfected cells were cultivated for 7 days before supernatants were harvested for purification. The bispecific FolR1/CD3-kappa-lambda antibodies were produced and purified as follows.

1. Transient Rransfection and Production

The kappa-lambda bispecific antibody was transiently produced in HEK293 EBNA cells using a PEI mediated transfection procedure for the required vectors as described below. HEK293 EBNA cells were cultivated in suspension serum free in CD CHO culture medium. For the production in 500 ml shake flask 400 million HEK293 EBNA cells were seeded 24 hours before transfection (for alternative scales all amounts were adjusted accordingly). For transfection cells were centrifuged for 5 min by 210×g, supernatant is replaced by pre-warmed 20 ml CD CHO medium. Expression vectors were mixed in 20 ml CD CHO medium to a final amount of 200 μg DNA. After addition of 540 μl PEI solution is vortexed for 15 s and subsequently incubated for 10 min at room temperature. Afterwards cells were mixed with the DNA/PEI solution, transferred to a 500 ml shake flask and incubated for 3 hours by 37° C. in an incubator with a 5% CO2 atmosphere. After incubation time 160 ml F17 medium was added and cell were cultivated for 24 hours. One day after transfection 1 mM valporic acid and 7% Feed 1 was added. After 7 days cultivation supernatant was collected for purification by centrifugation for 15 min at 210×g, the solution is sterile filtered (0.22 μm filter) and sodium azide in a final concentration of 0.01% w/v was added, and kept at 4° C.

2. Purification

The kappa-lambda bispecific antibody was purified in three steps, using an affinity step specific for kappa light chains, followed by an affinity step specific for lambda light chains and finally by a size exclusion chromatography step for removal of aggregates. The supernatant obtained from transient production was adjusted to pH 8.0 (using 2 M TRIS pH 8.0) and applied to Capture Select kappa affinity matrix, or HiTrap KappaSelect, GE Healthcare, column volume (cv)=1 ml, equilibrated with 5 column volumes (cv) buffer A (50 mM Tris, 100 mM glycine, 150 mM NaCl, pH 8.0). After washing with 15 cv of buffer A, the protein was eluted using a pH gradient to buffer B (50 mM Tris, 100 mM glycine, 150 mM NaCl, pH 2.0) over 25 cv. Fractions containing the protein of interest were pooled and the pH of the solution was adjusted to pH 8.0 (using 2 M Tris pH 8.0). The neutralized pooled fractions were applied to Capture Select lambda affinity matrix (now: HiTrap LambdaFabSelect, GE Healthcare, column volume (cv)=1 ml) equilibrated with 5 column volumes (cv) buffer A (50 mM Tris, 100 mM glycine, 150 mM NaCl, pH 8.0). After washing with 15 cv of buffer A, the protein was eluted using a pH gradient to buffer B (50 mM Tris, 100 mM glycine, 150 mM NaCl, pH 2.0) over 25 cv. Fractions containing the protein of interest were pooled and the pH of the solution was adjusted to pH 8.0 (using 2 M Tris pH 8.0). This solution was concentrated using ultra-concentrators (Vivaspin 15R 30.000 MWCO HY, Sartorius) and subsequently applied to a Superdex™ 200 10/300 GL (GE Healthcare) equilibrated with 20 mM Histidine, pH 6.0, 140 mM NaCl, 0.01% Tween-20. The pooled fractions after size exclusion were again concentrated using ultra-concentrators (Vivaspin 15R 30.000 MWCO HY, Sartorius).

The protein concentration was determined by measuring the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence. Purity and molecular weight of the constructs were analyzed by SDS capillary electrophoresis in the presence and absence of a reducing agent following the manufacturer instructions (instrument Caliper LabChipGX, Perkin Elmer). Only small amounts of protein could be purified with a final yield of 0.17 mg/L.

Example 30 T Cell Mediated Killing with Bispecific FolR1/CD3-Kappa-Lambda Antibody

Activity of kappa lambda FolR1 TCB was tested on SKOV3 cells in the presence of freshly isolated PBMCs. As negative control DP47 TCB was included. T cell mediated killing of SKOV3 cells was determined after 24 h and 48 h by LDH release. After 48 h the T cells were harvested and CD69 and CD25 upregulation on CD4 T cells and CD8 T cells was measured by flow cytometry.

The kappa lambda FolR1 construct induces killing of SKOV3 cells in a concentration dependent manner which is accompanied by CD69 and CD25 upregulation both on CD4 T cells and on CD8 T cells.

SKOV3 cells were incubated with PBMCs in the presence of either kappa lambda FoLR1 TCB or DP47 TCB. After 24 h and 48 h killing of tumor cells was determined by measuring LDH release (FIG. 21). SKOV3 cells were incubated with PBMCs in the presence of either kappa lambda FoLR1 TCB or DP47 TCB. After 48 h CD25 and CD69 upregulation on CD4 T cells and CD8 T cells was measured by flow cytometry (FIG. 22).

Example 31 Biochemical Characterization of 16D5 and 36F2 FolR1 Binders by Surface Plasmon Resonance

Binding of anti-FolR1 16D5 in different monovalent or bivalent T-cell bispecific formats and of anti-FolR1 36F2 as IgG or as T-cell bispecific to recombinant human, cynomolgus and murine folate receptor 1 (all as Fc fusions) was assessed by surface plasmon resonance (SPR). All SPR experiments were performed on a Biacore T200 at 25° C. with HBS-EP as running buffer (0.01 M HEPES pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Surfactant P20, Biacore, GE Healthcare).

1. Molecules Tested

The molecules used for affinity and avidity determination are described in Table 24.

TABLE 24 Name and description of the 6 constructs used in SPR analysis Name Description 16D5 TCB 2 + 1 T-cell bispecific, inverted format (common light chain) 16D5 TCB 2 + 1 T-cell bispecific, classical classical format (common light chain) 16D5 TCB 1 + 1 1 + 1 T-cell bispecific (common light chain) 16D5 TCB 1 + 1 T-cell bispecific head- 1 + 1 HT to-tail (common light chain) 36F2 IgG Human IgG1 with P329G LALA 36F2 TCB 2 + 1 T-cell bispecific, inverted format, crossfab

2. Avidity to Folate Receptor 1

The avidity of the interaction between the anti-FolR1 IgG or T cell bispecifics and the recombinant folate receptors was determined as described below (Table 25).

Recombinant biotinylated monomeric Fc fusions of human, cynomolgus and murine Folate Receptor 1 (FolR1-Fc) were directly coupled on a SA chip using the standard coupling instruction (Biacore, GE Healthcare). The immobilization level was about 300-400 RU. The anti-FolR1 IgGs or T cell bispecifics were passed at a concentration range from 3.7 to 900 nM with a flow of 30 μL/minutes through the flow cells over 180 seconds. The dissociation was monitored for 240 or 600 seconds. The chip surface was regenerated after every cycle using a double injection of 30 sec 10 mM Glycine-HCl pH 2. Bulk refractive index differences were corrected for by subtracting the response obtained on reference flow cell immobilized with recombinant biotinylated murine CD134 Fc fusion. The binding curves resulting from the bivalent binding of the IgG or T cell bispecifics were approximated to a 1:1 Langmuir binding (even though it is a 1:2 binding) and fitted with that model to get an apparent KD representing the avidity of the bivalent binding. The apparent avidity constants for the interactions were derived from the rate constants of the fitting using the Bia Evaluation software (GE Healthcare). For the 1+1 T cell bispecifics format the interaction is a real 1:1 and the KD represents affinity since there is only one FolR1 binder in this construct.

TABLE 25 Bivalent binding (avidity with apparent KD) of anti-FolR1 16D5 and 36F2 as IgG or as T-cell bispecifics (TCB) on human, cyno and murine FolR1. Apparent Analyte Ligand ka (1/Ms) kd (1/s) KD 36F2 IgG huFolR1 2.07E+06  1.3E−02 6 nM cyFolR1 2.78E+06 1.75E−02 6 nM muFolR1 4.28E+05 8.23E−04 2 nM 36F2 TCB huFolR1 2.45E+06 9.120E−03   4 nM cyFolR1 4.31E+06 1.45E−02 3 nM muFolR1 6.97E+05 9.51E−04 1 nM 16D5 TCB huFolR1 1.57E+05 3.92E−04 3 nM cyFolR1 2.01E+05 3.81E−04 2 nM 16D5 TCB huFolR1 2.04E+05 1.84E−04 0.9 nM classical cyFolR1 2.50E+05 3.05E−04 1 nM 16D5 TCB huFolR1 5.00E+04 2.25E−03 45 nM 1 + 1 HT cyFolR1 5.75E+04 4.10E−03 70 nM 16D5 TCB huFolR1 3.65E+04 2.04E−03 56 nM 1 + 1 cyFolR1 4.09E+04 3.60E−03 90 nM

3. Affinity to Folate Receptor 1

The affinity of the interaction between the anti-FolR1 IgG or T cell bispecifics and the recombinant folate receptors was determined as described below (Table 26).

For affinity measurement, direct coupling of around 12000 resonance units (RU) of the anti-human Fab specific antibody (Fab capture kit, GE Healthcare) was performed on a CM5 chip at pH 5.0 using the standard amine coupling kit (GE Healthcare). Anti-FolR1 IgG or T cell bispecifics were captured at 20 nM with a flow rate of 10 μl/min for 40 sec, the reference flow cell was left without capture. Dilution series (12.3 to 3000 nM) of human, cyno or murine Folate Receptor 1 Fc fusion were passed on all flow cells at 30 μl/min for 240 sec to record the association phase. The dissociation phase was monitored for 300 s and triggered by switching from the sample solution to HBS-EP. The chip surface was regenerated after every cycle using a double injection of 60 sec 10 mM Glycine-HCl pH 1.5. Bulk refractive index differences were corrected for by subtracting the response obtained on the reference flow cell 1. The affinity constants for the interactions were derived from the rate constants by fitting to a 1:1 Langmuir binding using the Bia Evaluation software (GE Healthcare).

TABLE 26 Monovalent binding (affinity) of anti-16D5 and 36F2 as IgG or as T-cell bispecifics (TCB) on human, cyno and murine FolRl. Analyte Ligand ka (1/Ms) kd (1/s) KD 36F2 IgG huFolR1 9.10E+04 6.65E−02 730 nM cyFolR1 1.02E+05 5.78E−02 570 nM muFolR1 8.32E+04 1.78E−02 210 nM 36F2 TCB huFolR1 5.94E+04 6.13E−02 1000 nM cyFolR1 6.29E+04 5.42E−02 860 nM muFolR1 5.68E+04 1.75E−02 300 nM 16D5 TCB huFolR1 2.23E+04 7.33E−04 33 nM cyFolR1 1.57E+04 1.60E−03 100 nM 16D5 TCB huFolR1 1.03E+04 7.59E−04 74 nM classical cyFolR1 9.18E+03 1.61E−03 175 nM 16D5 TCB huFolR1 2.05E+04 7.08E−04 35 nM 1 + 1 HT cyFolR1 1.67E+04 1.53E−03 92 nM 16D5 TCB huFolR1 1.43E+04 9.91E−04 69 nM 1 + 1 cyFolR1 1.20E+04 1.80E−03 150 nM

The affinity (monovalent binding) to human and cyno FolR1-Fc of 36F2 TCB is similar and around 1000 nM for both, whereas the affinity to murine FolR1-Fc is slightly better and around 300 nM. The 36F2 can be used in murine and primate models, there is no need for a surrogate. The avidity (apparent KD) of 36F2 TCB to human FolR1 is around 30 times lower than the affinity of the 16D5 TCB to human FolR1. In the bivalent format, 36F2 TCB is in the low nanomolar range, whereas 16D5 TCB is in the low picomolar range (1000 fold difference).

FolR1 is expressed on tumor cells overexpressed, at intermittent and high levels, on the surface of cancer cells in a spectrum of epithelial malignancies, including ovarian, breast, renal, colorectal, lung and other solid cancers and is also expressed on the apical surface of a limited subset of polarized epithelial cells in normal tissue. These non-tumorous, normal cells express FolR1 only at low levels, and include, e.g., bronchiolal epithelial cells on alveolar surface, renal cortical luminal border of tubular cells, retinal pigment epithelium (basolateral membrane) and choroid plexus.

16D5 TCB binds to normal tissues cells expressing low amounts of FolR1 which results in their T cell mediated killing. This might, at least in part, account for limited tolerance observed at 10 μg/kg in cynomolgus monkeys. The inventors wanted to determine if lowering the affinity of the T cell bispecific molecule could increase the differentiation between high and low target density tissues and, thereby, lower toxicity by making use of bivalent binding and avidity. Low affinity binders are ordinarily not selected as suitable candidates for further analysis because low affinity is often associated with low potency and efficacy. Nevertheless, the low affinity FolR1 binder 36F2 was developed in several formats and characterized for its biological properties. For the 36F2 used in the bivalent T cell bispecific format the avidity effect (difference between monovalent and bivalent binding) is around 250 fold (1000 nM versus 4 nM). At low target density the affinity defined the interaction and with 1000 nM led to a low potency of the TCB. However, at high target density the molecule's avidity comes into play and with 4 nM led to a high activity of the TCB (see Example 32).

In an alternatively approach, the inventors generated monovalent formats of 16D5 and low affinity variant of 16D5 (affinity about 10-40 nM) in a bivalent format. The 16D5 binder used in a monovalent format (1+1) has an affinity of about 50 nM. The differentiation between high and low target density tissues can be better achieved by taking advantage of the avidity effect.

Example 32 T-Cell Killing of SKov-3 Cells Induced by 36F2 TCB, Mov19 TCB and 21A5 TCB

T-cell killing mediated by 36F2 TCB, Mov19 TCB and 21A5 TCB was assessed on SKov-3 cells (medium FolR1). Human PBMCs were used as effectors and the killing was detected at 24 h and 48 h of incubation with the bispecific antibodies. Briefly, target cells were harvested with Trypsin/EDTA, washed, and plated at density of 25 000 cells/well using flat-bottom 96-well plates. Cells were left to adhere overnight. Peripheral blood mononuclear cells (PBMCs) were prepared by Histopaque density centrifugation of enriched lymphocyte preparations (buffy coats) obtained from healthy human donors. Fresh blood was diluted with sterile PBS and layered over Histopaque gradient (Sigma, #H8889). After centrifugation (450×g, 30 minutes, room temperature), the plasma above the PBMC-containing interphase was discarded and PBMCs transferred in a new falcon tube subsequently filled with 50 ml of PBS. The mixture was centrifuged (400×g, 10 minutes, room temperature), the supernatant discarded and the PBMC pellet washed twice with sterile PBS (centrifugation steps 350×g, 10 minutes). The resulting PBMC population was counted automatically (ViCell) and stored in RPMI1640 medium containing 10% FCS and 1% L-alanyl-L-glutamine (Biochrom, K0302) at 37° C., 5% CO2 in cell incubator until further use (no longer than 24 h). For the killing assay, the antibody was added at the indicated concentrations (range of 0.005 pM-5 nM in triplicates). PBMCs were added to target cells at final E:T ratio of 10:1. Target cell killing was assessed after 24 h and 48 h of incubation at 37° C., 5% CO2 by quantification of LDH released into cell supernatants by apoptotic/necrotic cells (LDH detection kit, Roche Applied Science, #11 644 793 001). Maximal lysis of the target cells (=100%) was achieved by incubation of target cells with 1% Triton X-100. Minimal lysis (=0%) refers to target cells co-incubated with effector cells without bispecific construct.

The results show that the killing induced by 36F2 is strongly reduced in comparison to Mov19 TCB and 21A5 TCB (FIGS. 23A-B). The EC50 values related to killing assays, calculated using GraphPadPrism6 are summarized in Table 27.

TABLE 27 EC50 values (pM) for T-cell mediated killing of FolR1-expressing SKov-3 cells induced by 36F2 TCB, Mov19 TCB and 21A5 TCB. EC50 [pM] Antibody 24 h 48 h 36F2 TCB 1406.07* 134.5 Mov19 TCB 0.75 0.05 21A5 TCB 2.83 0.10 *curve did not reach saturation, value is hypothetical

Example 33 T-Cell Killing Induced by 36F2 TCB and 16D5 TCB in Different Monovalent and Bivalent T-Cell Bispecific Formats

T-cell killing mediated by 36F2 TCB, 16D5 TCB, 16D5 TCB classical, 16D5 TCB 1+1 and 16D5 TCB HT antibodies of Hela (high FolR1, about 2 million copies, Table 14, FIG. 27), Skov-3 (medium FolR1, about 70000-90000 copies, Table 14, FIG. 27) and HT-29 (low FolR1, about 10000, Table 14, FIG. 27) human tumor cells was assessed. DP47 TCB antibody was included as negative control. Human PBMCs were used as effectors and the killing was detected at 24 h of incubation with the bispecific antibody. Briefly, target cells were harvested with Trypsin/EDTA, washed, and plated at density of 25 000 cells/well using flat-bottom 96-well plates. Cells were left to adhere overnight. Peripheral blood mononuclear cells (PBMCs) were prepared by Histopaque density centrifugation of enriched lymphocyte preparations (buffy coats) obtained from healthy human donors. Fresh blood was diluted with sterile PBS and layered over Histopaque gradient (Sigma, #H8889). After centrifugation (450×g, 30 minutes, room temperature), the plasma above the PBMC-containing interphase was discarded and PBMCs transferred in a new falcon tube subsequently filled with 50 ml of PBS. The mixture was centrifuged (400×g, 10 minutes, room temperature), the supernatant discarded and the PBMC pellet washed twice with sterile PBS (centrifugation steps 350×g, 10 minutes). The resulting PBMC population was counted automatically (ViCell) and stored in RPMI1640 medium containing 10% FCS and 1% L-alanyl-L-glutamine (Biochrom, K0302) at 37° C., 5% CO2 in cell incubator until further use (no longer than 24 h). For the killing assay, the antibody was added at the indicated concentrations (range of 0.01 pM-100 nM in triplicates). PBMCs were added to target cells at final E:T ratio of 10:1. Target cell killing was assessed after 24 h of incubation at 37° C., 5% CO2 by quantification of LDH released into cell supernatants by apoptotic/necrotic cells (LDH detection kit, Roche Applied Science, #11 644 793 001). Maximal lysis of the target cells (=100%) was achieved by incubation of target cells with 1% Triton X-100. Minimal lysis (=0%) refers to target cells co-incubated with effector cells without bispecific construct.

The results show that target-specific killing of all three FolR1+ target cell lines induced by 36F2 TCB is much weaker compared to the killing induced by 16D5 TCB (FIGS. 24A-C, Table 29). Target-specific killing induced by the monovalent 16D5 TCBs (16D5 HT and 16D5 1+1) is worse compared to the bivalent 16D5 TCBs (16D5 TCB and 16D5 TCB classical). The EC50 values related to killing assays, calculated using GraphPadPrism6, are summarized in Table 28. Importantly, this data shows that using the 36F2 FolR1 binder in the bivalent 2+1 TCB format widens the therapeutic window compared to the 16D5 FOLR1 TCB (FIG. 24A-C). Whereas the potency reduction between 16D5 and 36F2 FOLR1 TCB is approximately 45-fold for Hela cells (high FOLR1 expression, see Table 28: 16D5 TCB=0.8 versus 36F2 TCB 36.0) and approximately 297-fold for Skov3 cells (medium FOLR1 expression, see Table 28: 16D5 TCB=0.6 versus 36F2 TCB 178.4), this reduction is almost 7000-fold for HT29 with low FOLR1 expression (see Table 28: 16D5 TCB=5.7 versus 36F2 TCB 39573). Thus, the 36F2 FOLR1 TCB differentiates between high and low expressing cells which is of special importance to reduce toxicity as the cells of some normal, non-tumorous tissues express very low levels of FolR1 (approximately less than 1000 copies per cell). Consistent with this observation, the results discussed in Example 35 below show that 36F2 TCB does not induce T-cell killing of primary cells (FIGS. 26A-D) whereas for 16D5 TCB some killing can be observed on HRCEpiC and HRPEpiC cells after 48 h of incubation (FIGS. 26B and C). This important characteristic of 36F2 TCB allows for dosing for the treatment of FolR1-positive tumors so that it mediates potent killing of tumor tissues with high or medium FOLR1 expression, but not of normal tissues with low (partially polarized) expression. Notably, this characteristic appears to be mediated by the avidity of 36F2 TCB in the bivalent 2+1 inverted format, as it was not observed when using the 1+1 monovalent formats carrying the same low affinity 36F2 binder.

Stated another way, 36F2 TCB in the bivalent 2+1 format comprises FolR1 binding moieties of relatively low affinity but it possesses an avidity effect which allows for differentiation between high and low FolR1 expressing cells. Because tumor cells express FolR1 at high or intermediate levels, this TCB selectively binds to tumor cells and not normal, non-cancerous cells that express FolR1 at low levels or not at all.

In addition to the above advantageous characteristics, the 36F2 TCB in the bivalent 2+1 inverted format also has the advantage that it does not require chemical cross linking or other hybrid approach. This makes it suitable for manufacture of a medicament to treat patients, for example patients having FolR1-positive cancerous tumors. The 36F2 TCB in the bivalent 2+1 inverted format can be produced using standard CHO processes with low aggregates. Further, the 36F2 TCB in the bivalent 2+1 comprises human and humanized sequences making it superior to molecules that employ rat and murine polypeptides that are highly immunogenic when administered to humans. Furthermore, the 36F2 TCB in the bivalent 2+1 format was engineered to abolish FcgR binding and, as such, does not cause FcgR crosslinking and infusion reactions, further enhancing its safety when administered to patients.

As demonstrated by the results described above, its head-to-tail geometry make the 36F2 TCB in the bivalent 2+1 inverted format a highly potent molecule that induces absolute target cell killing. Its bivalency enhance avidity and potency, but also allow for differentiation between high and low expressing cells. Its preference for high or medium target expressing cells due to its avidity affect reduce toxicity resulting from T cell mediated killing of normal cells that express FolR1 at low levels.

A further advantage of the 36F2 TCB in the bivalent 2+1 format and other embodiments disclosed herein is that their clinical development does not require the use of surrogate molecules as they bind to human, cynomous and murine FolR1. As such, the molecules disclosed herein recognize a different epitope than antibodies to FolR1 previously described that do not recognize FolR1 from all three species.

TABLE 28 EC50 values (pM) for T-cell mediated killing of FolR1-expressing tumor cells induced by 36F2 TCB and16D5 TCB in different monovalent and bivalent T-cell bispecific formats after 24 h of incubation. Skov-3 Hela (FolR1 HT-29 Antibody (FolR1 high) medium) (FolR1 low) 16D5 TCB 0.8 0.6 5.7 16D5 TCB 4.6 2.0 13.0 classical 16D5 TCB 11.6 12.3 15.1 HT 16D5 TCB 23.8 48.9 883.8* 1 + 1 36F2 TCB 36.0 178.4 39573.0* *curve did not reach saturation, only hypothetical value

Table 29 shows a comparison of EC50 values of 16D5 TCB and 36F2 TCB on the different cell lines tested. Out of the obtained EC50 values the delta (EC50 of 16D5 TCB minus EC50 of 36F2 TCB) and the x-fold difference (EC50 of 16D5 TCB divided by the EC50 of 36F2 TCB) was calculated.

TABLE 29 Comparison of EC50 values of 16D5 TCB and 36F2 TCB. Hela Skov-3 HT-29 Antibody (FolR1 high) (FolR1 medium) (FolR1 low) 16D5 TCB 0.82 0.63 5.73 36F2 TCB 35.99 178.40 39573.00* Δ 35.17 177.77 39567.27 x-fold 43.83 284.61 6906.58 *curve did not reach saturation, only hypothetical value

The calculated EC50 values clearly show that the difference between 36F2 TCB and 16D5 TCB gets larger the lower the FolR1 expression on the target cells is.

The same calculations as done for the comparison of the EC50 values of 16D5 TCB and 36F2 TCB were done for 16D5 TCB and the two monovalent 16D5 TCBs (16D5 TCB HT and 16D5 1+1). Tables 30 and 31 show the comparisons of the EC50 values of 16D5 TCB vs 16D5 TCB HT (Table 30) and 16D5 TCB vs 16D5 TCB 1+1 (Table 31) as well as the corresponding deltas (EC50 of 16D5 TCB minus EC50 of 16D5 TCB HT/1+1) and the x-fold differences (EC50 of 16D5 TCB divided by the EC50 of 16D5 TCB HT/1+1).

TABLE 30 Comparison of EC50 values of 16D5 TCB (2 + 1 inverted) and 16D5 TCB HT. Hela Skov-3 HT-29 Antibody (FolR1 high) (FolR1 medium) (FolR1 low) 16D5 TCB 0.82 0.63 5.73 16D5 TCB HT 11.61 12.27 15.11 Δ 10.79 11.65 9.38 x-fold 14.14 19.58 2.64

TABLE 31 Comparison of EC50 values of 16D5 TCB and 16D5 TCB 1 + 1. Hela Skov-3 HT-29 Antibody (FolR1 high) (FolR1 medium) (FolR1 low) 16D5 TCB 0.82 0.63 5.73 16D5 TCB 1 + 1 23.84 48.86 883.78* Δ 23.02 48.24 878.05 x-fold 29.03 77.95 154.24 *curve did not reach saturation, only hypothetical value

The comparison of the EC50 values of 16D5 TCB and 36F2 TCB (Table 29) shows that the difference in the EC50 values gets larger the lower the FolR1 expression on the target cells is. This effect cannot be seen in the comparison of 16D5 TCB and the monovalent 16D5 TCBs (Table 29 and Table 30). For 16D5 TCB 1+1 (Table 31) there is also a slight increase in the difference between the EC50 of 16D5 TCB and 16D5 TCB 1+1 with decreasing FolR1 expression but by far not as pronounced as can be seen in the comparison of 16D5 TCB vs 36F2 TCB.

Example 34 CD25 and CD69 Upregulation on CD8+ and CD4+ Effector Cells after T Cell-Killing of FolR1-Expressing Tumor Cells Induced by 36F2 TCB and 16D5 TCB Antibody

Activation of CD8+ and CD4 T cells after T-cell killing of FolR1-expressing Hela, SKov-3 and HT-29 tumor cells mediated by 36F2 TCB and 16D5 TCB was assessed by FACS analysis using antibodies recognizing the T cell activation markers CD25 (late activation marker) and CD69 (early activation marker). DP47 TCB was included as non-binding control. The antibody and the killing assay conditions were essentially as described above (Example 32) using the same antibody concentration range (0.01 pM-100 nM in triplicates), E:T ratio 10:1 and an incubation time of 48 h.

After the incubation, PBMCs were transferred to a round-bottom 96-well plate, centrifuged at 400×g for 4 min and washed twice with PBS containing 0.1% BSA. Surface staining for CD8 (PE anti-human CD8, BD #555635), CD4 (Brilliant Violet 421™ anti-human CD4, Biolegend #300532), CD69 (FITC anti-human CD69, BD #555530) and CD25 (APC anti-human CD25 BD #555434) was performed according to the manufacturer's instructions. Cells were washed twice with 150 μl/well PBS containing 0.1% BSA. After centrifugation, the samples were resuspended in 200 μl/well PBS 0.1% for the FACS measurement. Samples were analyzed at BD FACS Canto II.

36F2 TCB induced a target-specific up-regulation of activation markers (CD25, CD69) on CD8+ and CD4+ T cells after killing of Hela (FIG. 25A) and SKov-3 (FIG. 25B) cells. In comparison to 16D5 TCB the up-regulation of CD25 and CD69 on CD8+ and CD4+ T cells induced by 36F2 is much weaker.

On HT-29 (low FolR1) an up-regulation of activation markers can only be seen at the highest concentration of 36F2 TCB. In contrast, with 16D5 TCB up-regulation of CD25 and CD69 can be seen already at much lower antibody concentrations (FIG. 25C).

As seen as well in the tumor lysis experiment, the analysis of activation markers (CD25 and CD69) on T cells (CD4+ and CD8+) after killing clearly shows that the difference between 16D5 TCB and 36F2 TCB becomes larger the lower the FolR1 expression level on the target cells is.

Example 35 T-Cell Killing of Primary Cells Induced by 36F2 TCB and 16D5 TCB

T-cell killing mediated by 36F2 TCB and 16D5 TCB was assessed on primary cells (Human Renal Cortical Epithelial Cells (HRCEpiC) (ScienCell Research Laboratories; Cat No 4110) and Human Retinal Pigment Epithelial Cells (HRPEpiC) (ScienCell Research Laboratories; Cat No 6540)). HT-29 cells (low FolR1) were included as control cell line. DP47 TCB served as non-binding control. Human PBMCs were used as effectors and the killing was detected at 24 h and 48 h of incubation with the bispecific antibodies. Briefly, target cells were harvested with Trypsin/EDTA, washed, and plated at density of 25 000 cells/well using flat-bottom 96-well plates. Cells were left to adhere overnight. Peripheral blood mononuclear cells (PBMCs) were prepared by Histopaque density centrifugation of enriched lymphocyte preparations (buffy coats) obtained from healthy human donors. Fresh blood was diluted with sterile PBS and layered over Histopaque gradient (Sigma, #H8889). After centrifugation (450×g, 30 minutes, room temperature), the plasma above the PBMC-containing interphase was discarded and PBMCs transferred in a new falcon tube subsequently filled with 50 ml of PBS. The mixture was centrifuged (400×g, 10 minutes, room temperature), the supernatant discarded and the PBMC pellet washed twice with sterile PBS (centrifugation steps 350×g, 10 minutes). The resulting PBMC population was counted automatically (ViCell) and stored in RPMI1640 medium containing 10% FCS and 1% L-alanyl-L-glutamine (Biochrom, K0302) at 37° C., 5% CO2 in cell incubator until further use (no longer than 24 h). For the killing assay, the antibody was added at the indicated concentrations (range of 0.01 pM-10 nM in triplicates). PBMCs were added to target cells at final E:T ratio of 10:1. Target cell killing was assessed after 24 h and 48 h of incubation at 37° C., 5% CO2 by quantification of LDH released into cell supernatants by apoptotic/necrotic cells (LDH detection kit, Roche Applied Science, #11 644 793 001). Maximal lysis of the target cells (=100%) was achieved by incubation of target cells with 1% Triton X-100. Minimal lysis (=0%) refers to target cells co-incubated with effector cells without bispecific construct.

The results show that 36F2 TCB does not induce T-cell killing of primary cells (FIG. 26A-D) whereas for 16D5 TCB some killing can be observed on HRCEpiC and HRPEpiC cells after 48 h of incubation (FIG. 26B and D). As described above, a strong difference in T-cell killing between of HT-29 cells was observed between 16D5 TCB and 36F2 TCB (FIG. 26E, F).

Example 36 Preparation of DP47 GS TCB (2+1 Crossfab-IgG P329G LALA Inverted=“Untargeted TCB”)

The “untargeted TCB” was used as a control in the above experiments. The bispecific antibody engages CD3e but does not bind to any other antigen and therefore cannot crosslink T cells to any target cells (and subsequently cannot induce any killing). It was therefore used as negative control in the assays to monitor any unspecific T cell activation. This untargeted TCB was prepared as described in WO2014/131712. In brief, the variable region of heavy and light chain DNA sequences have been subcloned in frame with either the constant heavy chain or the constant light chain pre-inserted into the respective recipient mammalian expression vector. The antibody expression was driven by an MPSV promoter and carries a synthetic polyA signal sequence at the 3′ end of the CDS. In addition each vector contains an EBV OriP sequence.

The molecule was produced by co-transfecting HEK293-EBNA cells with the mammalian expression vectors using polyethylenimine. The cells were transfected with the corresponding expression vectors in a 1:2:1:1 ratio (“vector heavy chain Fc(hole)”: “vector light chain”: “vector light chain Crossfab”: “vector heavy chain Fc(knob)-FabCrossfab”).

For transfection HEK293 EBNA cells were cultivated in suspension serum free in CD CHO culture medium. For the production in 500 ml shake flask 400 million HEK293 EBNA cells were seeded 24 hours before transfection. For transfection cells were centrifuged for 5 min by 210×g, supernatant is replaced by pre-warmed 20 ml CD CHO medium. Expression vectors were mixed in 20 ml CD CHO medium to a final amount of 200 g DNA. After addition of 540 μl PEI solution was vortexed for 15 s and subsequently incubated for 10 min at room temperature. Afterwards cells were mixed with the DNA/PEI solution, transferred to a 500 ml shake flask and incubated for 3 hours by 37° C. in an incubator with a 5% CO2 atmosphere. After incubation time 160 ml F17 medium was added and cell were cultivated for 24 hours. One day after transfection 1 mM valporic acid and 7% Feed lwas added. After 7 days cultivation supernatant was collected for purification by centrifugation for 15 min at 210×g, the solution was sterile filtered (0.22·m filter) and sodium azide in a final concentration of 0.01% w/v was added, and kept at 4° C.

The secreted protein was purified from cell culture supernatants by affinity chromatography using ProteinA. Supernatant was loaded on a HiTrap ProteinA HP column (CV=5 mL, GE Healthcare) equilibrated with 40 ml 20 mM sodium phosphate, 20 mM sodium citrate, 0.5 M sodium chloride, pH 7.5. Unbound protein was removed by washing with at least 10 column volume 20 mM sodium phosphate, 20 mM sodium citrate, 0.5 M sodium chloride, pH 7.5. Target protein was eluted during a gradient over 20 column volume from 20 mM sodium citrate, 0.5 M sodium chloride, pH 7.5 to 20 mM sodium citrate, 0.5 M sodium chloride, pH 2.5. Protein solution was neutralized by adding 1/10 of 0.5 M sodium phosphate, pH 8. Target protein was concentrated and filtrated prior loading on a HiLoad Superdex 200 column (GE Healthcare) equilibrated with 20 mM Histidine, 140 mM sodium chloride solution of pH 6.0.

The protein concentration of purified protein samples was determined by measuring the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence.

Purity and molecular weight of molecules were analyzed by CE-SDS analyses in the presence and absence of a reducing agent. The Caliper LabChip GXII system (Caliper lifescience) was used according to the manufacturer's instruction. 2ug sample is used for analyses.

The aggregate content of antibody samples was analyzed using a TSKgel G3000 SW XL analytical size-exclusion column (Tosoh) in 25 mM K2HPO4, 125 mM NaCl, 200 mM L-Arginine Monohydrocloride, 0.02% (w/v) NaN3, pH 6.7 running buffer at 25° C.

TABLE 32 Summary production and purification of DP47 GS TCB. Aggregate after 1st Mono- Titer Yield purification HMW LMW mer Construct [mg/l] [mg/l] step [%] [%] [%] [%] DP47 103.7 8.04 8 2.3 6.9 91.8 GS TCB

TABLE 33 CE-SDS analyses of DP47 GS TCB. Peak kDa Corresponding Chain DP47 GS TCB non reduced 1 165.22 Molecule with 2 missing (A) light chains 2 181.35 Molecule with 1 missing light chain 3 190.58 Correct molecule without N-linked glycosylation 4 198.98 Correct molecule DP47 GS TCB reduced (B) 1 27.86 Light chain DP47 GS 2 35.74 Light chain huCH2527 3 63.57 Fc (hole) 4 93.02 Fc (knob)

Example 37 Binding of 16D5 TCB and 9D11 TCB and their corresponding CD3 Deamidation Variants N100A and S100aA to CD3-Expressing Jurkat Cells

The binding of 16D5 TCB and the corresponding CD3 deamidation variants 16D5 TCB N100A and 16D5 TCB S100aA and 9D11 TCB and the deamidation variants 9D11 TCB N100A and 9D11 TCB S100aA to human CD3 was assessed on a CD3-expressing immortalized T lymphocyte line (Jurkat). Briefly, cells were harvested, counted, checked for viability and resuspended at 2×106 cells/ml in FACS buffer (100 μl PBS 0.1% BSA). 100 μl of cell suspension (containing 0.2×106 cells) was incubated in round-bottom 96-well plates for 30 min at 4° C. with different concentrations of the bispecific antibodies (686 pM-500 nM). After two washing steps with cold PBS 0.1% BSA, samples were re-incubated for further 30 min at 4° C. with a PE-conjugated AffiniPure F(ab′)2 Fragment goat anti-human IgG Fcg Fragment Specific secondary antibody (Jackson Immuno Research Lab PE #109-116-170). After washing the samples twice with cold PBS 0.1% BSA they were immediately analyzed by FACS using a FACS Cantoll (Software FACS Diva). Binding curves were obtained using GraphPadPrism6 (FIG. 28A-B).

The results show reduced binding of the deamidation variants N100A and S100aA to CD3 compared to the parental antibodies 16D5 TCB (FIG. 28A) and 9D11 TCB (FIG. 28B).

Example 38 T-Cell Killing of SKov-3 and HT-29 Cells induced by 16D5 TCB and 9D11 TCB and their CD3 Deamidation Variants N100A and S100aA

T-cell killing mediated by 16D5 TCB and the corresponding CD3 deamidation variants 16D5 TCB N100A and 16D5 TCB S100aA and 9D11 TCB and the deamidation variants 9D11 TCB N100A and 9D11 TCB S100aA was assessed on SKov-3 (medium FolR1) and HT-29 (low FolR1) cells. Human PBMCs were used as effectors and the killing was detected at 24 h of incubation with the bispecific antibodies. Briefly, target cells were harvested with Trypsin/EDTA, washed, and plated at a density of 25 000 cells/well using flat-bottom 96-well plates. Cells were left to adhere overnight. Peripheral blood mononuclear cells (PBMCs) were prepared by Histopaque density centrifugation of enriched lymphocyte preparations (buffy coats) obtained from healthy human donors. Fresh blood was diluted with sterile PBS and layered over Histopaque gradient (Sigma, #H8889). After centrifugation (450×g, 30 minutes, room temperature), the plasma above the PBMC-containing interphase was discarded and PBMCs transferred in a new falcon tube subsequently filled with 50 ml of PBS. The mixture was centrifuged (400×g, 10 minutes, room temperature), the supernatant discarded and the PBMC pellet washed twice with sterile PBS (centrifugation steps 350×g, 10 minutes). The resulting PBMC population was counted automatically (ViCell) and stored in RPMI1640 medium containing 10% FCS and 1% L-alanyl-L-glutamine (Biochrom, K0302) at 37° C., 5% CO2 in cell incubator until further use (no longer than 24 h). For the killing assay, the antibody was added at the indicated concentrations (range of 0.01 pM-10 nM in triplicates). PBMCs were added to target cells at final E:T ratio of 10:1. Target cell killing was assessed after 24 h of incubation at 37° C., 5% CO2 by quantification of LDH released into cell supernatants by apoptotic/necrotic cells (LDH detection kit, Roche Applied Science, #11 644 793 001). Maximal lysis of the target cells (=100%) was achieved by incubation of target cells with 1% Triton X-100. Minimal lysis (=0%) refers to target cells co-incubated with effector cells without bispecific construct.

The results show that on SKov-3 cells the killing induced by the CD3 deamidation variants 16D5 TCB N100A and 16D5 S100aA is comparable to the one induced by 16D5 TCB (FIG. 29A). The same is true for 9D11 TCB and its variants 9D11 TCB N100A and 9D11 TCB S100aA (FIG. 29B). On FolR1 low expressing HT-29 cells the S100aA variant shows an impaired killing efficiency which is the case for 16D5 TCB (FIG. 30A) as well as for 9D11 TCB (FIG. 30B). The EC50 values related to killing assays, calculated using GraphPadPrism6 are given in Table 35.

TABLE 35 EC50 values (pM) for T-cell mediated killing of FolR1-expressing SKov-3 and HT-29 cells induced by 16D5 TCB and 9D11 TCB and their deamidation variants N100A and A100aA. EC50 [pM] Antibody SKov-3 HT-29 16D5 TCB 1.283 56.67 16D5 TCB 1.886 91.95 N100A 16D5 TCB 1.939 165.6 S100aA 9D11 TCB 1.283 2.827 9D11 TCB 1.886 37.72 N100A 9D11 TCB 1.939 n.d.* S100aA * not determined

Example 39 Generation of Mucin-1 T Cell Bi-Specific Constructs that Contain a Common Light Chain

Gene Synthesis

Desired gene segments were synthesized at Geneart AG (Regensburg, Germany) from synthetic oligonucleotides and PCR products by automated gene synthesis.

Production and Purification of MUC1 Antigen

For the generation of common light chain (CLC) antibodies against the “sea urchin sperm protein, enterokinase and agrin” (SEA) domain of human Mucin-1 (MUC1), a DNA fragment encoding SEA domain was synthesized (Uniprot P15941, amino acids 1041-1151). In order to prevent autoproteolysis of the SEA between positions G1097 and S1098, a process described previously, by Ligtenber et al. (1992). Cell-associated episialin is a complex containing two proteins derived from a common precursor. J Biol Chem 267, 6171-7. Parry et al., Identification of MUC1 Proteolytic Cleavage Sites in Vivo. Biochem Biophys Res Commun 283, 715-20, four additional glycine residues were inserted between G1097 and S1098. This insertion results in the relief of the conformational stress and the protein remains intact. The DNA fragment was inserted into pETXX, an inducible bacterial expression vector. The resulting plasmid expresses the SEA domain with a C-terminal avi tag and a His6 tag (SEQ ID NO: 47). While the avi tag was used for BirA-mediated in vivo biotinylation, the His6 tag (SEQ ID NO: 47) was used for purification.

A 500 ml culture was inoculated with the bacterial strain BL21 D3, transformed with the corresponding plasmid and a plasmid expressing BirA, and induced with 1 mM IPTG at an OD600 0.8. Afterwards, the cultures were incubated at 25° C. overnight and harvested by centrifugation. The bacterial pellet was resuspended with 25 ml BugBuster® Protein Extraction Reagent (Millipore) and incubated for 20 min at room temperature as described in the protocol. After centrifugation for 20 min at 16000×g, the supernatant was filtered and loaded on an IMAC column (His gravitrap, GE Healthcare). The column was washed with 40 ml washing buffer (500 mM NaCl, 20 mM Imidazole, 20 mM NaH2PO4 pH 7.4). After the elution from the column (500 mM NaCl, 500 mM Imidazole, 20 mM NaH2PO4 pH 7.4), the eluate was re-buffered using PD10 columns (GE Healthcare).

Selection of Anti-Human MUC1 SEA Domain Binders from the CLC Fab Libraries

Selections against the SEA domain of human MUC1 were carried out using E. coli-derived and in vivo biotinylated MUC1. The antigens were enzymatically biotinylated by co-expression of the biotin ligase Bir A via an C-terminal avi-tag. Panning rounds were performed in solution according to the following pattern: 1. Binding of the phagemid particles of the CLC libraries to 100 nM biotinylated antigen protein for 0.5 h in a total volume of 1 ml, 2. capture of biotinylated antigen and attached specifically binding phage by addition of 5.4×107 streptavidin-coated magnetic beads for 10 min, 3. washing of beads using 5×1 ml PBS/Tween20 and 5×1 ml PBS, 4. elution of phage particles by addition of 1 ml 100 mM triethylamine (TEA) for 10 min and neutralization by addition of 500 ul 1M Tris/HCl pH 7.4, 5. Re-infection of log-phase E. coli TG1 cells with the phage particles in the supernatant, infection with helperphage VCSM13 and subsequent PEG/NaCl precipitation of phagemid particles to be used in subsequent selection rounds. Selections were carried out over 3 rounds using either constant or decreasing (from 10−7M to 2×10−9M) antigen concentrations. In round 2, capture of antigen:phage complexes was performed using neutravidin plates instead of streptavidin beads. Specific binders were identified by ELISA as follows: 100 ul of 50 nM biotinylated antigen per well were coated on neutravidin plates. Fab-containing bacterial supernatants were added and binding Fabs were detected via their Flag-tags by using an anti-Flag/HRP secondary antibody. The VH domains of clones exhibiting significant signals over background were short-listed for sequencing (58D6 VH, 106D2 VH, 110A5 VH) and further analyses. All clones derive from the IGHV3-23 germline sequence (FIG. 31). Of note, clone 58D6 and 110A5 originate from a library that was randomized only in CDR3, while clone 106D2 was identified from a library randomized in all 3 CDRs. Positions in CDR1 and 2 that deviate from the germline sequence are printed italic. All VH variants were expressed in combination with the same common light chain (common light chain VL).

Purification of Fabs

Fabs from bacterial cultures (protein sequence of variable heavy chains domains for 58D6 VH, 106D2 VH, 110A5 VH, all clones expressed the same CLC variable domain listed as SEQ ID NO: 31) were purified for the exact analysis of the kinetic parameters. For each clone, a 500 ml culture was inoculated with bacteria harboring the corresponding phagemid and induced with 1 mM IPTG at an OD600 0.9. Afterwards, the cultures were incubated at 25° C. overnight and harvested by centrifugation. After the incubation of the resuspended pellet for 20 min in 25 ml PPB buffer (30 mM Tris-HCl pH8, 1 mM EDTA, 20% sucrose), bacteria were centrifuged again and the supernatant was harvested. This incubation step was repeated once with 25 ml of a 5 mM MgSO4 solution. The supernatants of both incubation steps were pooled, filtered and loaded on an IMAC column (His gravitrap, GE Healthcare). Subsequently, the column was washed with 40 ml washing buffer (500 mM NaCl, 20 mM Imidazole, 20 mM NaH2PO4 pH 7.4). After the elution (500 mM NaCl, 500 mM Imidazole, 20 mM NaH2PO4 pH 7.4) the eluate was re-buffered using PD10 columns (GE Healthcare). The kinetic parameters of the purified Fabs were then studied by SPR-analysis (Proteon XPR36, Biorad) in a dilution row that ranged from 100 nM to 6.25 nM.

Affinity-Determination by SPR Using BioRad's ProteOn XPR36 Biosensor

Affinity (KD) of selected Fab clones was measured by surface plasmon resonance using a ProteOn XPR36 instrument (Biorad) at 25° C. with biotinylated MUC1 antigen immobilized on NLC chips by neutravidin capture. Immobilization of recombinant antigens (ligand): Antigens were diluted with PBST (10 mM phosphate, 150 mM sodium chloride pH 7.4, 0.005% Tween 20) to 10 μg/ml, then injected at 30 μl/minute at varying contact times, to achieve immobilization levels of about 200 response units (RU) in vertical orientation. Injection of analytes: For one-shot kinetics measurements, injection direction was changed to horizontal orientation, two-fold dilution series of purified Fab (varying concentration ranges between 100 and 6.25 nM) were injected simultaneously at 50 μl/min along separate channels 1-5, with association times of 250 or 300 s, and dissociation times of 300 s. Buffer (PBST) was injected along the sixth channel to provide an “in-line” blank for referencing. Association rate constants (kon) and dissociation rate constants (koff) were calculated using a simple one-to-one Langmuir binding model in ProteOn Manager v3.1 software by simultaneously fitting the association and dissociation sensorgrams. The equilibrium dissociation constant (KD) was calculated as the ratio koff/kon. Regeneration was performed in horizontal orientation using 10 mM Glycine, pH 1.5 at a flow rate of 100 ul/min for a contact time of 30 s. 3 clones bound specifically to MUC1 (FIG. 32A), but not to the “in-line” blank demonstrating the specificity of these binders. The kinetic and thermodynamic data of all measurements are summarized in FIG. 32B.

Cloning, Expression and Characterization of the MUC1-Based

All Fabs demonstrating specific binding to MUC1 by SPR were converted into the T cell bi-specific (TCB) format. For this, the PCR-amplified DNA fragments of heavy and light chain VH-domains were inserted in frame into both Fc-containing Ig chains needed for the generation of a TCB (FIG. 33). The antibody chain expression is driven by an MPSV promoter and transcription is terminated by a synthetic polyA signal sequence located downstream of the CDS. In addition to the expression cassette, each vector contains an EBV oriP sequence for autonomous replication in EBV-EBNA expressing cell lines. The resulting DNA constructs were co-expressed in combination with the CLC (clone 58D6; clone 110A5) and purified from mammalian-derived cell culture supernatant (clone 58D6: clone 110A5). A summary of the analytical data for two bi-specific antibodies is shown clones in FIG. 34A and B.

Binding Analysis of the MUC1-Specific TCBs Using BioRad's ProteOn XPR36 Biosensor

Binding of the produced MUC1-specific TCBs was measured by surface plasmon resonance using a ProteOn XPR36 instrument (Biorad) at 25° C. Biotinylated MUC1 antigen and an unrelated biotinylated antigen were immobilized on NLC chips by neutravidin capture. Immobilization of the antigens was performed as described before. For one-shot kinetics measurements, injection direction was changed to horizontal orientation, two-fold dilution series of the purified constructs (varying concentration ranges between 30 and 1.88 nM or between 100 and 6.25 nM) were injected simultaneously at 50 μl/min along separate channels 1-5, with association times of 120 s, and dissociation times of up to 600 s. Buffer (PBST) was injected along the sixth channel to provide an “in-line” blank for referencing.

Regeneration was performed in horizontal orientation using 10 mM Glycine, pH 1.5 at a flow rate of 100 ul/min for a contact time of 30 s. 3 clones specifically bound to MUC1 (FIG. 35), but not to the “in-line” blank demonstrating the specificity of these binders. Due to the avidity effect of the bivalent TCB format, very strong binding to MUC1 was observed. In contrast, no binding to the unrelated antigen was detected indicating the specific binding of the TCB constructs.

Example 40 Generation of Anti-BCMA Antibodies

1.1: Production of Antigens and Tool Reagents

1.1.1: Recombinant, Soluble, Human BCMA Extracellular Domain

The extracellular domains of human, cynomolgus and murine BCMA that were used as antigens for phage display selections were transiently expressed as N-terminal monomeric Fc-fusion in HEK EBNA cells and in vivo site-specifically biotinylated via co-expression of BirA biotin ligase at the avi-tag recognition sequence located at the C-terminus of the Fc portion carrying the receptor chain (Fc knob chain). The extracellular domains of human, cynomolgus and murine BCMA comprised methionine 4 to asparagine 53, methionine 4 to asparagine 52, and alanine 2 to threonine 49, respectively. These were N-terminally fused to the hinge of a human IgG1 enabling heterodimerization with an unfused human IgG1 Fc portion (hole chain) by knobs-into-holes technology.

For recovering of the extracellular domain of BCMA the following primers were used:

(SEQ ID NO: 48) AAGCTTGGATCCATGTTGCAGATGGCTGGGCAGTGCTCC-3

incorporating a BamH1 site (bold, underlined) and

reverse primer (SEQ ID NO: 49) 5-GAATTCGCGGCCGCTCATCCTTTCACTGAATTGGTCACACTTGCATTA C-3 primer (SEQ ID NO: 50) 5-ACGTTAGATCTCCACTCAGTCCTGCATCTTGTTCCAGTTAAC-3 and reverse primer (SEQ ID NO: 51) 5-AACGTTGCGGCCGCTAGTTTCACAAACCCCAGG-3 (SEQ ID NO: 52) GAATTCAAGCTTGCCACCATGTTGCAGATGGCTGGGCAGTGCTCC-3

including a HindIII restriction site (bold, underlined) and Kozak consensus sequence and

reverse primer (SEQ ID NO: 53) 5-GAATTCTCTAGATTACCTAGCAGAAATTGATTTCTCTATCTCCGTAG C-3

Gene synthesis can be also used to obtain the extracellular domain of BCMA.

1.2: BCMA-Expressing Cells as Tools

1.2.1: Human Myeloma Cell Line Expressing BCMA on their Surface

BCMA expression was assessed on four human myeloma cell lines (NCI-H929, RPMI-8226, U266B1 and L-363) by flow cytometry. NCI-H929 cells ((H929) ATCC® CRL-9068™) were cultured in 80-90% RPMI 1640 with 10-20% heat-inactivated FCS and could contain 2 mM L-glutamine, 1 mM sodium pyruvate and 50 μM mercaptoethanol. RPMI-8226 cells ((RPMI) ATCC® CCL-155™) were cultured in a media containing 90% RPMI 1640 and 10% heat-inactivated FCS. U266B1 ((U266) ATCC® TIB-196™) cells were cultured in RPMI-1640 medium modified to contain 2 mM L-glutamine, 10 mM HEPES, 1 mM sodium pyruvate, 4500 mg/L glucose, and 1500 mg/L sodium bicarbonate and 15% heat-inactivated FCS. L-363 cell line (Leibniz Institute DSMZ—German collection of microorganisms and cell cultures; DSMZ No. ACC 49) was cultured in 85% RPMI 1640 and 15% heat-inactivated FCS. Briefly, cells were harvested, washed, counted for viability, resuspended at 50,000 cells/well of a 96-well round bottom plate and incubated with anti-human BCMA antibody (Abcam, #ab54834, mouse IgG1) at 10 μg/ml for 30 min at 4° C. (to prevent internalization). A mouse IgG1 was used as isotype control (BD Biosciences, #554121). Cells were then centrifuged (5 min at 350×g), washed twice and incubated with the FITC-conjugated anti mouse secondary antibody for 30 min at 4° C. At the end of incubation time, cells were centrifuged (5 min at 350×g), washed twice with FACS buffer, resuspended in 100 ul FACS buffer and analyzed on a Cantoll device running FACS Diva software. The relative quantification of BCMA receptor number on the surface membrane of H929, U266B1, RPMI-8226 and L-363 myeloma cell lines was assessed by QIFIKIT analysis (Dako, #K0078, following manufacturer's instructions). H929 cells expressed human BCMA with the highest density, up to 3.8 to 27-fold higher than other myeloma cell lines. H929 is considered as a BCMAhi-expressing myeloma cell line as compared to U266 which is BCMAmed/lo-expressing myeloma cells and RPMI-8226 and L363 which are BCMAlo-expressing myeloma cells. Table 36 summarizes the relative BCMA receptor number on the cell surface of human multiple myeloma cell lines.

TABLE 36 Quantification of BCMA receptor number on cell surface of NCI-H929, U266B1, RPMI-8226 and L-363 human myeloma cell lines Relative binding sites Myeloma cell lines per cell H929 50000 U266 13000 RPMI-8226 2000 L363 1800

1.3: Obtaining BCMA Binders Out of an In Vitro, Recombinant Library

1.3.1: Construction of Common Light Chain Fab-Libraries

Antibody libraries in the Fab-format are constructed on the basis of the humanized anti-CD3 antibody light chain were generated. Diversity was only introduced in the heavy chain. Six different heavy chain frameworks were used: VH1-46, VH1-69, VH3-15, VH3-23, VH4-59 and VH5-1. CDRs 1, 2 and 3 were randomized and each heavy chain library was combined with the non-randomized light chain. Libraries were generated as described by Nissim et al EMBO J. 1994 Feb. 1; 13(3):692-8, and Silacci et al. Proteomics. 2005 June; 5(9):2340-50.

1.3.2: Selection of Anti-BCMA Fab Clones

Anti-BCMA Fabs were established by phage display from synthetic Fab libraries consisting one constant VL and six different human VH sequences

TABLE 37 Anti-BCMA clones and respective VL/VH pairings Fab clone VL domain VH domain pSCHLI372 QAVVTQEPSLTVSPGGTVTLT EVQLVESGGGLVKPGGSLRLSC CGSSTGAVTTSNYANWVQEK AASGFTFSNSGMIWVRQAPGK PGQAFRGLIGGTNKRAPGTPA GLEWVGHIRSKTDGGTTDYAA RFSGSLLGGKAALTLSGAQPE PVKGRFTISRDDSKNTLYLQMN DEAEYYCALWYSNLWVFGG SLKTEDTAVYYCTTGGSGSFD GTKLTVL (SEQ ID NO: 31) YWGQGTLVTVSS (SEQ ID NO: 54) pSCHLI373 QAVVTQEPSLTVSPGGTVTLT EVQLVESGGGLVKPGGSLRLSC CGSSTGAVTTSNYANWVQEK AASGFSFSNSWMNWVRQAPG PGQAFRGLIGGTNKRAPGTPA KGLEWVGTIRQKTYGGTTDYA RFSGSLLGGKAALTLSGAQPE APVKGRFTISRDDSKNTLYLQM DEAEYYCALWYSNLWVFGG NSLKTEDTAVYYCTTGGLFGY GTKLTVL (SEQ ID NO: 31) WDYWGQGTLVTVSS (SEQ ID NO: 55)

Selection rounds (biopanning) were performed in solution according to the following pattern: 1) pre-clearing of ˜1012 phagemid particles per library pool in immunotubes coated with 10 ug/ml of an unrelated human IgG to deplete the libraries of antibodies recognizing the Fc-portion of the antigens; 2) incubation of the non-Fc-binding phagemid particles with 100 nM biotinylated BCMA for 0.5 h in the presence of 100 nM unrelated non-biotinylated Fc knobs-into-holes construct for further depletion of Fc-binders in a total volume of 2 ml; 3) capture of biotinylated BCMA and specifically binding phage by splitting up and transferring the panning reaction into 16 wells on a neutravidin or streptavidin pre-coated microtiter plate for 20 min on a shaker; 4) washing of respective wells 10-30× with PBS/Tween20 and 10-30× with PBS using a plate washer; 5) optional competitive washing step by addition of 230 nM murine APRIL to displace Fab clones that recognize the binding site of the natural ligand thus selecting for APRIL-non-competing phage antibodies; 6) elution of phage particles by addition of 125 ul 100 mM TEA (triethylamine) per well for 5-10 min and neutralization by addition of an equal volume of 1M Tris/HCl pH 7.4; 7) re-infection of log-phase E. coli TG1 cells with the eluted phage particles, infection with helperphage VCSM13, incubation on a shaker at 30° C. overnight and subsequent PEG/NaCl precipitation of phagemid particles to be used in the next selection round. Selections were carried out over 3 to 5 rounds using constant antigen concentrations of 100 nM. Apart from selection campaigns during which only human BCMA was used as antigen, additional selection campaigns were carried out during which also cynomolgus or murine BCMA were used in an alternating fashion with human BCMA in order to select for cross-reactive antibodies. Moreover, as an alternative to streptavidin plate-based capture, capture of antigen: phage complexes was performed by addition of 5.4×107 streptavidin-coated magnetic beads to the panning reaction followed by washing steps using respective magnets under the conditions described above.

Specific binders were identified by surface plasmon resonance-screening of Fab-containing bacterial culture supernatants using BioRad's ProteOn XPR36 biosensor. In brief, after infection of log-phase E. coli TG1 cells with the eluted phage particles, single colony forming units (cfu) were plated and picked for inoculation of 1 ml expression cultures in 96-deep well plates. Fabs were captured from the supernatants on a ProteOn GLM chip that was derivatized with 8.000-10.000 RU of a goat anti-human IgG, F(ab′)2 fragment specific polyclonal antibody (Jackson ImmunoResearch, #109-005-006) in vertical orientation. Subsequently, human, cynomolgus and murine BCMA as well as an unrelated Fc knobs-into-holes construct were injected as analytes in horizontal orientation. Clones that exhibited significant binding responses to BCMA and did not bind the Fc-portion of the antigens, were bacterially expressed in a 0.5 liter culture volume, affinity purified and kinetically characterized by SPR-analysis using a one-shot-kinetics protocol on BioRad's ProteOn XPR36 biosensor.

Example 41 BCMA Binding Assays: Surface Plasmon Resonance

Assessment of binding of anti-BCMA antibodies to recombinant BCMA by surface plasmon resonance (SPR) as follow. All SPR experiments were performed on a Biacore T200 at 25° C. with HBS-EP as running buffer (0.01 M HEPES pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Surfactant P20, Biacore, Freiburg/Germany). The avidity of the interaction between anti-BCMA antibodies and recombinant BCMA Fc(kih) (human, cynomolgus and murine) was determined (Tables 38-40). Biotinylated recombinant human, cynomolgus and murine BCMA Fc(kih) were directly coupled on a SA chip following instructions (Biacore, Freiburg/Germany). The immobilization level ranged from 80 to 120 RU. The anti-BCMA antibodies were passed at a 4-fold concentration range (1.95 to 500 nM) with a flow of 30 μL/minutes through the flow cells over 120 seconds. The dissociation was monitored for 180 seconds. Bulk refractive index differences were corrected for by subtracting the response obtained on the reference flow cell. Here, the anti-BCMA antibodies were flown over an empty surface previously activated and deactivated as described in the standard amine coupling kit. Apparent kinetic constants were derived using the Biacore T200 Evaluation Software (v 1.0, Biacore AB, Uppsala/Sweden), to fit rate equations for 1:1 Langmuir binding by numerical integration, despite the bivalency of the interaction for comparison purposes.

TABLE 38 Avidity values for comparison purposes only (Experiment 1) Kon Analyte Ligand [1/Ms] Koff [1/s] KD [nM] pSCHLI333 huBCMA Fc(kih) 4.55E+06 1.13E−02 2.5 anti-BCMA IgG pSCHLI333 cyBCMA Fc(kih) 9.39E+06 1.24E−02 1.3 anti-BCMA IgG pSCHLI333 muBCMA 8.26E+06 1.62E−01 19.6 anti-BCMA IgG Fc(kih)

TABLE 39 Avidity values for comparison purposes only (Experiment 2) Kon Analyte Ligand [1/Ms] Koff [1/s] KD [nM] pSCHLI372 huBCMA Fc(kih) 3.13E+05 2.66E−03 8.5 anti-BCMA IgG pSCHLI372 cyBCMA Fc(kih) 7.85E+05 3.50E−03 4.5 anti-BCMA IgG pSCHLI372 muBCMA 1.30E+05 5.53E−01 42.7 anti-BCMA IgG Fc(kih) pSCHLI373 huBCMA Fc(kih) 1.40E+05 3.23E−03 23.0 anti-BCMA IgG pSCHLI373 cyBCMA Fc(kih) 9.36E+04 9.28-04 9.9 anti-BCMA IgG

TABLE 40 Avidity values for comparison purposes only (Experiment 3) Kon Analyte Ligand [1/Ms] Koff [1/s] KD [nM] pSCHLI372 huBCMA Fc(kih) 8.03E+05 2.84E−03 3.6 anti-BCMA IgG pSCHLI372 cyBCMA Fc(kih) 1.63E+06 4.69E−03 2.9 anti-BCMA IgG pSCHLI372 muBCMA 4.69E+05 1.21E−02 25.7 anti-BCMA IgG Fc(kih) pSCHLI373 huBCMA Fc(kih) 9.78E+04 1.18E−03 12.1 anti-BCMA IgG pSCHLI373 cyBCMA Fc(kih) 9.39E+04 7.98E−04 8.5 anti-BCMA IgG

The affinity of the interaction between anti-BCMA antibodies or BCMA-TCB CLC antibodies and recombinant human BCMA Fc(kih) was also determined. Anti-human Fab antibody (GE Healthcare) was directly coupled on a CM5 chip at pH 5.0 using the standard amine coupling kit (Biacore, Freiburg/Germany). The immobilization level was about 6500 RU. Anti-BCMA antibody or BCMA-TCB CLC antibody was captured for 90 seconds at 25 nM. Recombinant human BCMA Fc(kih) was passed at a 4-fold concentration range (1.95 to 500 nM) with a flow of 30 μL/minutes through the flow cells over 120 or 180 seconds. The dissociation was monitored for 120 or 400 seconds. Bulk refractive index differences were corrected for by subtracting the response obtained on reference flow cell. Here, recombinant BCMA was flown over a surface with immobilized anti-human Fab antibody but on which HBS-EP has been injected rather than anti-BCMA antibody or BCMA-TCB CLC antibody. Kinetic constants were derived using the Biacore T200 Evaluation Software (v 1.0, Biacore AB, Uppsala/Sweden), to fit rate equations for 1:1 Langmuir binding by numerical integration (Table 41).

TABLE 41 Affinity constants determined by fitting rate equations for 1:1 Langmuir binding Kon Analyte Ligand [1/Ms] Koff [1/s] KD [nM] pSCHLI372 anti-BCMA IgG huBCMA Fc(kih) 4.59E+04 5.23E−03 114 pSCHLI372 anti-BCMA IgG cyBCMA Fc(kih) 2.68E+04 7.52E−03 281 pSCHLI372 anti-BCMA IgG muBCMA 9.92E+04 1.30E−01 1310 Fc(kih) pSCHLI373 anti-BCMA IgG huBCMA Fc(kih) 4.22E+04 5.70E−03 135 pSCHLI373 anti-BCMA IgG cyBCMA Fc(kih) 2.10E+04 1.12E−02 535 pSCHLI373 anti-BCMA IgG muBCMA 1.27E+05 9.19E−02 724 Fc(kih) pSCHLI333 BCMAxCD3 TCB huBCMA Fc(kih) 9.10E+03 1.49E−03 164 pSCHLI333 BCMAxCD3 TCB cyBCMA Fc(kih) 1.44E+04 2.63E−03 183 pSCHLI333 BCMAxCD3 TCB muBCMA 4.83E+02 3.47E−03 7190 Fc(kih) pSCHLI372 BCMAxCD3 TCB huBCMA Fc(kih) 5.59E+04 3.52E−03 63 pSCHLI372 BCMAxCD3 TCB cyBCMA Fc(kih) 4.58E+04 7.57E−03 165 pSCHLI372 BCMAxCD3 TCB muBCMA 1.42E+04 4.87E−03 344 Fc(kih)

Example 42 Generation of Bivalent Anti-BCMA IgG Antibody with the Common Light Chain

To verify the hypothesis that the use of the common light chain could be applied to any BCMA antibodies, 83A10-CLC bivalent BCMA antibody was generated by substituting the native light chain of the bivalent BCMA antibody 83A10, previously described in WO/2014/122143, with the common light chain (CD3 LC (CLC)). The anti-BCMA antibody comprising two heavy chains of 83A10 and two common light chains were produced using the general techniques for generation of bivalent IgG antibodies as described in the Material and general methods section. Affinity of 83A10-CLC IgG antibody to human BCMA was measured by SPR with methods similar to the ones described in Example 41. Table 42 depicts the binding of recombinant human BCMA Fc(kih) to 83A10-CLC BCMA IgG antibody.

TABLE 42 Affinity constants determined by fitting rate equations for 1:1 Langmuir binding:Binding of recombinant BCMA Fc(kih) to 83A10-CLC anti-BCMA antibody Kon Analyte Ligand [1/Ms] Koff [1/s] KD [nM] 83A10 CLC huBCMA Fc(kih) 8.78E+04 2.13E−3 24.3 anti-BCMA IgG

Example 43 Specificity Test of Anti-BCMA IgG Antibodies to huTACI-R and huBAFF-R

As members of the TNF-TNF-R superfamily, TACI and BAFF receptors are related to BCMA receptor with respectively 22% and 18.5% homology in the extracellular domain. Therefore, surface plasmon resonance (SPR) binding experiments were performed to examine the specificity of anti-BCMA IgG antibodies. All SPR experiments were performed on a Biacore T200 (GE Healthcare) at 25° C. with HBS-EP as running buffer (0.01 M HEPES pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Surfactant P20). Fc fused huBCMA, huBAFF-R and huTACI-R were chemically immobilized with a high immobilization level (˜5000 RU) on different flow channels of a Biacore CM5 sensor chip at pH 5.0 using the standard amine coupling kit (GE Healthcare). Initially high concentrated solutions (3 μM, dissolved in HBS-EP) of anti-BCMA IgG pSCHLI372 as well as a-huTACI-R IgG or a-huBAFF-R IgG as positive controls were injected (association time: 80 s, dissociation time: 600 s, flow: 30 μl/min) to check if binding occurs. A positive binding event of the a-huTACI-R IgG to huTACI-R as well as for a-huBAFF-R IgG to huBAFF-R and anti-BCMA IgG antibodies to huBCMA indicated that all receptors were still recognized after immobilization. For anti-BCMA IgG antibodies binding with fast kinetic rate constants to huBAFF-R and/or huTACI-R, a careful examination of kinetic parameter with low immobilization levels (300 RU) was performed on a new CM5 sensor chip. Anti-BCMA IgG antibody dilutions at concentrations of 3000-93.75 nM (2-fold dilution in HBS-EP) are injected (association time: 80 s, dissociation time: 300 s, flow: 30 μl/min), and sample(s) were tested in duplicate. Regeneration was also performed when applicable i.e no fast and complete dissociation. Kinetic evaluation of the interaction between anti-BCMA IgG antibodies and huBAFF-R or huTACI-R was performed by global fitting of the data to a 1:1 interaction model that includes a term for mass transport (Biacore T200 evaluation Version 1.0). A steady state analysis was also performed. As depicted in FIG. 37, curve 1 corresponds to the signal on reference channel, curve 2 to the channel where the binding occurs (binding channel) and the curve 2−1 is the subtracted signal (binding channel—reference channel), meaning that this is the signal due to the binding event. As shown in FIG. 37, the SPR binding assay clearly demonstrated that pSCHLI372 IgG did not bind to human TACI receptor. As positive control for binding, another BCMA IgG known to slightly bind to TACI receptor but with very fast on- and off-rate was used (data not shown).

Example 44 Production and Purification of BCMA-TCB CLC Fc-containing (2+1) Antibodies

For the production of the bispecific antibodies, bispecific antibodies were expressed by transient polymer-based cotransfection of the respective mammalian expression vectors in HEK293-EBNA cells, which were cultivated in suspension. One day prior to transfection the HEK293-EBNA cells were seeded at 1.5 Mio viable cells/mL in Ex-Cell medium, supplemented with 6 mM of L-Glutamine. For every mL of final production volume 2.0 Mio viable cells were centrifuged (5 minutes at 210×g). The supernatant was aspirated and the cells resuspended in 100 μL of CD CHO medium. The DNA for every mL of final production volume was prepared by mixing 1 μg of DNA (Ratio Bispecific Antibody Production: heavy chain: modified heavy chain: light chain: modified light chain=1:1:2:1; Ratio standard antibody: heavy chain: light chain=1:1) in 100 μL of CD CHO medium. After addition of 0.27 μL of PEI solution (1 mg/mL) the mixture was vortexed for 15 seconds and left at room temperature for 10 minutes. After 10 minutes, the resuspended cells and DNA/PEI mixture were put together and then transferred into an appropriate container which was placed in a shaking device (37° C., 5% CO2). After a 3 hours incubation time 800 μL of Ex-Cell Medium, supplemented with 6 mM L-Glutamine, 1.25 mM valproic acid and 12.5% Pepsoy (50 g/L), was added for every mL of final Production volume. After 24 hours, 70 μL of feed solution was added for every mL of final production volume. After 7 days or when the cell viability was equal or lower than 70%, the cells were separated from the supernatant by centrifugation and sterile filtration. The antibodies were purified by an affinity step and one polishing step size exclusion chromatography. For the affinity step the supernatant was loaded on a protein A column (HiTrap Protein A FF, 5 mL, GE Healthcare) equilibrated with 6 CV 20 mM sodium phosphate, 20 mM sodium citrate, pH 7.5. After a washing step with the same buffer the antibody was eluted from the column by step elution with 20 mM sodium phosphate, 100 mM sodium chloride, 100 mM Glycine, pH 3.0. The fractions with the desired antibody were immediately neutralized by 0.5 M Sodium Phosphate, pH 8.0 (1:10), pooled and concentrated by centrifugation. The concentrate was sterile filtered and processed further by size exclusion chromatography.

For the size exclusion step the concentrated protein was injected in a XK16/60 HiLoad Superdex 200 column (GE Healthcare), and 20 mM Histidine, 140 mM Sodium Chloride, pH 6.0 with or without Tween20 as formulation buffer. The fractions containing the monomers were pooled, concentrated by centrifugation and sterile filtered into a sterile vial.

Determination of the antibody concentration was done by measurement of the absorbance at 280 nm, using the theoretical value of the absorbance of a 0.1% solution of the antibody. This value was based on the amino acid sequence and calculated by GPMAW software (Lighthouse data). Purity and monomer content of the final protein preparation was determined by CE-SDS (Caliper LabChip GXII system (Caliper Life Sciences)) resp. HPLC (TSKgel G3000 SW XL analytical size exclusion column (Tosoh)) in a 25 mM potassium phosphate, 125 mM Sodium chloride, 200 mM L-arginine monohydrochloride, 0.02% (w/v) Sodium azide, pH 6.7 buffer. FIGS. 38A-C depicts the CE-SDS graphs (non-reduced (top panels) and reduced (bottom panels)) of the final protein preparations after Protein A (PA) affinity chromatography and size exclusion chromatographic (SEC) purification steps (FIG. 38A) pSCHLI333-TCB CLC, (FIG. 38B) pSCHLI372-TCB CLC, (FIG. 38C) pSCHLI373-TCB CLC. PA affinity chromatography and SEC purification steps applied to pSCHLI333-TCB CLC antibody resulted in a purity of 89.2% and 100% of monomer content (FIG. 38A), a purity of 88.5% and 100% of monomer content for pSCHLI372-TCB CLC antibody (FIG. 38B), and a purity of 91.8% and 100% of monomer content for pSCHLI372-TCB CLC antibody (FIG. 38C). Table 43 summarizes the properties of pSCHLI333-TCB CLC, pSCHLI372-TCB CLC, and pSCHLI373-TCB CLC antibodies following PA affinity chromatography and SEC purification steps. In all BCMA-TCB CLC antibodies, >88% purity and 100% monomer content were consistently reached. The overall results clearly demonstrate that advantages in production/purification features could be achieved with using a common light chain (CLC) to TCB antibodies and that only two purification steps (i.e PA affinity chromatography and SEC) were required to achieve already high quality protein preparations with very good developability properties.

TABLE 43 Production/purification profile of BCMA-TCB CLC antibodies following protein A affinity chromatography and size exclusion chromatography purification steps pSCHLI333-TCB pSCHLI372-TCB pSCHLI373-TCB CLC CLC CLC Purity (%) 89.2 88.5 91.8 Yield (mg/L) 4.2 4.1 2.73 Titer (mg/L) 26.9 21.2 37.1 Monomer 100 100 100 (%)

Example 44 Binding of BCMA-TCB CLC Antibodies to BCMA-Positive Multiple Myeloma Cell Lines

BCMA-TCB CLC antibodies (pSCHLI333, pSCHLI372, pSCHLI373) were analyzed by flow cytometry for binding to human BCMA on BCMAhi-expressing H929 cells. MKN45 (human gastric adenocarcinoma cell line that does not express BCMA) was used as negative control.

Briefly, cultured cells are harvested, counted and cell viability was evaluated using ViCell. Viable cells are then adjusted to 2×106 cells per ml in BSA-containing FACS Stain Buffer (BD Biosciences). 100 μl of this cell suspension were further aliquoted per well into a round-bottom 96-well plate and incubated with 30 μl of the BCMA-TCB CLC antibodies or corresponding TCB control for 30 min at 4° c. All BCMA-TCB CLC antibodies (and TCB controls) were titrated and analyzed in final concentration range between 0.12-500 nM. Cells were then centrifuged (5 min, 350×g), washed with 120W/well FACS Stain Buffer (BD Biosciences), resuspended and incubated for an additional 30 min at 4° C. with fluorochrome-conjugated PE-conjugated AffiniPure F(ab′)2 Fragment goat anti-human IgG Fc Fragment Specific (Jackson Immuno Research Lab; 109-116-170). Cells were then washed twice with Stain Buffer (BD Biosciences), fixed using 100 ul BD Fixation buffer per well (#BD Biosciences, 554655) at 4° C. for 20 min, resuspended in 120 μl FACS buffer and analyzed using BD FACS CantoII. As depicted in FIGS. 39A-B, the mean fluorescence intensity of BCMA-TCB CLC antibodies were plotted in function of antibody concentrations; (FIG. 39A) pSCHLI372-TCB CLC and pSCHLI373-TCB CLC on H929 cells; (FIG. 39B) pSCHLI372-TCB CLC and pSCHLI373-TCB CLC on MKN45 cells. BCMA-TCB CLC antibodies (pSCHLI333, pSCHLI372, pSCHLI373) antibodies did not bind to BCMA-negative and CD3-negative MKN45 cells at concentrations below 100 nM. When applicable, EC50 were calculated using Prism GraphPad (LaJolla, Calif., USA) and EC50 values denoting the antibody concentration required to reach 50% of the maximal binding for the binding of anti-BCMA/anti-CD3 TCB antibodies to H929 cells are summarized in Table 44.

TABLE 44 EC50 values for binding of BCMA-TCB CLC antibodies to H929 cells Anti-BCMA/anti-CD3 TCB molecules EC50 (nM) EC50 (μg/ml) pSCHLI372-TCB CLC 344.8 65.9 pSCHLI373-TCB CLC no EC50 value no EC50 value

Example 45 Binding of BCMA-TCB CLC Antibodies to CD3-Positive Jurkat T Cell Line (Flow Cytometry)

BCMA-TCB CLC antibodies (pSCHLI333, pSCHLI372, pSCHLI373) were also analyzed by flow cytometry for their binding properties to human CD3 expressed on human leukemic T cells Jurkat (ATCC TIB-152). Jurkat T cells were cultured in RPMI supplemented with 10% heat-inactivated FCS. Briefly, cultured cells were harvested, counted and cell viability was evaluated using ViCell. Viable cells were then adjusted to 2×106 cells per ml in FACS Stain Buffer (BD Biosciences) containing 0.1% BSA. 100 μl of this cell suspension were further aliquoted per well into a round-bottom 96-well plate. 30 μl of BCMA-TCB CLC antibodies or corresponding TCB control were added to the cell-containing wells to obtain final concentrations of 0.12 nM to 500 nM. BCMA-TCB CLC antibodies and control IgG were used at the same molarity. After incubation for 30 min at 4° C., cells were centrifuged (5 min, 350×g), washed twice with 150 μl/well BSA-containing FACS Stain Buffer (BD Biosciences), then cells were fixed using 100 ul BD Fixation buffer per well (#BD Biosciences, 554655) at 4° C. for 20 min, resuspended in 120 μl FACS buffer and analyzed using BD FACS CantoII. Binding of BCMA-TCB CLC antibodies to T cells were evaluated and the median fluorescence intensity was determined gated on CD3-expressing Jurkat T cells and plotted in histograms or dot plots. FIGS. 40A-B show the median fluorescence intensity for BCMA-TCB CLC antibodies (pSCHLI333, pSCHLI372, pSCHLI373) binding to Jurkat T cells (FIG. 40A) or MKN45 cells (FIG. 40B) and plotted in function of antibody concentration. EC50 values and maximal binding of anti-BCMA/anti-CD3 TCB antibodies to CD3-positive Jurkat T cells were not reached. Isotype control antibody did not bind to Jurkat T cells and BCMA-TCB CLC antibodies (pSCHLI333, pSCHLI372, pSCHLI373) antibodies did not bind to BCMA-negative and CD3-negative MKN45 cells at concentrations below 100 nM.

Example 46 Activation of Human T Cells Upon Binding of BCMA-TCB CLC Antibodies to CD3-Positive T Cells and BCMA-Positive Multiple Myeloma Cell Lines

BCMA-TCB CLC antibodies (pSCHLI372, pSCHLI373) were analyzed by flow cytometry for their ability to induce T cell activation by evaluating the surface expression of the early activation marker CD69, or the late activation marker CD25 on CD4+ and CD8+ T cells in the presence or absence of human BCMA-expressing MM cells. Briefly, BCMA-positive H929 cells were harvested with Cell Dissociation buffer, counted and checked for viability. Cells were adjusted to 0.3×106 (viable) cells per ml in modified RPMI-1640 medium, 100 μl of this cell suspension were pipetted per well into a round-bottom 96-well plate (as indicated). 50 μl of the (diluted) BCMA-TCB CLC antibodies were added to the cell-containing wells to obtain a final concentration of 0.012 pM-100 nM. Human PBMC effector cells were isolated from fresh blood of a healthy donor and adjusted to 6×106 (viable) cells per ml in modified RPMI-1640 medium. 50 μl of this cell suspension was added per well of the assay plate to obtain a final E:T ratio of PBMC to myeloma tumor cells of 10:1. To analyze whether the BCMA-TCB CLC antibodies were able to activate T cells specifically in the presence of target cells expressing human BCMA, wells were included that contained 3 to 10 nM of the respective BCMA-TCB CLC antibodies molecules, as well as PBMCs, but no target cells. After 48 h incubation at 37° C., 5% CO2, cells were centrifuged (5 min, 350×g) and washed twice with 150 μl/well PBS containing 0.1% BSA. Surface staining for CD4 (mouse IgG1, K; clone RPA-T4), CD8 (mouse IgG1, K; clone HIT8a; BD #555635), CD69 (mouse IgG2; clone L78; BD #340560) and CD25 (mouse IgG1, K; clone M-A251; BD #555434) was performed at 4° C. for 30 min, according to the supplier's suggestions. Cells were washed twice with 150 μl/well PBS containing 0.1% BSA and fixed for 15 min at 4° C., using 100 μl/well fixation buffer (BD #554655). After centrifugation, the samples were resuspended in 200 μl/well PBS with 0.1% BSA and analyzed using a FACS CantoII machine (Software FACS Diva). FIG. 41 depicts the expression level of the early activation marker CD69 and the late activation marker CD25 on CD4+ and CD8+ T cells after 48 hours of incubation (representative results from two independent experiments). pSCHLI372-TCB CLC and pSCHLI373-TCB CLC antibodies induced an up-regulation of CD69 and CD25 activation markers in a concentration-dependent and specific manner in the presence of BCMA-positive target cells. No activation of CD4+ and CD8+ T cells was observed when human PBMCs were treated with DP47-TCB control antibody, suggesting that despite binding to CD3 on the T cells T-cell activation does not occur when the TCB antibody does not bind to BCMA-positive target cells.

Example 47 Redirected T-Cell Cytotoxicity of BCMAhi-Expressing H929 Myeloma Cells Induced by Anti-BCMA/Anti-CD3 T Cell Bispecific Antibodies (Colorimetric LDH Release Assay)

BCMA-TCB CLC antibodies (pSCHLI372, pSCHLI373) were also analyzed for their potential to induce T cell-mediated apoptosis in BCMA-high expressing MM cells upon crosslinking of the construct via binding of the antigen binding moieties to BCMA on cells. Briefly, human BCMA-expressing H929 multiple myeloma target cells were harvested with Cell Dissociation Buffer, washed and resuspended in RPMI supplemented with 10% fetal bovine serum (Invitrogen). Approximately, 30,000 cells per well were plated in a round-bottom 96-well plate and the respective dilution of the construct was added for a desired final concentration (in triplicates); final concentrations ranging from 0.12 pM to 100 nM. For an appropriate comparison, all TCB constructs and controls were adjusted to the same molarity. Human total T cells (effector) were added into the wells to obtain a final E:T ratio of 5:1. When human PBMC were used as effector cells, a final E:T ratio of 10:1 was used. Negative control groups were represented by effector or target cells only. As a positive control for the activation of human pan T cells, 1 μg/ml PHA-M (Sigma #L8902) was used. For normalization, maximal lysis of the H929 MM target cells (=100%) was determined by incubation of the target cells with a final concentration of 1% Triton X-100, inducing cell death. Minimal lysis (=0%) was represented by target cells co-incubated with effector cells only, i.e. without any T cell bispecific antibody. After 20-24 h or 48 h incubation at 37° C., 5% CO2, LDH release from the apoptotic/necrotic MM target cells into the supernatant was then measured with the LDH detection kit (Roche Applied Science), following the manufacturer's instructions. The percentage of LDH release was plotted against the concentrations of BCMA-TCB CLC antibodies in concentration-response curves. The EC50 values were measured using Prism software (GraphPad) and determined as the TCB antibody concentration that results in 50% of maximum LDH release. As shown in FIGS. 42A-B, BCMA-TCB CLC antibodies, pSCHLI372-TCB CLC (FIG. 42A, B) and pSCHLI373-TCB CLC (FIG. 42B) induced a concentration-dependent killing of BCMA-positive H929 myeloma cells as measured by LDH release. The killing of H929 cells was specific since DP47-TCB control antibody which does not bind to BCMA-positive target cells did not induce LDH release, even at the highest concentration tested of 100 nM. Table 45 summarizes the EC50 values for redirected T-cell killing of BCMA-positive H92 cells induced by BCMA-TCB CLC antibodies.

TABLE 45 EC50 values for redirected T-cell killing of H929 cells induced by BCMA-TCB CLC antibodies Anti-BCMA/anti-CD3 TCB molecules EC50 (pM) EC50 (μg/ml) pSCHLI333-TCB CLC 980 0.19 pSCHLI372-TCB CLC (Experiment 1) 450 0.08 pSCHLI373-TCB CLC 1590 0.31 pSCHLI372-TCB CLC (Experiment 2) 900 0.17

Example 48 Redirected T-Cell Cytotoxicity of BCMAmed/lo-Expressing U266 Myeloma Cells Induced by BCMA-TCB CLC Antibodies (LDH Release Assay)

BCMA-TCB CLC antibodies (pSCHLI372, pSCHLI373) were analyzed for their ability to induce T cell-mediated apoptosis in BCMAmed/lo-expressing MM cells upon crosslinking of the construct via binding of the antigen binding moieties to BCMA on cells. Briefly, human BCMAmed/lo-expressing U266 multiple myeloma target cells were harvested with Cell Dissociation Buffer, washed and resuspended in RPMI supplemented with 10% fetal bovine serum (Invitrogen). Approximately, 30,000 cells per well are plated in a round-bottom 96-well plate and the respective dilution of the construct was added for a desired final concentration (in triplicates); final concentrations ranging from 0.12 pM to 100 nM. For an appropriate comparison, all TCB constructs and controls were adjusted to the same molarity. Human total T cells (effector) were added into the wells to obtain a final E:T ratio of 5:1. When human PBMC were used as effector cells, a final E:T ratio of 10:1 was used. Negative control groups were represented by effector or target cells only. As a positive control for the activation of human T cells, 1 μg/ml PHA-M (Sigma #L8902) was used. For normalization, maximal lysis of the MM target cells (=100%) was determined by incubation of the target cells with a final concentration of 1% Triton X-100, inducing cell death. Minimal lysis (=0%) was represented by target cells co-incubated with effector cells only, i.e. without any T cell bispecific antibody. After 20-24 h incubation at 37° C., 5% CO2, LDH release from the apoptotic/necrotic MM target cells into the supernatant was then measured with the LDH detection kit (Roche Applied Science), following the manufacturer's instructions. The percentage of LDH release was plotted against the concentrations of BCMA-TCB CLC antibodies in concentration-response curves. The EC50 values were measured using Prism software (GraphPad) and determined as the TCB antibody concentration that results in 50% of maximum LDH release. Table 46 summarizes the EC50 values for redirected T-cell killing of BCMA-positive U266 cells induced by BCMA-TCB CLC antibodies.

TABLE 46 EC50 values for redirected T-cell killing of U266 cells induced by BCMA-TCB CLC antibodies Anti-BCMA/anti-CD3 TCB molecules EC50 (pM) EC50 (μg/ml) pSCHLI372-TCB CLC 5700 1.1

Amino Acid Sequences of Exemplary Embodiments

1) FolR binders useful in common light chain format, variable heavy chain Description Sequence Seq ID No 16A3 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 1 WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA VYYCARNYYAGVTPFDYWGQGTLVTVSS 18D3 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 2 WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA VYYCARNYYTGGSSAFDYWGQGTLVTVS 15H7 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 3 WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA VYYCARNYYLFSTSFDYWGQGTLVTVSS 15B6 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 4 WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA VYYCARNYYIGIVPFDYWGQGTLVTVSS 21D1 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 5 WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA VYYCARNYYVGVSPFDYWGQGTLVTVSS 16F12 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 6 WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA VYYCARNFTVLRVPFDYWGQGTLVTVSS 15A1 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 7 WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA VYYCARNYYIGVVTFDYWGQGTLVTVSS 15A1_CDR1 SYYMH 8 15A1_CDR2 IINPSGGSTSYAQKFQG 9 15A1_CDR3 NYYIGVVTFDY 10 19E5 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 11 WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA VYYCARGEWRRYTSFDYWGQGTLVTVSS 19E5_CDR1 SYYMH 8 19E5_CDR2 IINPSGGSTSYAQKFQG 9 19E5_CDR3 GEWRRYTSFDY 12 19A4 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 13 WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA VYYCARGGWIRWEHFDYWGQGTLVTVSS 19A4_CDR1 SYYMH 8 19A4_CDR2 IINPSGGSTSYAQKFQG 9 19A4_CDR3 GGWIRWEHFDY 14 16D5 EVQLVESGGGLVKPGGSLRLSCAASGFTFSNAWMSWVRQAPGKGLE 15 WVGRIKSKTDGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTED TAVYYCTTPWEWSWYDYWGQGTLVTVSS 16D5_CDR1 NAWMS 16 16D5_CDR2 RIKSKTDGGTTDYAAPVKG 17 16D5_CDR3 PWEWSWYDY 18 15E12 EVQLVESGGGLVKPGGSLRLSCAASGFTFSNAWMSWVRQAPGKGLE 19 WVGRIKSKTDGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTED TAVYYCTTPWEWSYFDYWGQGTLVTVSS 15E12_CDR1 NAWMS 16 15E12_CDR2 RIKSKTDGGTTDYAAPVKG 17 15E12_CDR3 PWEWSYFDY 20 21A5 EVQLVESGGGLVKPGGSLRLSCAASGFTFSNAWMSWVRQAPGKGLE 21 WVGRIKSKTDGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTED TAVYYCTTPWEWAWFDYWGQGTLVTVSS 21A5_CDR1 NAWMS 16 21A5_CDR2 RIKSKTDGGTTDYAAPVKG 17 21A5_CDR3 PWEWAWFDY 22 21G8 EVQLVESGGGLVKPGGSLRLSCAASGFTFSNAWMSWVRQAPGKGLE 23 WVGRIKSKTDGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTED TAVYYCTTPWEWAYFDYWGQGTLVTVSS 21G8_CDR1 NAWMS 16 21G8_CDR2 RIKSKTDGGTTDYAAPVKG 17 21G8_CDR3 PWEWAYFDY 24 19H3 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 25 WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA VYYCARTGWSRWGYMDYWGQGTLVTVSS 19H3_CDR1 SYYMH 8 19H3_CDR2 IINPSGGSTSYAQKFQG 9 19H3_CDR3 TGWSRWGYMDY 26 20G6 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 27 WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA VYYCARGEWIRYYHFDYWGQGTLVTVSS 20G6_CDR1 SYYMH 8 20G6_CDR2 IINPSGGSTSYAQKFQG 9 20G6_CDR3 GEWIRYYHFDY 28 20H7 QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLE 29 WMGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTA VYYCARVGWYRWGYMDYWGQGTLVTVSS 20H7_CDR1 SYYMH 8 20H7_CDR2 IINPSGGSTSYAQKFQG 9 20H7_CDR3 VGWYRWGYMDY 30

2) CD3 binder common light chain (CLC) Description Sequence Seq ID No common QAVVTQEPSLTVSPGGTVTLTCGSSTGAVTTSNYANWVQEKP 31 CD3 light GQAFRGLIGGTNKRAPGTPARFSGSLLGGKAALTLSGAQPED chain (VL) EAEYYCALWYSNLWVFGGGTKLTVL common GSSTGAVTTSNYAN 32 CD3 light chain_CDR1 common GTNKRAP 33 CD3 light chain_CDR2 common ALWYSNLWV 34 CD3 light chain_CDR3 common QAVVTQEPSLTVSPGGTVTLTCGSSTGAVTTSNYANWVQEKP 35 CD3 light GQAFRGLIGGTNKRAPGTPARFSGSLLGGKAALTLSGAQPED chain EAEYYCALWYSNLWVFGGGTKLTVLGQPKAAPSVTLFPPSSE (VLCL) ELQANKATLVCLISDFYPGAVTVAWKADSSPVKAGVETTTPS KQSNNKYAASSYLSLTPEQWKSHRSYSCQVTHEGSTVEKTVA PTECS

3) CD3 binder, heavy chain Seq ID Description Sequence No CD3 EVQLLESGGGLVQPGGSLRLSCAASGFTFSTYAMNWVRQAPGKG 36 variable LEWVSRIRSKYNNYATYYADSVKGRFTISRDDSKNTLYLQMNSL heavy chain RAEDTAVYYCVRHGNFGNSYVSWFAYWGQGTLVTVSS (VH) CD3 heavy TYAMN 37 chain (VH)_CDR1 CD3 heavy RIRSKYNNYATYYADSVKG 38 chain (VH)_CDR2 CD3 heavy HGNFGNSYVSWFAY 39 chain (VH)_CDR3 CD3 full EVQLLESGGGLVQPGGSLRLSCAASGFTFSTYAMNWVRQAPGKG 40 heavy chain LEWVSRIRSKYNNYATYYADSVKGRFTISRDDSKNTLYLQMNSL (VHCH1) RAEDTAVYYCVRHGNFGNSYVSWFAYWGQGTLVTVSSASTKGPS VFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVH TFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVD KKVEPKSC CD3 ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSG 84 constant ALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHK heavy chain PSNTKVDKKVEPKSC CH1

4) MUC1 binders useful in common light chain format, variable heavy chain SEQ ID Amino Acid NOS Description Nucleotide Sequence MUC1 Antigens 57 and MUC1 SEA- ATGAGCTTTTTTTTCCTGAGCTTTCATATTAGC MSFFFLSFHISNLQ 58 avi-His AACCTGCAGTTTAATAGCAGCCTGGAAGATCCG FNSSLEDPSTDYYQ AGCACCGATTATTATCAAGAACTGCAGCGTGAT ELQRDISEMFLQIY ATCAGCGAAATGTTTCTGCAGATCTATAAACAG KQGGFLGLSNIKFR GGTGGTTTTCTGGGTCTGAGCAACATCAAATTT PGGGGGSVVVQLTL CGTCCGGGTGGTGGCGGTGGTTCAGTTGTTGTG AFREGTINVHDVET CAGCTGACCCTGGCATTTCGTGAAGGCACCATT QFNQYKTEAASRYN AATGTTCATGATGTGGAAACCCAGTTTAACCAG LTISDVSVSDVPFP TATAAAACCGAAGCAGCAAGCCGTTATAATCTG FSAQSVDGLNDIFE ACCATTAGTGATGTTAGCGTTTCCGATGTTCCG AQKIEWHELEHHHH TTTCCGTTTAGCGCACAGAGTGTCGACGGTCTG HH AATGATATTTTTGAAGCCCAGAAAATCGAATGG CATGAACTCGAGCACCACCACCACCACCAC MUC1 Binders 59 and 58D6 VH GAGGTGCAATTGTTGGAGTCTGGGGGAGGCTTG EVQLLESGGGLVQP 60 GTACAGCCTGGGGGGTCCCTGAGACTCTCCTGT GGSLRLSCAASGFT GCAGCCTCCGGATTCACCTTTAGCAGTTATGCC FSSYAMSWVRQAPG ATGAGCTGGGTCCGCCAGGCTCCAGGGAAGGGG KGLEWVSAISGSGG CTGGAGTGGGTCTCAGCTATTAGTGGTAGTGGT STYYADSVKGRFTI GGTAGCACATACTACGCAGACTCCGTGAAGGGC SRDNSKNTLYLQMN CGGTTCACCATCTCCAGAGACAATTCCAAGAAC SLRAEDTAVYYCAK ACGCTGTATCTGCAGATGAACAGCCTGAGAGCC VEWSTLLYFDYWGQ GAGGACACGGCCGTATATTACTGTGCGAAAGTT GTLVTVSS GAATGGTCTACTCTGCTGTACTTCGACTACTGG GGCCAAGGAACCCTGGTCACCGTCTCGAGT 61 and 106D2 VH GAGGTGCAATTGTTGGAGTCTGGGGGAGGCTTG EVQLLESGGGLVQP 62 GTACAGCCTGGGGGGTCCCTGAGACTCTCCTGT GGSLRLSCAASGFT GCAGCCTCCGGATTCACCTTTCGGGTTTATGCA FRVYAMSWVRQAPG ATGAGCTGGGTCCGCCAGGCTCCAGGGAAGGGG KGLEWVSGISETGS CTGGAGTGGGTCTCAGGTATTTCTGAAACTGGT YTYYADSVKGRFTI TCTTACACATACTACGCAGACTCCGTGAAGGGC SRDNSKNTLYLQMN CGGTTCACCATCTCCAGAGACAATTCCAAGAAC SLRAEDTAVYYCAR ACGCTGTATCTGCAGATGAACAGCCTGAGAGCC YPYGFDYWGQGTLV GAGGACACGGCCGTATATTACTGTGCGCGTTAC TVSS CCGTACGGTTTCGACTACTGGGGCCAAGGAACC CTGGTCACCGTCTCGAGT 63 and 110A5 VH GAGGTGCAATTGTTGGAGTCTGGGGGAGGCTTG EVQLLESGGGLVQP 64 GTACAGCCTGGGGGGTCCCTGAGACTCTCCTGT GGSLRLSCAASGFT GCAGCCTCCGGATTCACCTTTAGCAGTTATGCC FSSYAMSWVRQAPG ATGAGCTGGGTCCGCCAGGCTCCAGGGAAGGGG KGLEWVSAISGSGG CTGGAGTGGGTCTCAGCTATTAGTGGTAGTGGT STYYADSVKGRFTI GGTAGCACATACTACGCAGACTCCGTGAAGGGC SRDNSKNTLYLQMN CGGTTCACCATCTCCAGAGACAATTCCAAGAAC SLRAEDTAVYYCAK ACGCTGTATCTGCAGATGAACAGCCTGAGAGCC YSYRYVLAFDYWGQ GAGGACACGGCCGTATATTACTGTGCGAAATAC GTLVTVSS TCTTACCGTTACGTTCTGGCTTTCGACTACTGG GGCCAAGGAACCCTGGTCACCGTCTCGAGT 65 and commom CAGGCCGTCGTGACCCAGGAACCCAGCCTGACA QAVVTQEPSLTVSP 66 light chain GTGTCTCCTGGCGGCACCGTGACCCTGACATGT GGTVTLTCGSSTGA VL GGCAGTTCTACAGGCGCCGTGACCACCAGCAAC VTTSNYANWVQEKP TACGCCAACTGGGTGCAGGAAAAGCCCGGCCAG GQAFRGLIGGTNKR GCCTTCAGAGGACTGATCGGCGGCACCAACAAG APGTPARESGSLLG AGAGCCCCTGGCACCCCTGCCAGATTCAGCGGA GKAALTLSGAQPED TCTCTGCTGGGAGGAAAGGCCGCCCTGACACTG EAEYYCALWYSNLW TCTGGCGCCCAGCCAGAAGATGAGGCCGAGTAC VFGGGTKLTVL TACTGCGCCCTGTGGTACAGCAACCTGTGGGTG TTCGGCGGAGGCACCAAGCTGACAGTCCTA 67 and Common CAGGCCGTCGTGACCCAGGAACCCAGCCTGACA QAVVTQEPSLTVSP 68 light chain GTGTCTCCTGGCGGCACCGTGACCCTGACATGT GGTVTLTCGSSTGA GGCAGTTCTACAGGCGCCGTGACCACCAGCAAC VTTSNYANWVQEKP TACGCCAACTGGGTGCAGGAAAAGCCCGGCCAG GQAFRGLIGGTNKR GCCTTCAGAGGACTGATCGGCGGCACCAACAAG APGTPARESGSLLG AGAGCCCCTGGCACCCCTGCCAGATTCAGCGGA GKAALTLSGAQPED TCTCTGCTGGGAGGAAAGGCCGCCCTGACACTG EAEYYCALWYSNLW TCTGGCGCCCAGCCAGAAGATGAGGCCGAGTAC VFGGGTKLTVLRTQ TACTGCGCCCTGTGGTACAGCAACCTGTGGGTG PKAAPSVTLFPPSS TTCGGCGGAGGCACCAAGCTGACAGTGCTGCGT EELQANKATLVCLI ACGCAACCCAAGGCTGCCCCCAGCGTGACCCTG SDFYPGAVTVAWKA TTCCCCCCCAGCAGCGAGGAACTGCAGGCCAAC DSSPVKAGVETTTP AAGGCCACCCTGGTCTGCCTGATCAGCGACTTC SKQSNNKYAASSYL TACCCAGGCGCCGTGACCGTGGCCTGGAAGGCC SLTPEQWKSHRSYS GACAGCAGCCCCGTGAAGGCCGGCGTGGAGACC CQVTHEGSTVEKTV ACCACCCCCAGCAAGCAGAGCAACAACAAGTAC APTECS GCCGCCAGCAGCTACCTGAGCCTGACCCCCGAG CAGTGGAAGAGCCACAGGTCCTACAGCTGCCAG GTGACCCACGAGGGCAGCACCGTGGAGAAAACC GTGGCCCCCACCGAGTGCTCC 69 and 58D6 knob GAGGTGCAATTGTTGGAGTCTGGGGGAGGCTTG EVQLLESGGGLVQP 70 heavy chain GTACAGCCTGGGGGGTCCCTGAGACTCTCCTGT GGSLRLSCAASGFT GCAGCCTCCGGATTCACCTTTAGCAGTTATGCC FSSYAMSWVRQAPG ATGAGCTGGGTCCGCCAGGCTCCAGGGAAGGGG KGLEWVSAISGSGG CTGGAGTGGGTCTCAGCTATTAGTGGTAGTGGT STYYADSVKGRFTI GGTAGCACATACTACGCAGACTCCGTGAAGGGC SRDNSKNTLYLQMN CGGTTCACCATCTCCAGAGACAATTCCAAGAAC SLRAEDTAVYYCAK ACGCTGTATCTGCAGATGAACAGCCTGAGAGCC VEWSTLLYFDYWGQ GAGGACACGGCCGTATATTACTGTGCGAAAGTT GTLVTVSSASTKGP GAATGGTCTACTCTGCTGTACTTCGACTACTGG SVFPLAPSSKSTSG GGCCAAGGAACCCTGGTCACCGTCTCGAGTGCT GTAALGCLVKDYFP AGCACAAAGGGCCCTAGCGTGTTCCCTCTGGCC EPVTVSWNSGALTS CCCAGCAGCAAGAGCACAAGCGGCGGAACAGCC GVHTFPAVLQSSGL GCCCTGGGCTGCCTCGTGAAGGACTACTTCCCC YSLSSVVTVPSSSL GAGCCCGTGACAGTGTCTTGGAACAGCGGAGCC GTQTYICNVNHKPS CTGACAAGCGGCGTGCACACTTTCCCTGCCGTG NTKVDKKVEPKSCD CTGCAGAGCAGCGGCCTGTACTCCCTGAGCAGC GGGGSGGGGSEVQL GTGGTCACCGTGCCTAGCAGCAGCCTGGGCACC LESGGGLVQPGGSL CAGACCTACATCTGCAACGTGAACCACAAGCCC RLSCAASGFTFSTY AGCAACACCAAAGTGGACAAGAAGGTGGAGCCC AMNWVRQAPGKGLE AAGAGCTGTGATGGCGGAGGAGGGTCCGGAGGC WVSRIRSKYNNYAT GGAGGATCCGAGGTGCAGCTGCTGGAATCTGGC YYADSVKGRFTISR GGCGGACTGGTGCAGCCTGGCGGATCTCTGAGA DDSKNTLYLQMNSL CTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGC RAEDTAVYYCVRHG ACCTACGCCATGAACTGGGTGCGCCAGGCCCCT NFGNSYVSWFAYWG GGCAAAGGCCTGGAATGGGTGTCCCGGATCAGA QGTLVTVSSASTKG AGCAAGTACAACAACTACGCCACCTACTACGCC PSVFPLAPSSKSTS GACAGCGTGAAGGGCCGGTTCACCATCAGCCGG GGTAALGCLVKDYF GACGACAGCAAGAACACCCTGTACCTGCAGATG PEPVTVSWNSGALT AACAGCCTGCGGGCCGAGGACACCGCCGTGTAC SGVHTFPAVLQSSG TATTGTGTGCGGCACGGCAACTTCGGCAACAGC LYSLSSVVTVPSSS TATGTGTCTTGGTTTGCCTACTGGGGCCAGGGC LGTQTYICNVNHKP ACCCTCGTGACCGTGTCAAGCGCTAGTACCAAG SNTKVDKKVEPKSC GGCCCCAGCGTGTTCCCCCTGGCACCCAGCAGC DKTHTCPPCPAPEA AAGAGCACATCTGGCGGAACAGCCGCTCTGGGC AGGPSVFLFPPKPK TGTCTGGTGAAAGACTACTTCCCCGAGCCCGTG DTLMISRTPEVTCV ACCGTGTCTTGGAACTCTGGCGCCCTGACCAGC VVDVSHEDPEVKFN GGCGTGCACACCTTTCCAGCCGTGCTGCAGAGC WYVDGVEVHNAKTK AGCGGCCTGTACTCCCTGTCCTCCGTGGTCACC PREEQYNSTYRVVS GTGCCCTCTAGCTCCCTGGGAACACAGACATAT VLTVLHQDWLNGKE ATCTGTAATGTCAATCACAAGCCTTCCAACACC YKCKVSNKALGAPI AAAGTCGATAAGAAAGTCGAGCCCAAGAGCTGC EKTISKAKGQPREP GACAAAACTCACACATGCCCACCGTGCCCAGCA QVYTLPPCRDELTK CCTGAAGCTGCAGGGGGACCGTCAGTCTTCCTC NQVSLWCLVKGFYP TTCCCCCCAAAACCCAAGGACACCCTCATGATC SDIAVEWESNGQPE TCCCGGACCCCTGAGGTCACATGCGTGGTGGTG NNYKTTPPVLDSDG GACGTGAGCCACGAAGACCCTGAGGTCAAGTTC SFFLYSKLTVDKSR AACTGGTACGTGGACGGCGTGGAGGTGCATAAT WQQGNVFSCSVMHE GCCAAGACAAAGCCGCGGGAGGAGCAGTACAAC ALHNHYTQKSLSLS AGCACGTACCGTGTGGTCAGCGTCCTCACCGTC PGK CTGCACCAGGACTGGCTGAATGGCAAGGAGTAC AAGTGCAAGGTCTCCAACAAAGCCCTCGGCGCC CCCATCGAGAAAACCATCTCCAAAGCCAAAGGG CAGCCCCGAGAACCACAGGTGTACACCCTGCCC CCATGCCGGGATGAGCTGACCAAGAACCAGGTC AGCCTGTGGTGCCTGGTCAAAGGCTTCTATCCC AGCGACATCGCCGTGGAGTGGGAGAGCAATGGG CAGCCGGAGAACAACTACAAGACCACGCCTCCC GTGCTGGACTCCGACGGCTCCTTCTTCCTCTAC AGCAAGCTCACCGTGGACAAGAGCAGGTGGCAG CAGGGGAACGTCTTCTCATGCTCCGTGATGCAT GAGGCTCTGCACAACCACTACACGCAGAAGAGC CTCTCCCTGTCTCCGGGTAAA 71 and 58D6 hole GAGGTGCAATTGTTGGAGTCTGGGGGAGGCTTG EVQLLESGGGLVQP 72 heavy chain GTACAGCCTGGGGGGTCCCTGAGACTCTCCTGT GGSLRLSCAASGFT GCAGCCTCCGGATTCACCTTTAGCAGTTATGCC FSSYAMSWVRQAPG ATGAGCTGGGTCCGCCAGGCTCCAGGGAAGGGG KGLEWVSAISGSGG CTGGAGTGGGTCTCAGCTATTAGTGGTAGTGGT STYYADSVKGRFTI GGTAGCACATACTACGCAGACTCCGTGAAGGGC SRDNSKNTLYLQMN CGGTTCACCATCTCCAGAGACAATTCCAAGAAC SLRAEDTAVYYCAK ACGCTGTATCTGCAGATGAACAGCCTGAGAGCC VEWSTLLYFDYWGQ GAGGACACGGCCGTATATTACTGTGCGAAAGTT GTLVTVSSASTKGP GAATGGTCTACTCTGCTGTACTTCGACTACTGG SVFPLAPSSKSTSG GGCCAAGGAACCCTGGTCACCGTCTCGAGTGCT GTAALGCLVKDYFP AGCACCAAGGGCCCCTCCGTGTTCCCCCTGGCC EPVTVSWNSGALTS CCCAGCAGCAAGAGCACCAGCGGCGGCACAGCC GVHTFPAVLQSSGL GCTCTGGGCTGCCTGGTCAAGGACTACTTCCCC YSLSSVVTVPSSSL GAGCCCGTGACCGTGTCCTGGAACAGCGGAGCC GTQTYICNVNHKPS CTGACCTCCGGCGTGCACACCTTCCCCGCCGTG NTKVDKKVEPKSCD CTGCAGAGTTCTGGCCTGTATAGCCTGAGCAGC KTHTCPPCPAPEAA GTGGTCACCGTGCCTTCTAGCAGCCTGGGCACC GGPSVFLFPPKPKD CAGACCTACATCTGCAACGTGAACCACAAGCCC TLMISRTPEVTCVV AGCAACACCAAGGTGGACAAGAAGGTGGAGCCC VDVSHEDPEVKFNW AAGAGCTGCGACAAAACTCACACATGCCCACCG YVDGVEVHNAKTKP TGCCCAGCACCTGAAGCTGCAGGGGGACCGTCA REEQYNSTYRVVSV GTCTTCCTCTTCCCCCCAAAACCCAAGGACACC LTVLHQDWLNGKEY CTCATGATCTCCCGGACCCCTGAGGTCACATGC KCKVSNKALGAPIE GTGGTGGTGGACGTGAGCCACGAAGACCCTGAG KTISKAKGQPREPQ GTCAAGTTCAACTGGTACGTGGACGGCGTGGAG VCTLPPSRDELTKN GTGCATAATGCCAAGACAAAGCCGCGGGAGGAG QVSLSCAVKGFYPS CAGTACAACAGCACGTACCGTGTGGTCAGCGTC DIAVEWESNGQPEN CTCACCGTCCTGCACCAGGACTGGCTGAATGGC NYKTTPPVLDSDGS AAGGAGTACAAGTGCAAGGTCTCCAACAAAGCC FFLVSKLTVDKSRW CTCGGCGCCCCCATCGAGAAAACCATCTCCAAA QQGNVFSCSVMHEA GCCAAAGGGCAGCCCCGAGAACCACAGGTGTGC LHNHYTQKSLSLSP ACCCTGCCCCCATCCCGGGATGAGCTGACCAAG GK AACCAGGTCAGCCTCTCGTGCGCAGTCAAAGGC TTCTATCCCAGCGACATCGCCGTGGAGTGGGAG AGCAATGGGCAGCCGGAGAACAACTACAAGACC ACGCCTCCCGTGCTGGACTCCGACGGCTCCTTC TTCCTCGTGAGCAAGCTCACCGTGGACAAGAGC AGGTGGCAGCAGGGGAACGTCTTCTCATGCTCC GTGATGCATGAGGCTCTGCACAACCACTACACG CAGAAGAGCCTCTCCCTGTCTCCGGGTAAA 73 and 110A5 knob GAGGTGCAATTGTTGGAGTCTGGGGGAGGCTTG EVQLLESGGGLVQP 74 heavy chain GTACAGCCTGGGGGGTCCCTGAGACTCTCCTGT GGSLRLSCAASGFT GCAGCCTCCGGATTCACCTTTAGCAGTTATGCC FSSYAMSWVRQAPG ATGAGCTGGGTCCGCCAGGCTCCAGGGAAGGGG KGLEWVSAISGSGG CTGGAGTGGGTCTCAGCTATTAGTGGTAGTGGT STYYADSVKGRFTI GGTAGCACATACTACGCAGACTCCGTGAAGGGC SRDNSKNTLYLQMN CGGTTCACCATCTCCAGAGACAATTCCAAGAAC SLRAEDTAVYYCAK ACGCTGTATCTGCAGATGAACAGCCTGAGAGCC YSYRYVLAFDYWGQ GAGGACACGGCCGTATATTACTGTGCGAAATAC GTLVTVSSASTKGP TCTTACCGTTACGTTCTGGCTTTCGACTACTGG SVFPLAPSSKSTSG GGCCAAGGAACCCTGGTCACCGTCTCGAGTGCT GTAALGCLVKDYFP AGCACAAAGGGCCCTAGCGTGTTCCCTCTGGCC EPVTVSWNSGALTS CCCAGCAGCAAGAGCACAAGCGGCGGAACAGCC GVHTFPAVLQSSGL GCCCTGGGCTGCCTCGTGAAGGACTACTTCCCC YSLSSVVTVPSSSL GAGCCCGTGACAGTGTCTTGGAACAGCGGAGCC GTQTYICNVNHKPS CTGACAAGCGGCGTGCACACTTTCCCTGCCGTG NTKVDKKVEPKSCD CTGCAGAGCAGCGGCCTGTACTCCCTGAGCAGC GGGGSGGGGSEVQL GTGGTCACCGTGCCTAGCAGCAGCCTGGGCACC LESGGGLVQPGGSL CAGACCTACATCTGCAACGTGAACCACAAGCCC RLSCAASGFTFSTY AGCAACACCAAAGTGGACAAGAAGGTGGAGCCC AMNWVRQAPGKGLE AAGAGCTGTGATGGCGGAGGAGGGTCCGGAGGC WVSRIRSKYNNYAT GGAGGATCCGAGGTGCAGCTGCTGGAATCTGGC YYADSVKGRFTISR GGCGGACTGGTGCAGCCTGGCGGATCTCTGAGA DDSKNTLYLQMNSL CTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGC RAEDTAVYYCVRHG ACCTACGCCATGAACTGGGTGCGCCAGGCCCCT NFGNSYVSWFAYWG GGCAAAGGCCTGGAATGGGTGTCCCGGATCAGA QGTLVTVSSASTKG AGCAAGTACAACAACTACGCCACCTACTACGCC PSVFPLAPSSKSTS GACAGCGTGAAGGGCCGGTTCACCATCAGCCGG GGTAALGCLVKDYF GACGACAGCAAGAACACCCTGTACCTGCAGATG PEPVTVSWNSGALT AACAGCCTGCGGGCCGAGGACACCGCCGTGTAC SGVHTFPAVLQSSG TATTGTGTGCGGCACGGCAACTTCGGCAACAGC LYSLSSVVTVPSSS TATGTGTCTTGGTTTGCCTACTGGGGCCAGGGC LGTQTYICNVNHKP ACCCTCGTGACCGTGTCAAGCGCTAGTACCAAG SNTKVDKKVEPKSC GGCCCCAGCGTGTTCCCCCTGGCACCCAGCAGC DKTHTCPPCPAPEA AAGAGCACATCTGGCGGAACAGCCGCTCTGGGC AGGPSVFLFPPKPK TGTCTGGTGAAAGACTACTTCCCCGAGCCCGTG DTLMISRTPEVTCV ACCGTGTCTTGGAACTCTGGCGCCCTGACCAGC VVDVSHEDPEVKFN GGCGTGCACACCTTTCCAGCCGTGCTGCAGAGC WYVDGVEVHNAKTK AGCGGCCTGTACTCCCTGTCCTCCGTGGTCACC PREEQYNSTYRVVS GTGCCCTCTAGCTCCCTGGGAACACAGACATAT VLTVLHQDWLNGKE ATCTGTAATGTCAATCACAAGCCTTCCAACACC YKCKVSNKALGAPI AAAGTCGATAAGAAAGTCGAGCCCAAGAGCTGC EKTISKAKGQPREP GACAAAACTCACACATGCCCACCGTGCCCAGCA QVYTLPPCRDELTK CCTGAAGCTGCAGGGGGACCGTCAGTCTTCCTC NQVSLWCLVKGFYP TTCCCCCCAAAACCCAAGGACACCCTCATGATC SDIAVEWESNGQPE TCCCGGACCCCTGAGGTCACATGCGTGGTGGTG NNYKTTPPVLDSDG GACGTGAGCCACGAAGACCCTGAGGTCAAGTTC SFFLYSKLTVDKSR AACTGGTACGTGGACGGCGTGGAGGTGCATAAT WQQGNVFSCSVMHE GCCAAGACAAAGCCGCGGGAGGAGCAGTACAAC ALHNHYTQKSLSLS AGCACGTACCGTGTGGTCAGCGTCCTCACCGTC PGK CTGCACCAGGACTGGCTGAATGGCAAGGAGTAC AAGTGCAAGGTCTCCAACAAAGCCCTCGGCGCC CCCATCGAGAAAACCATCTCCAAAGCCAAAGGG CAGCCCCGAGAACCACAGGTGTACACCCTGCCC CCATGCCGGGATGAGCTGACCAAGAACCAGGTC AGCCTGTGGTGCCTGGTCAAAGGCTTCTATCCC AGCGACATCGCCGTGGAGTGGGAGAGCAATGGG CAGCCGGAGAACAACTACAAGACCACGCCTCCC GTGCTGGACTCCGACGGCTCCTTCTTCCTCTAC AGCAAGCTCACCGTGGACAAGAGCAGGTGGCAG CAGGGGAACGTCTTCTCATGCTCCGTGATGCAT GAGGCTCTGCACAACCACTACACGCAGAAGAGC CTCTCCCTGTCTCCGGGTAAA 75 and 110A5 hole GAGGTGCAATTGTTGGAGTCTGGGGGAGGCTTG EVQLLESGGGLVQP 76 heavy chain GTACAGCCTGGGGGGTCCCTGAGACTCTCCTGT GGSLRLSCAASGFT GCAGCCTCCGGATTCACCTTTAGCAGTTATGCC FSSYAMSWVRQAPG ATGAGCTGGGTCCGCCAGGCTCCAGGGAAGGGG KGLEWVSAISGSGG CTGGAGTGGGTCTCAGCTATTAGTGGTAGTGGT STYYADSVKGRFTI GGTAGCACATACTACGCAGACTCCGTGAAGGGC SRDNSKNTLYLQMN CGGTTCACCATCTCCAGAGACAATTCCAAGAAC SLRAEDTAVYYCAK ACGCTGTATCTGCAGATGAACAGCCTGAGAGCC YSYRYVLAFDYWGQ GAGGACACGGCCGTATATTACTGTGCGAAATAC GTLVTVSSASTKGP TCTTACCGTTACGTTCTGGCTTTCGACTACTGG SVFPLAPSSKSTSG GGCCAAGGAACCCTGGTCACCGTCTCGAGTGCT GTAALGCLVKDYFP AGCACCAAGGGCCCCTCCGTGTTCCCCCTGGCC EPVTVSWNSGALTS CCCAGCAGCAAGAGCACCAGCGGCGGCACAGCC GVHTFPAVLQSSGL GCTCTGGGCTGCCTGGTCAAGGACTACTTCCCC YSLSSVVTVPSSSL GAGCCCGTGACCGTGTCCTGGAACAGCGGAGCC GTQTYICNVNHKPS CTGACCTCCGGCGTGCACACCTTCCCCGCCGTG NTKVDKKVEPKSCD CTGCAGAGTTCTGGCCTGTATAGCCTGAGCAGC KTHTCPPCPAPEAA GTGGTCACCGTGCCTTCTAGCAGCCTGGGCACC GGPSVFLFPPKPKD CAGACCTACATCTGCAACGTGAACCACAAGCCC TLMISRTPEVTCVV AGCAACACCAAGGTGGACAAGAAGGTGGAGCCC VDVSHEDPEVKFNW AAGAGCTGCGACAAAACTCACACATGCCCACCG YVDGVEVHNAKTKP TGCCCAGCACCTGAAGCTGCAGGGGGACCGTCA REEQYNSTYRVVSV GTCTTCCTCTTCCCCCCAAAACCCAAGGACACC LTVLHQDWLNGKEY CTCATGATCTCCCGGACCCCTGAGGTCACATGC KCKVSNKALGAPIE GTGGTGGTGGACGTGAGCCACGAAGACCCTGAG KTISKAKGQPREPQ GTCAAGTTCAACTGGTACGTGGACGGCGTGGAG VCTLPPSRDELTKN GTGCATAATGCCAAGACAAAGCCGCGGGAGGAG QVSLSCAVKGFYPS CAGTACAACAGCACGTACCGTGTGGTCAGCGTC DIAVEWESNGQPEN CTCACCGTCCTGCACCAGGACTGGCTGAATGGC NYKTTPPVLDSDGS AAGGAGTACAAGTGCAAGGTCTCCAACAAAGCC FFLVSKLTVDKSRW CTCGGCGCCCCCATCGAGAAAACCATCTCCAAA QQGNVFSCSVMHEA GCCAAAGGGCAGCCCCGAGAACCACAGGTGTGC LHNHYTQKSLSLSP ACCCTGCCCCCATCCCGGGATGAGCTGACCAAG GK AACCAGGTCAGCCTCTCGTGCGCAGTCAAAGGC TTCTATCCCAGCGACATCGCCGTGGAGTGGGAG AGCAATGGGCAGCCGGAGAACAACTACAAGACC ACGCCTCCCGTGCTGGACTCCGACGGCTCCTTC TTCCTCGTGAGCAAGCTCACCGTGGACAAGAGC AGGTGGCAGCAGGGGAACGTCTTCTCATGCTCC GTGATGCATGAGGCTCTGCACAACCACTACACG CAGAAGAGCCTCTCCCTGTCTCCGGGTAAA

5) BCMA binders useful in common light chain format, variable heavy chain Amino Acid Sequences pSCHLI333-TCB pSCHLI372-TCB pSCHLI373-TCB BCMA VH QVQLVQSGAEVKKPGA EVQLVESGGGLVKPGGSLR EVQLVESGGGLVKPGGSLRLSC SVKVSCKASGYMFSSF LSCAASGFTFSNSGMIWVR AASGFSFSNSWMNWVRQAPGKG GMSWVRQAPGQGLEWM QAPGKGLEWVGHIRSKTDG LEWVGTIRQKTYGGTTDYAAPV GWIYPVGQGTWYAQKF GTTDYAAPVKGRFTISRDD KGRFTISRDDSKNTLYLQMNSL QGRVTMTRDTSTSTVY SKNTLYLQMNSLKTEDTAV KTEDTAVYYCTTGGLFGYWDYW MELSSLRSEDTAVYYC YYCTTGGSGSFDYWGQGTL GQGTLVTVSS (SEQ ID NO: ARVSYPPSHFDYWGQG VTVSS (SEQ ID NO: 79) TLVTVSS (SEQ ID 78) NO: 77) BCMA CDR1 GYMFSSFGMS (SEQ GFTFSNSGMI (SEQ ID GFSFSNSWMN (SEQ ID NO: (VH) ID NO: 80) NO: 81) 82) BCMA CDR2 WIYPVGQGTWYAQKFQ HIRSKTDGGTTDYAAPVKG TIRQKTYGGTTDYAAPVKG (VH) G (SEQ ID NO: (SEQ ID NO: 85) (SEQ ID NO: 86) 83) BCMA CDR3 VSYPPSHFDY (SEQ GGSGSFDY (SEQ ID GGLFGYWDY (SEQ ID NO: (VH) ID NO: 87) NO: 88) 89) CD3 VL QAVVTQEPSLTVSPGG QAVVTQEPSLTVSPGGTVT QAVVTQEPSLTVSPGGTVTLTC TVTLTCGSSTGAVTTS LTCGSSTGAVTTSNYANWV GSSTGAVTTSNYANWVQEKPGQ NYANWVQEKPGQAFRG QEKPGQAFRGLIGGTNKRA AFRGLIGGTNKRAPGTPARFSG LIGGTNKRAPGTPARF PGTPARFSGSLLGGKAALT SLLGGKAALTLSGAQPEDEAEY SGSLLGGKAALTLSGA LSGAQPEDEAEYYCALWYS YCALWYSNLWVFGGGTKLTVL QPEDEAEYYCALWYSN NLWVFGGGTKLTVL (SEQ (SEQ ID NO: 92) LWVFGGGTKLTVL ID NO: 91) (SEQ ID NO: 90) CD3 CDR1 GSSTGAVTTSNYAN GSSTGAVTTSNYAN (SEQ GSSTGAVTTSNYAN (SEQ ID (VL) (SEQ ID NO: 93) ID NO: 93) NO: 93) CD3 CDR2 GTNKRAP (SEQ ID GTNKRAP (SEQ ID NO: GTNKRAP (SEQ ID NO: (VL) NO: 94) 94) 94) CD3 CDR3 ALWYSNLWV (SEQ ALWYSNLWV (SEQ ID ALWYSNLWV (SEQ ID NO: (VL) ID NO: 95) NO: 95) 95) CD3 VH EVQLLESGGGLVQPGG EVQLLESGGGLVQPGGSLR EVQLLESGGGLVQPGGSLRLSC SLRLSCAASGFTFSTY LSCAASGFTFSTYAMNWVR AASGFTFSTYAMNWVRQAPGKG AMNWVRQAPGKGLEWV QAPGKGLEWVSRIRSKYNN LEWVSRIRSKYNNYATYYADSV SRIRSKYNNYATYYAD YATYYADSVKGRFTISRDD KGRFTISRDDSKNTLYLQMNSL SVKGRFTISRDDSKNT SKNTLYLQMNSLRAEDTAV RAEDTAVYYCVRHGNFGNSYVS LYLQMNSLRAEDTAVY YYCVRHGNFGNSYVSWFAY WFAYWGQGTLVTVSS (SEQ YCVRHGNFGNSYVSWF WGQGTLVTVSS (SEQ ID ID NO: 96) AYWGQGTLVTVSS NO: 96) (SEQ ID NO: 96) CD3 CDR1 TYAMN (SEQ ID TYAMN (SEQ ID NO: TYAMN (SEQ ID NO: 98) (VH) NO: 98) 98) CD3 CDR2 RIRSKYNNYATYYADS RIRSKYNNYATYYADSVKG RIRSKYNNYATYYADSVKG (VH) VKG (SEQ ID NO: (SEQ ID NO: 99) (SEQ ID NO: 99) 99) CD3 CDR3 HGNFGNSYVSWFAY HGNFGNSYVSWFAY (SEQ HGNFGNSYVSWFAY (SEQ ID (VH) (SEQ ID NO: 100) ID NO: 100) NO: 100) CD3 LC QAVVTQEPSLTVSPGG QAVVTQEPSLTVSPGGTVT QAVVTQEPSLTVSPGGTVTLTC (CLC) TVTLTCGSSTGAVTTS LTCGSSTGAVTTSNYANWV GSSTGAVTTSNYANWVQEKPGQ NYANWVQEKPGQAFRG QEKPGQAFRGLIGGTNKRA AFRGLIGGTNKRAPGTPARFSG LIGGTNKRAPGTPARF PGTPARFSGSLLGGKAALT SLLGGKAALTLSGAQPEDEAEY SGSLLGGKAALTLSGA LSGAQPEDEAEYYCALWYS YCALWYSNLWVFGGGTKLTVLG QPEDEAEYYCALWYSN NLWVFGGGTKLTVLGQPKA QPKAAPSVTLFPPSSEELQANK LWVFGGGTKLTVLGQP APSVTLFPPSSEELQANKA ATLVCLISDFYPGAVTVAWKAD KAAPSVTLFPPSSEEL TLVCLISDFYPGAVTVAWK SSPVKAGVETTTPSKQSNNKYA QANKATLVCLISDFYP ADSSPVKAGVETTTPSKQS ASSYLSLTPEQWKSHRSYSCQV GAVTVAWKADSSPVKA NNKYAASSYLSLTPEQWKS THEGSTVEKTVAPTECS (SEQ GVETTTPSKQSNNKYA HRSYSCQVTHEGSTVEKTV ID NO: 101) ASSYLSLTPEQWKSHR APTECS (SEQ ID NO: SYSCQVTHEGSTVEKT 101) VAPTECS (SEQ ID NO: 101) 2 + 1 Fc containing TCB (full molecule) BCMA QVQLVQSGAEVKKPGA n/a n/a pSCHLI333 SVKVSCKASGYMFSSF VH_CH1 × GMSWVRQAPGQGLEWM CD3 GWIYPVGQGTWYAQKF VH_CH1 Fcknob QGRVTMTRDTSTSTVY LALA MELSSLRSEDTAVYYC PG ARVSYPPSHFDYWGQG TLVTVSSASTKGPSVF PLAPSSKSTSGGTAAL GCLVKDYFPEPVTVSW NSGALTSGVHTFPAVL QSSGLYSLSSVVTVPS SSLGTQTYICNVNHKP SNTKVDKKVEPKSCDG GGGSGGGGSEVQLLES GGGLVQPGGSLRLSCA ASGFTFSTYAMNWVRQ APGKGLEWVSRIRSKY NNYATYYADSVKGRFT ISRDDSKNTLYLQMNS LRAEDTAVYYCVRHGN FGNSYVSWFAYWGQGT LVTVSSASTKGPSVFP LAPSSKSTSGGTAALG CLVKDYFPEPVTVSWN SGALTSGVHTFPAVLQ SSGLYSLSSVVTVPSS SLGTQTYICNVNHKPS NTKVDKKVEPKSCDKT HTCPPCPAPEAAGGPS VFLFPPKPKDTLMISR TPEVTCVVVDVSHEDP EVKFNWYVDGVEVHNA KTKPREEQYNSTYRVV SVLTVLHQDWLNGKEY KCKVSNKALGAPIEKT ISKAKGQPREPQVYTL PPCRDELTKNQVSLWC LVKGFYPSDIAVEWES NGQPENNYKTTPPVLD SDGSFFLYSKLTVDKS RWQQGNVFSCSVMHEA LHNHYTQKSLSLSPGK (SEQ ID NO: 102) BCMA QVQLVQSGAEVKKPGA n/a n/a pSCHLI333 SVKVSCKASGYMFSSF HC hole GMSWVRQAPGQGLEWM LALA PG GWIYPVGQGTWYAQKF QGRVTMTRDTSTSTVY MELSSLRSEDTAVYYC ARVSYPPSHFDYWGQG TLVTVSSASTKGPSVF PLAPSSKSTSGGTAAL GCLVKDYFPEPVTVSW NSGALTSGVHTFPAVL QSSGLYSLSSVVTVPS SSLGTQTYICNVNHKP SNTKVDKKVEPKSCDK THTCPPCPAPEAAGGP SVFLFPPKPKDTLMIS RTPEVTCVVVDVSHED PEVKFNWYVDGVEVHN AKTKPREEQYNSTYRV VSVLTVLHQDWLNGKE YKCKVSNKALGAPIEK TISKAKGQPREPQVCT LPPSRDELTKNQVSLS CAVKGFYPSDIAVEWE SNGQPENNYKTTPPVL DSDGSFFLVSKLTVDK SRWQQGNVFSCSVMHE ALHNHYTQKSLSLSPG K (SEQ ID NO: 103) BCMA n/a EVQLVESGGGLVKPGGSLR n/a pSCHLI372 LSCAASGFTFSNSGMIWVR VH_CH1 × QAPGKGLEWVGHIRSKTDG CD3 GTTDYAAPVKGRFTISRDD VH_CH1 Fcknob SKNTLYLQMNSLKTEDTAV LALA YYCTTGGSGSFDYWGQGTL PG VTVSSASTKGPSVFPLAPS SKSTSGGTAALGCLVKDYF PEPVTVSWNSGALTSGVHT FPAVLQSSGLYSLSSVVTV PSSSLGTQTYICNVNHKPS NTKVDKKVEPKSCDGGGGS GGGGSEVQLLESGGGLVQP GGSLRLSCAASGFTFSTYA MNWVRQAPGKGLEWVSRIR SKYNNYATYYADSVKGRFT ISRDDSKNTLYLQMNSLRA EDTAVYYCVRHGNFGNSYV SWFAYWGQGTLVTVSSAST KGPSVFPLAPSSKSTSGGT AALGCLVKDYFPEPVTVSW NSGALTSGVHTFPAVLQSS GLYSLSSVVTVPSSSLGTQ TYICNVNHKPSNTKVDKKV EPKSCDKTHTCPPCPAPEA AGGPSVFLFPPKPKDTLMI SRTPEVTCVVVDVSHEDPE VKFNWYVDGVEVHNAKTKP REEQYNSTYRVVSVLTVLH QDWLNGKEYKCKVSNKALG APIEKTISKAKGQPREPQV YTLPPCRDELTKNQVSLWC LVKGFYPSDIAVEWESNGQ PENNYKTTPPVLDSDGSFF LYSKLTVDKSRWQQGNVFS CSVMHEALHNHYTQKSLSL SPGK (SEQ ID NO: 110) BCMA n/a EVQLVESGGGLVKPGGSLR n/a pSCHLI372 LSCAASGFTFSNSGMIWVR HC hole QAPGKGLEWVGHIRSKTDG LALA PG GTTDYAAPVKGRFTISRDD SKNTLYLQMNSLKTEDTAV YYCTTGGSGSFDYWGQGTL VTVSSASTKGPSVFPLAPS SKSTSGGTAALGCLVKDYF PEPVTVSWNSGALTSGVHT FPAVLQSSGLYSLSSVVTV PSSSLGTQTYICNVNHKPS NTKVDKKVEPKSCDKTHTC PPCPAPEAAGGPSVFLFPP KPKDTLMISRTPEVTCVVV DVSHEDPEVKFNWYVDGVE VHNAKTKPREEQYNSTYRV VSVLTVLHQDWLNGKEYKC KVSNKALGAPIEKTISKAK GQPREPQVCTLPPSRDELT KNQVSLSCAVKGFYPSDIA VEWESNGQPENNYKTTPPV LDSDGSFFLVSKLTVDKSR WQQGNVFSCSVMHEALHNH YTQKSLSLSPGK (SEQ ID NO: 111) BCMA n/a n/a EVQLVESGGGLVKPGGSLRLSC pSCHLI373 AASGFSFSNSWMNWVRQAPGKG VH_CH1 × LEWVGTIRQKTYGGTTDYAAPV CD3 KGRFTISRDDSKNTLYLQMNSL VH_CH1 Fc KTEDTAVYYCTTGGLFGYWDYW knob LALA GQGTLVTVSSASTKGPSVFPLA PG PSSKSTSGGTAALGCLVKDYFP EPVTVSWNSGALTSGVHTFPAV LQSSGLYSLSSVVTVPSSSLGT QTYICNVNHKPSNTKVDKKVEP KSCDGGGGSGGGGSEVQLLESG GGLVQPGGSLRLSCAASGFTFS TYAMNWVRQAPGKGLEWVSRIR SKYNNYATYYADSVKGRFTISR DDSKNTLYLQMNSLRAEDTAVY YCVRHGNFGNSYVSWFAYWGQG TLVTVSSASTKGPSVFPLAPSS KSTSGGTAALGCLVKDYFPEPV TVSWNSGALTSGVHTFPAVLQS SGLYSLSSVVTVPSSSLGTQTY ICNVNHKPSNTKVDKKVEPKSC DKTHTCPPCPAPEAAGGPSVFL FPPKPKDTLMISRTPEVTCVVV DVSHEDPEVKFNWYVDGVEVHN AKTKPREEQYNSTYRVVSVLTV LHQDWLNGKEYKCKVSNKALGA PIEKTISKAKGQPREPQVYTLP PCRDELTKNQVSLWCLVKGFYP SDIAVEWESNGQPENNYKTTPP VLDSDGSFFLYSKLTVDKSRWQ QGNVFSCSVMHEALHNHYTQKS LSLSPGK (SEQ ID NO: 113) BCMA n/a n/a EVQLVESGGGLVKPGGSLRLSC pSCHLI373 AASGFSFSNSWMNWVRQAPGKG HC hole LEWVGTIRQKTYGGTTDYAAPV LALA PG KGRFTISRDDSKNTLYLQMNSL KTEDTAVYYCTTGGLFGYWDYW GQGTLVTVSSASTKGPSVFPLA PSSKSTSGGTAALGCLVKDYFP EPVTVSWNSGALTSGVHTFPAV LQSSGLYSLSSVVTVPSSSLGT QTYICNVNHKPSNTKVDKKVEP KSCDKTHTCPPCPAPEAAGGPS VFLFPPKPKDTLMISRTPEVTC VVVDVSHEDPEVKFNWYVDGVE VHNAKTKPREEQYNSTYRVVSV LTVLHQDWLNGKEYKCKVSNKA LGAPIEKTISKAKGQPREPQVC TLPPSRDELTKNQVSLSCAVKG FYPSDIAVEWESNGQPENNYKT TPPVLDSDGSFFLVSKLTVDKS RWQQGNVFSCSVMHEALHNHYT QKSLSLSPGK (SEQ ID NO: 114) 83A10 VH EVQLLESGGGLVQPGG SLRLSCAASGFTFSSY AMSWVRQAPGKGLEWV SAISGSGGSTYYADSV KGRFTISRDNSKNTLY LQMNSLRAEDTAVYYC AKVLGWFDYWGQGTLV TVSS (SEQ ID NO: 115) 83A10 SYAMS (SEQ ID CDRH1 NO: 116) 83A10 AISGSGGSTYYADSVK CDRH2 G (SEQ ID NO: 117) 83A10 VLGWFDY (SEQ ID CDRH3 NO: 118)

6) Untargeted DP47 Description Sequence Seq ID No Light Chain DP47 GS EIVLTQSPGTLSLSPGERATLSCRASQSVSSS 119 YLAWYQQKPGQAPRLLIYGASSRATGIPDR FSGSGSGTDFTLTISRLEPEDFAVYYCQQYG SSPLTFGQGTKVEIKRTVAAPSVFIFPPSDEQ LKSGTASVVCLLNNFYPREAKVQWKVDNA LQSGNSQESVTEQDSKDSTYSLSSTLTLSKA DYEKHKVYACEVTHQGLSSPVTKSFNRGEC Light Chain humanized QAVVTQEPSLTVSPGGTVTLTCGSSTGAVT 120 CD3CH2527 (Crossfab, VL- TSNYANWVQEKPGQAFRGLIGGTNKRAPG CH1) TPARFSGSLLGGKAALTLSGAQPEDEAEYY CALWYSNLWVFGGGTKLTVLSSASTKGPS VFPLAPSSKSTSGGTAALGCLVKDYFPEPVT VSWNSGALTSGVHTFPAVLQSSGLYSLSSV VTVPSSSLGTQTYICNVNHKPSNTKVDKKV EPKSC DP47 GS (VH-CH1) - EVQLLESGGGLVQPGGSLRLSCAASGFTFSS 121 humanized CD3CH2527 YAMSWVRQAPGKGLEWVSAISGSGGSTYY (Crossfab VH-Ck) - ADSVKGRFTISRDNSKNTLYLQMNSLRAED Fc(knob) P329GLALA TAVYYCAKGSGFDYWGQGTLVTVSSASTK GPSVFPLAPSSKSTSGGTAALGCLVKDYFPE PVTVSWNSGALTSGVHTFPAVLQSSGLYSL SSVVTVPSSSLGTQTYICNVNHKPSNTKVD KKVEPKSCDGGGGSGGGGSEVQLLESGGG LVQPGGSLRLSCAASGFTFSTYAMNWVRQ APGKGLEWVSRIRSKYNNYATYYADSVKG RFTISRDDSKNTLYLQMNSLRAEDTAVYYC VRHGNFGNSYVSWFAYWGQGTLVTVSSAS VAAPSVFIFPPSDEQLKSGTASVVCLLNNFY PREAKVQWKVDNALQSGNSQESVTEQDSK DSTYSLSSTLTLSKADYEKHKVYACEVTHQ GLSSPVTKSFNRGECDKTHTCPPCPAPEAA GGPSVFLFPPKPKDTLMISRTPEVTCVVVDV SHEDPEVKFNWYVDGVEVHNAKTKPREEQ YNSTYRVVSVLTVLHQDWLNGKEYKCKVS NKALGAPIEKTISKAKGQPREPQVYTLPPCR DELTKNQVSLWCLVKGFYPSDIAVEWESN GQPENNYKTTPPVLDSDGSFFLYSKLTVDK SRWQQGNVFSCSVMHEALHNHYTQKSLSL SPGK DP47 GS (VH-CH1) - EVQLLESGGGLVQPGGSLRLSCAASGFTFSS 122 Fc(hole)P329GLALA YAMSWVRQAPGKGLEWVSAISGSGGSTYY ADSVKGRFTISRDNSKNTLYLQMNSLRAED TAVYYCAKGSGFDYWGQGTLVTVSSASTK GPSVFPLAPSSKSTSGGTAALGCLVKDYFPE PVTVSWNSGALTSGVHTFPAVLQSSGLYSL SSVVTVPSSSLGTQTYICNVNHKPSNTKVD KKVEPKSCDKTHTCPPCPAPEAAGGPSVFL FPPKPKDTLMISRTPEVTCVVVDVSHEDPE VKFNWYVDGVEVHNAKTKPREEQYNSTY RVVSVLTVLHQDWLNGKEYKCKVSNKAL GAPIEKTISKAKGQPREPQVCTLPPSRDELT KNQVSLSCAVKGFYPSDIAVEWESNGQPEN NYKTTPPVLDSDGSFFLVSKLTVDKSRWQQ GNVFSCSVMHEALHNHYTQKSLSLSPGK

7) Exemplary target sequences Description Sequence Seq ID No Human CD3 Human FolR1 (ECD 25- RIAWARTELLNVCMNAKHHKEKPGPEDKLHEQCRPWRKNA 227 234) CCSTNTSQEAHKDVSYLYRENWNHCGEMAPACKRHFIQDT CLYECSPNLGPWIQQVDQSWRKERVLNVPLCKEDCEQWWE DCRTSYTCKSNWHKGWNWTSGFNKCAVGAACQPFHFYFPT PTVLCNEIWTHSYKVSNYSRGSGRCIQMWFDPAQGNPNEE VARFYAAAM murine FolR1 (ECD 25- TRARTELLNVCMDAKHHKEKPGPEDNLHDQCSPWKTNSCC 230 232) STNTSQEAHKDISYLYRFNWNHCGTMTSECKRHFIQDTCL YECSPNLGPWIQQVDQSWRKERILDVPLCKEDCQQWWEDC QSSFTCKSNWHKGWNWSSGHNECPVGASCHPFTFYFPTSA ALCEEIWSHSYKLSNYSRGSGRCIQMWFDPAQGNPNEEVA RFYAEAMS cynomolgus FolR1 (ECD EAQTRTARARTELLNVCMNAKHHKEKPGPEDKLHEQCRPW 231 25-234) KKNACCSTNTSQEAHKDVSYLYRENWNHCGEMAPACKRHF IQDTCLYECSPNLGPWIQQVDQSWRKERVLNVPLCKEDCE RWWEDCRTSYCKSNWHKGWNWTSGFNKCPVGAACQPFHFY FPTPTVLCNEIWTYSYKVSNYSRGSGRCIQMWFDPAQGNP NEEVARFYAAAMS

8) Nudeotide sequences of exemplary embodiments Description Sequence Seq ID No 16A3 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 151 CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA GTGTACTACTGTGCACGCAACTACTACGCTGGTGTTACTCCGTTCG ACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT 15A1 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 152 CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA GTGTACTACTGTGCACGCAACTACTACATCGGTGTTGTTACTTTCG ACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT 18D3 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 153 CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA GTGTACTACTGTGCACGCAACTACTACACTGGTGGTTCTTCTGCTT TCGACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT 19E5 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 154 NTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA GTGTACTACTGTGCACGCGGTGAATGGCGTCGTTACACTTCTTTCG ACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT 19A4 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 155 CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA GTGTACTACTGTGCACGCGGTGGTTGGATCCGTTGGGAACATTTCG ACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT 15H7 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 156 CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA GTGTACTACTGTGCACGCAACTACTACCTGTTCTCTACTTCTTTCG ACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT 15B6 CAGGTGCAATTGGTTCAATCTGGTGCTGAGGTAAAAAAACCGGGCG 157 CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA GTGTACTACTGTGCACGCAACTACTACATCGGTATCGTTCCGTTCG ACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT 16D5 GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCG 158 GTTCCCTGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAA CGCGTGGATGAGCTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAG TGGGTTGGTCGTATCAAGTCTAAAACTGACGGTGGCACCACGGATT ACGCGGCTCCAGTTAAAGGTCGTTTTACCATTTCCCGCGACGATAG CAAAAACACTCTGTATCTGCAGATGAACTCTCTGAAAACTGAAGAC ACCGCAGTCTACTACTGTACTACCCCGTGGGAATGGTCTTGGTACG ATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCC 15E12 GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCG 159 GTTCCCNGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAA CGCGTGGATGAGCTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAG TGGGTTGGTCGTATCAAGTCTAAAACTGACGGTGGCACCACGGATT ACGCGGCTCCAGTTAAAGGTCGTTTTACCATTTCCCGCGACGATAG CAAAAACACTCTGTATCTGCAGATGAACTCTCTGAAAACCGAAGAC ACCGCAGTCTACTACTGTACTACCCCGTGGGAATGGTCTTACTTCG ATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCC 21D1 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 160 CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA GTGTACTACTGTGCACGCAACTACTACGTTGGTGTTTCTCCGTTCG ACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT 16F12 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 161 NTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCNTACGCGC AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA GTGTACTACTGTGCACGCAACTTCACTGTTCTGCGTGTTCCGTTCG ACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT 21A5 GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCG 162 GTTCCCTGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAA CGCGTGGATGAGCTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAG TGGGTTGGTCGTATCAAGTCTAAAACTGACGGTGGCACCACGGATT ACGCGGCTCCAGTTAAAGGTCGTTTTACCATTTCCCGCGACGATAG CAAAAACACTCTGTATCTGCAGATGAACTCTCTGAAAACTGAAGAC ACCGCAGTCTACTACTGTACTACCCCGTGGGAATGGGCTTGGTTCG ATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCC 21G8 GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCG 163 GTTCCCTGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAA CGCGTGGATGAGCTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAG TGGGTTGGTCGTATCAAGTCTAAAACTGACGGTGGCACCACGGATT ACGCGGCTCCAGTTAAAGGTCGTTTTACCATTTCCCGCGACGATAG CAAAAACACTCTGTATCTGCAGATGAACTCTCTGAAAACCGAAGAC ACCGCAGTCTACTACTGTACTACCCCTTGGGAATGGGCTTACTTCG ATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCC 19H3 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 164 CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA GTGTACTACTGTGCACGCACTGGTTGGTCTCGTTGGGGTTACATGG ACTATTGGGGCCAAGGCACCCTCGTAACGGTTTCTTCT 20G6 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 165 CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA GTGTACTACTGTGCACGCGGTGAATGGATCCGTTACTACCATTTCG ACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT 20H7 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 166 CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA GTGTACTACTGTGCACGCGTTGGTTGGTACCGTTGGGGTTACATGG ACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT 11F8_VH CAGGTGCAATTGGTGCAGTCTGGGGCTGAGGTGAAGAAGCCTGGGT 167 CCTCGGTGAAGGTCTCCTGCAAGGCCTCCGGAGGCACATTCAGCAG CTACGCTATAAGCTGGGTGCGACAGGCCCCTGGACAAGGGCTCGAG TGGATGGGAGGGATCATCCCTATCTTTGGTACAGCAAACTACGCAC AGAAGTTCCAGGGCAGGGTAACCATTACTGCAGACAAATCCACGAG CACAGCCTACATGGAGCTGAGCAGCCTGAGATCTGAGGACACCGCC GTGTATTACTGTGCGAGAGCTGTTTTCTACCGTGCTTGGTACTCTT TCGACTACTGGGGCCAAGGGACCACCGTGACCGTCTCCTCA 11F8_VL GACATCCAGATGACCCAGTCTCCTTCCACCCTGTCTGCATCTGTAG 168 GAGACCGTGTCACCATCACTTGCCGTGCCAGTCAGAGTATTAGTAG CTGGTTGGCCTGGTATCAGCAGAAACCAGGGAAAGCCCCTAAGCTC CTGATCTATGATGCCTCCAGTTTGGAAAGTGGGGTCCCATCACGTT TCAGCGGCAGTGGATCCGGGACAGAATTCACTCTCACCATCAGCAG CTTGCAGCCTGATGATTTTGCAACTTATTACTGCCAACAGTATACC AGCCCACCACCAACGTTTGGCCAGGGCACCAAAGTCGAGATCAAG 36F2_VH CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 169 CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC AGAAATTCCAGGGTCGCGTCACGATGACCCATGACACTAGCACCTC TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA GTGTACTACTGTGCACGCTCTTTCTTCACTGGTTTCCATCTGGACT ATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT 36F2_VL GAAATCGTGTTAACGCAGTCTCCAGGCACCCTGTCTTTGTCTCCAG 170 GGGAAAGAGCCACCCTCTCTTGCAGGGCCAGTCAGAGTGTTAGCAG CAGCTACTTAGCCTGGTACCAGCAGAAACCTGGCCAGGCTCCCAGG CTCCTCATCTATGGAGCATCCAGCAGGGCCACTGGCATCCCAGACA GGTTCAGTGGCAGTGGATCCGGGACAGACTTCACTCTCACCATCAG CAGACTGGAGCCTGAAGATTTTGCAGTGTATTACTGTCAGCAGTAT ACCAACGAACATTATTATACGTTCGGCCAGGGGACCAAAGTGGAAA TCAAA 9D11_VH CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 171 CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA TGGATGGGCATCATTAACCCAAGCGGTGGCCCTACCTCCTACGCGC AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA GTGTACTACTGTGCACGCGGTGACTTCGCTTGGCTGGACTATTGGG GTCAAGGCACCCTCGTAACGGTTTCTTCT 9D11_VL GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAG 172 GCGAACCGGCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCA CTCCAACGGCTACAACTATCTCGATTGGTACCTGCAAAAACCGGGT CAGAGCCCTCAGCTGCTGATCTACCTGGGCTCTAACCGCGCTTCCG GTGTACCGGACCGTTTCAGCGGCTCTGGATCCGGCACCGATTTCAC GTTGAAAATCAGCCGTGTTGAAGCAGAAGACGTGGGCGTTTATTAC TGTATGCAGGCAAGCATTATGAACCGGACTTTTGGTCAAGGCACCA AGGTCGAAATTAAA 9D11_VL GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAG 173 N95S GCGAACCGGCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCA CTCCAACGGCTACAACTATCTCGATTGGTACCTGCAAAAACCGGGT CAGAGCCCTCAGCTGCTGATCTACCTGGGCTCTAACCGCGCTTCCG GTGTACCGGACCGTTTCAGCGGCTCTGGATCCGGCACCGATTTCAC GTTGAAAATCAGCCGTGTTGAAGCAGAAGACGTGGGCGTTTATTAC TGTATGCAGGCAAGCATTATGAGCCGGACTTTTGGTCAAGGCACCA AGGTCGAAATTAAA 9D11_VL GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAG 174 N95Q GCGAACCGGCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCA CTCCAACGGCTACAACTATCTCGATTGGTACCTGCAAAAACCGGGT CAGAGCCCTCAGCTGCTGATCTACCTGGGCTCTAACCGCGCTTCCG GTGTACCGGACCGTTTCAGCGGCTCTGGATCCGGCACCGATTTCAC GTTGAAAATCAGCCGTGTTGAAGCAGAAGACGTGGGCGTTTATTAC TGTATGCAGGCAAGCATTATGCAGCGGACTTTTGGTCAAGGCACCA AGGTCGAAATTAAA 9D11_VL GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAG 175 T97A GCGAACCGGCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCA CTCCAACGGCTACAACTATCTCGATTGGTACCTGCAAAAACCGGGT CAGAGCCCTCAGCTGCTGATCTACCTGGGCTCTAACCGCGCTTCCG GTGTACCGGACCGTTTCAGCGGCTCTGGATCCGGCACCGATTTCAC GTTGAAAATCAGCCGTGTTGAAGCAGAAGACGTGGGCGTTTATTAC TGTATGCAGGCAAGCATTATGAACCGGGCTTTTGGTCAAGGCACCA AGGTCGAAATTAAA 9D11_VL GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAG 176 T97N GCGAACCGGCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCA CTCCAACGGCTACAACTATCTCGATTGGTACCTGCAAAAACCGGGT CAGAGCCCTCAGCTGCTGATCTACCTGGGCTCTAACCGCGCTTCCG GTGTACCGGACCGTTTCAGCGGCTCTGGATCCGGCACCGATTTCAC GTTGAAAATCAGCCGTGTTGAAGCAGAAGACGTGGGCGTTTATTAC TGTATGCAGGCAAGCATTATGAACCGGAATTTTGGTCAAGGCACCA AGGTCGAAATTAAA 5D9_VH CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 177 CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA GTGTACTACTGTGCACGCTCTTACATCGACATGGACTATTGGGGTC AAGGCACCCTCGTAACGGTTTCTTCT 5D9_VL GAAATCGTGTTAACGCAGTCTCCAGGCACCCTGTCTTTGTCTCCAG 178 GGGAAAGAGCCACCCTCTCTTGCAGGGCCAGTCAGAGTGTTAGCAG CAGCTACTTAGCCTGGTACCAGCAGAAACCTGGCCAGGCTCCCAGG CTCCTCATCTATGGAGCATCCAGCAGGGCCACTGGCATCCCAGACA GGTTCAGTGGCAGTGGATCCGGGACAGACTTCACTCTCACCATCAG CAGACTGGAGCCTGAAGATTTTGCAGTGTATTACTGTCAGCAGGAT AACTGGAGCCCAACGTTCGGCCAGGGGACCAAAGTGGAAATCAAA 6B6_VH CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 179 CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA TGGATGGGCATCATTAACCCAAGCGGTGGCTCTACCTCCTACGCGC AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA GTGTACTACTGTGCACGCTCTTACGTTGACATGGACTATTGGGGTC AAGGCACCCTCGTAACGGTTTCTTCT 6B6_VL GAAATCGTGTTAACGCAGTCTCCAGGCACCCTGTCTTTGTCTCCAG 180 GGGAAAGAGCCACCCTCTCTTGCAGGGCCAGTCAGAGTGTTAGCAG CAGCTACCTAGCCTGGTACCAGCAGAAACCTGGCCAGGCTCCCAGG CTCCTCATCTATGGAGCATCCAGCAGGGCCACTGGCATCCCAGACA GGTTCAGTGGCAGTGGATCCGGGACAGACTTCACTCTCACCATCAG CAGACTGGAGCCTGAAGATTTTGCAGTGTATTACTGTCAGCAGGAT ATTTGGAGCCCAACGTTCGGCCAGGGGACCAAAGTGGAAATCAAA 14E4_VH GAGGTGCAATTGTTGGAGTCTGGGGGAGGCTTGGTACAGCCTGGGG 181 GGTCCCTGAGACTCTCCTGTGCAGCCTCCGGATTCACCTTTAGCAG TTATGCCATGAGCTGGGTCCGCCAGGCTCCAGGGAAGGGGCTGGAG TGGGTCTCAGCTATTAGTGGTAGTGGTGGTAGCACATACTACGCAG ACTCCGTGAAGGGCCGGTTCACCATCTCCAGAGACAATTCCAAGAA CACGCTGTATCTGCAGATGAACAGCCTGAGAGCCGAGGACACGGCC GTATATTACTGTGCGAAAGACTCTTCTTACGTTGAATGGTACGCTT TCGACTACTGGGGCCAAGGAACCCTGGTCACCGTCTCGAGT 14E4_VL GAAATCGTGTTAACGCAGTCTCCAGGCACCCTGTCTTTGTCTCCAG 182 GGGAAAGAGCCACCCTCTCTTGCAGGGCCAGTCAGAGTGTTAGCAG CAGCTACTTAGCCTGGTACCAGCAGAAACCTGGCCAGGCTCCCAGG CTCCTCATCTATGGAGCATCCAGCAGGGCCACTGGCATCCCAGACA GGTTCAGTGGCAGTGGATCCGGGACAGACTCCACTCTCACCATCAG CAGACTGGAGCCTGAAGATTTTGCAGTGTATTACTGTCAGCAGCCA ACCAGCAGCCCAATTACGTTCGGCCAGGGGACCAAAGTGGAAATCA AA CD3 heavy GAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGGTGCAGCCT 183 chain GGCGGATCTCTGAGACTGAGCTGTGCCGCCAGCGGCTTCACC (VHCH1) TTCAGCACCTACGCCATGAACTGGGTGCGCCAGGCCCCTGGC AAAGGCCTGGAATGGGTGTCCCGGATCAGAAGCAAGTACAAC AACTACGCCACCTACTACGCCGACAGCGTGAAGGGCCGGTTC ACCATCAGCCGGGACGACAGCAAGAACACCCTGTACCTGCAG ATGAACAGCCTGCGGGCCGAGGACACCGCCGTGTACTATTGT GTGCGGCACGGCAACTTCGGCAACAGCTATGTGTCTTGGTTT GCCTACTGGGGCCAGGGCACCCTCGTGACCGTGTCAAGCGCT AGTACCAAGGGCCCCAGCGTGTTCCCCCTGGCACCCAGCAGC AAGAGCACATCTGGCGGAACAGCCGCTCTGGGCTGTCTGGTG AAAGACTACTTCCCCGAGCCCGTGACCGTGTCTTGGAACTCT GGCGCCCTGACCAGCGGCGTGCACACCTTTCCAGCCGTGCTG CAGAGCAGCGGCCTGTACTCCCTGTCCTCCGTGGTCACCGTG CCCTCTAGCTCCCTGGGAACACAGACATATATCTGTAATGTC AATCACAAGCCTTCCAACACCAAAGTCGATAAGAAAGTCGAG CCCAAGAGCTGC Crossed GAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGGTGCAGCCTGGCG 184 CD3 heavy GATCTCTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCAC chain CTACGCCATGAACTGGGTGCGCCAGGCCCCTGGCAAAGGCCTGGAA (VHCκ) TGGGTGTCCCGGATCAGAAGCAAGTACAACAACTACGCCACCTACT ACGCCGACAGCGTGAAGGGCCGGTTCACCATCAGCCGGGACGACAG CAAGAACACCCTGTACCTGCAGATGAACAGCCTGCGGGCCGAGGAC ACCGCCGTGTACTATTGTGTGCGGCACGGCAACTTCGGCAACAGCT ATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCACCCTCGTGACCGT GTCAAGCGCTAGTGTGGCCGCTCCCTCCGTGTTTATCTTTCCCCCA TCCGATGAACAGCTGAAAAGCGGCACCGCCTCCGTCGTGTGTCTGC TGAACAATTTTTACCCTAGGGAAGCTAAAGTGCAGTGGAAAGTGGA TAACGCACTGCAGTCCGGCAACTCCCAGGAATCTGTGACAGAACAG GACTCCAAGGACAGCACCTACTCCCTGTCCTCCACCCTGACACTGT CTAAGGCTGATTATGAGAAACACAAAGTCTACGCCTGCGAAGTCAC CCATCAGGGCCTGAGCTCGCCCGTCACAAAGAGCTTCAACAGGGGA GAGTGT Mutagenesis GCAGGCAAGCATTATGCAGCGGACTTTTGGTCAAGG 185 primer GAB7734 N95Q Mutagenesis CAGGCAAGCATTATGAGCCGGACTTTTGGTCAAGG 186 primer GAB7735 N95S Mutagenesis CATTATGAACCGGGCTTTTGGTCAAGGCACCAAGGTC 187 primer GAB7736 T97A Mutagenesis CATTATGAACCGGAATTTTGGTCAAGGCACCAAGGTC 188 primer GAB7737 T97N VHCH1[16D5]_VHCH1 GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCG 189 [CD3]_Fcknob_PGLALA GTTCCCTGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAA pCON999 CGCGTGGATGAGCTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAG (Inverted TGGGTTGGTCGTATCAAGTCTAAAACTGACGGTGGCACCACGGATT TCB with ACGCGGCTCCAGTTAAAGGTCGTTTTACCATTTCCCGCGACGATAG 16D5 2 + 1: CAAAAACACTCTGTATCTGCAGATGAACTCTCTGAAAACTGAAGAC pCON999 + ACCGCAGTCTACTACTGTACTACCCCGTGGGAATGGTCTTGGTACG pCON983 + ATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCCGCTAGCAC pETR13197) AAAGGGCCCTAGCGTGTTCCCTCTGGCCCCCAGCAGCAAGAGCACA AGCGGCGGAACAGCCGCCCTGGGCTGCCTCGTGAAGGACTACTTCC CCGAGCCCGTGACAGTGTCTTGGAACAGCGGAGCCCTGACAAGCGG CGTGCACACTTTCCCTGCCGTGCTGCAGAGCAGCGGCCTGTACTCC CTGAGCAGCGTGGTCACCGTGCCTAGCAGCAGCCTGGGCACCCAGA CCTACATCTGCAACGTGAACCACAAGCCCAGCAACACCAAAGTGGA CAAGAAGGTGGAGCCCAAGAGCTGTGATGGCGGAGGAGGGTCCGGA GGCGGAGGATCCGAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGG TGCAGCCTGGCGGATCTCTGAGACTGAGCTGTGCCGCCAGCGGCTT CACCTTCAGCACCTACGCCATGAACTGGGTGCGCCAGGCCCCTGGC AAAGGCCTGGAATGGGTGTCCCGGATCAGAAGCAAGTACAACAACT ACGCCACCTACTACGCCGACAGCGTGAAGGGCCGGTTCACCATCAG CCGGGACGACAGCAAGAACACCCTGTACCTGCAGATGAACAGCCTG CGGGCCGAGGACACCGCCGTGTACTATTGTGTGCGGCACGGCAACT TCGGCAACAGCTATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCAC CCTCGTGACCGTGTCAAGCGCTAGTACCAAGGGCCCCAGCGTGTTC CCCCTGGCACCCAGCAGCAAGAGCACATCTGGCGGAACAGCCGCTC TGGGCTGTCTGGTGAAAGACTACTTCCCCGAGCCCGTGACCGTGTC TTGGAACTCTGGCGCCCTGACCAGCGGCGTGCACACCTTTCCAGCC GTGCTGCAGAGCAGCGGCCTGTACTCCCTGTCCTCCGTGGTCACCG TGCCCTCTAGCTCCCTGGGAACACAGACATATATCTGTAATGTCAA TCACAAGCCTTCCAACACCAAAGTCGATAAGAAAGTCGAGCCCAAG AGCTGCGACAAAACTCACACATGCCCACCGTGCCCAGCACCTGAAG CTGCAGGGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGA CACCCTCATGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTG GACGTGAGCCACGAAGACCCTGAGGTCAAGTTCAACTGGTACGTGG ACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCA GTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCAC CAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACA AAGCCCTCGGCGCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGG GCAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATGCCGGGAT GAGCTGACCAAGAACCAGGTCAGCCTGTGGTGCCTGGTCAAAGGCT TCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCC GGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGC TCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGC AGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCA CAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA VHCH1[16D5]_Fchole_PGLALA_HYRF GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCG 190 pCON983 GTTCCCTGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAA CGCGTGGATGAGCTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAG TGGGTTGGTCGTATCAAGTCTAAAACTGACGGTGGCACCACGGATT ACGCGGCTCCAGTTAAAGGTCGTTTTACCATTTCCCGCGACGATAG CAAAAACACTCTGTATCTGCAGATGAACTCTCTGAAAACTGAAGAC ACCGCAGTCTACTACTGTACTACCCCGTGGGAATGGTCTTGGTACG ATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCCGCTAGCAC CAAGGGCCCCTCCGTGTTCCCCCTGGCCCCCAGCAGCAAGAGCACC AGCGGCGGCACAGCCGCTCTGGGCTGCCTGGTCAAGGACTACTTCC CCGAGCCCGTGACCGTGTCCTGGAACAGCGGAGCCCTGACCTCCGG CGTGCACACCTTCCCCGCCGTGCTGCAGAGTTCTGGCCTGTATAGC CTGAGCAGCGTGGTCACCGTGCCTTCTAGCAGCCTGGGCACCCAGA CCTACATCTGCAACGTGAACCACAAGCCCAGCAACACCAAGGTGGA CAAGAAGGTGGAGCCCAAGAGCTGCGACAAAACTCACACATGCCCA CCGTGCCCAGCACCTGAAGCTGCAGGGGGACCGTCAGTCTTCCTCT TCCCCCCAAAACCCAAGGACACCCTCATGATCTCCCGGACCCCTGA GGTCACATGCGTGGTGGTGGACGTGAGCCACGAAGACCCTGAGGTC AAGTTCAACTGGTACGTGGACGGCGTGGAGGTGCATAATGCCAAGA CAAAGCCGCGGGAGGAGCAGTACAACAGCACGTACCGTGTGGTCAG CGTCCTCACCGTCCTGCACCAGGACTGGCTGAATGGCAAGGAGTAC AAGTGCAAGGTCTCCAACAAAGCCCTCGGCGCCCCCATCGAGAAAA CCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCACAGGTGTGCAC CCTGCCCCCATCCCGGGATGAGCTGACCAAGAACCAGGTCAGCCTC TCGTGCGCAGTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGT GGGAGAGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCC CGTGCTGGACTCCGACGGCTCCTTCTTCCTCGTGAGCAAGCTCACC GTGGACAAGAGCAGGTGGCAGCAGGGGAACGTCTTCTCATGCTCCG TGATGCATGAGGCTCTGCACAACCGCTTCACGCAGAAGAGCCTCTC CCTGTCTCCGGGTAAA CD3_common CAGGCCGTCGTGACCCAGGAACCCAGCCTGACAGTGTCTCCTGGCG 191 light GCACCGTGACCCTGACATGTGGCAGTTCTACAGGCGCCGTGACCAC chain CAGCAACTACGCCAACTGGGTGCAGGAAAAGCCCGGCCAGGCCTTC pETR13197 AGAGGACTGATCGGCGGCACCAACAAGAGAGCCCCTGGCACCCCTG CCAGATTCAGCGGATCTCTGCTGGGAGGAAAGGCCGCCCTGACACT GTCTGGCGCCCAGCCAGAAGATGAGGCCGAGTACTACTGCGCCCTG TGGTACAGCAACCTGTGGGTGTTCGGCGGAGGCACCAAGCTGACAG TCCTAGGTCAACCCAAGGCTGCCCCCAGCGTGACCCTGTTCCCCCC CAGCAGCGAGGAACTGCAGGCCAACAAGGCCACCCTGGTCTGCCTG ATCAGCGACTTCTACCCAGGCGCCGTGACCGTGGCCTGGAAGGCCG ACAGCAGCCCCGTGAAGGCCGGCGTGGAGACCACCACCCCCAGCAA GCAGAGCAACAACAAGTACGCCGCCAGCAGCTACCTGAGCCTGACC CCCGAGCAGTGGAAGAGCCACAGGTCCTACAGCTGCCAGGTGACCC ACGAGGGCAGCACCGTGGAGAAAACCGTGGCCCCCACCGAGTGCAGC VHCH1[CD3]_VHCH1 GAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGGTGCAGCCTGGCG 192 [16D5]_Fcknob_PGLALA GATCTCTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCAC pETR13932 CTACGCCATGAACTGGGTGCGCCAGGCCCCTGGCAAAGGCCTGGAA (Classical TGGGTGTCCCGGATCAGAAGCAAGTACAACAACTACGCCACCTACT TCB with ACGCCGACAGCGTGAAGGGCCGGTTCACCATCAGCCGGGACGACAG 16D5; 2 + 1: CAAGAACACCCTGTACCTGCAGATGAACAGCCTGCGGGCCGAGGAC pETR13932 + ACCGCCGTGTACTATTGTGTGCGGCACGGCAACTTCGGCAACAGCT pCON983 + ATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCACCCTCGTGACCGT pETR13197) GTCATCTGCTAGCACAAAGGGCCCTAGCGTGTTCCCTCTGGCCCCC AGCAGCAAGAGCACAAGCGGCGGAACAGCCGCCCTGGGCTGCCTCG TGAAGGACTACTTCCCCGAGCCCGTGACAGTGTCTTGGAACAGCGG AGCCCTGACAAGCGGCGTGCACACCTTCCCTGCCGTGCTGCAGAGC AGCGGCCTGTACTCCCTGAGCAGCGTGGTCACCGTGCCTAGCAGCA GCCTGGGCACCCAGACCTACATCTGCAACGTGAACCACAAGCCCAG CAACACCAAAGTGGACAAGAAGGTGGAGCCCAAGAGCTGTGATGGC GGAGGAGGGTCCGGAGGCGGAGGATCCGAGGTGCAATTGGTTGAAT CTGGTGGTGGTCTGGTAAAACCGGGCGGTTCCCTGCGTCTGAGCTG CGCGGCTTCCGGATTCACCTTCTCCAACGCGTGGATGAGCTGGGTT CGCCAGGCCCCGGGCAAAGGCCTCGAGTGGGTTGGTCGTATCAAGT CTAAAACTGACGGTGGCACCACGGATTACGCGGCTCCAGTTAAAGG TCGTTTTACCATTTCCCGCGACGATAGCAAAAACACTCTGTATCTG CAGATGAACTCTCTGAAAACTGAAGACACCGCAGTCTACTACTGTA CTACCCCGTGGGAATGGTCTTGGTACGATTATTGGGGCCAGGGCAC GCTGGTTACGGTGTCTAGCGCTAGTACCAAGGGCCCCAGCGTGTTC CCCCTGGCACCCAGCAGCAAGAGCACATCTGGCGGAACAGCCGCTC TGGGCTGTCTGGTGAAAGACTACTTCCCCGAGCCCGTGACCGTGTC TTGGAACTCTGGCGCCCTGACCAGCGGCGTGCACACCTTTCCAGCC GTGCTGCAGAGCAGCGGCCTGTACTCCCTGTCCTCCGTGGTCACCG TGCCCTCTAGCTCCCTGGGAACACAGACATATATCTGTAATGTCAA TCACAAGCCTTCCAACACCAAAGTCGATAAGAAAGTCGAGCCCAAG AGCTGCGACAAAACTCACACATGCCCACCGTGCCCAGCACCTGAAG CTGCAGGGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGA CACCCTCATGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTG GACGTGAGCCACGAAGACCCTGAGGTCAAGTTCAACTGGTACGTGG ACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCA GTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCAC CAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACA AAGCCCTCGGCGCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGG GCAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATGCCGGGAT GAGCTGACCAAGAACCAGGTCAGCCTGTGGTGCCTGGTCAAAGGCT TCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCC GGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGC TCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGC AGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCA CAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA VHCH1[CD3]_Fcknob_PGLALA GAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGGTGCAGCCTGGCG 193 pETR13719 GATCTCTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCAC (16D5 IgG CTACGCCATGAACTGGGTGCGCCAGGCCCCTGGCAAAGGCCTGGAA format, 1 + 1: TGGGTGTCCCGGATCAGAAGCAAGTACAACAACTACGCCACCTACT pETR13719 + ACGCCGACAGCGTGAAGGGCCGGTTCACCATCAGCCGGGACGACAG pCON983 + CAAGAACACCCTGTACCTGCAGATGAACAGCCTGCGGGCCGAGGAC pETR13197) ACCGCCGTGTACTATTGTGTGCGGCACGGCAACTTCGGCAACAGCT ATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCACCCTCGTGACCGT GTCATCTGCTAGCACCAAGGGCCCATCGGTCTTCCCCCTGGCACCC TCCTCCAAGAGCACCTCTGGGGGCACAGCGGCCCTGGGCTGCCTGG TCAAGGACTACTTCCCCGAACCGGTGACGGTGTCGTGGAACTCAGG CGCCCTGACCAGCGGCGTGCACACCTTCCCGGCTGTCCTACAGTCC TCAGGACTCTACTCCCTCAGCAGCGTGGTGACCGTGCCCTCCAGCA GCTTGGGCACCCAGACCTACATCTGCAACGTGAATCACAAGCCCAG CAACACCAAGGTGGACAAGAAAGTTGAGCCCAAATCTTGTGACAAA ACTCACACATGCCCACCGTGCCCAGCACCTGAAGCTGCAGGGGGAC CGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGACACCCTCATGAT CTCCCGGACCCCTGAGGTCACATGCGTGGTGGTGGACGTGAGCCAC GAAGACCCTGAGGTCAAGTTCAACTGGTACGTGGACGGCGTGGAGG TGCATAATGCCAAGACAAAGCCGCGGGAGGAGCAGTACAACAGCAC GTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGACTGGCTG AATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCCTCGGCG CCCCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCCGAGA ACCACAGGTGTACACCCTGCCCCCATGCCGGGATGAGCTGACCAAG AACCAGGTCAGCCTGTGGTGCCTGGTCAAAGGCTTCTATCCCAGCG ACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCCGGAGAACAACTA CAAGACCACGCCTCCCGTGCTGGACTCCGACGGCTCCTTCTTCCTC TACAGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAGGGGAACG TCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACCACTACAC GCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA Fc_hole_PGLALA_HYRF GACAAAACTCACACATGCCCACCGTGCCCAGCACCTGAACTCCTGG 194 pETR10755 GGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGACACCCT (16D5 Head- CATGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTGGACGTG to-tail, 1 + 1: AGCCACGAAGACCCTGAGGTCAAGTTCAACTGGTACGTGGACGGCG pCON999 + TGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCAGTACAA pETR10755 + CAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGAC pETR13197) TGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCC TCCCAGCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCC CCGAGAACCACAGGTGTGCACCCTGCCCCCATCCCGGGATGAGCTG ACCAAGAACCAGGTCAGCCTCTCGTGCGCAGTCAAAGGCTTCTATC CCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCCGGAGAA CAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGCTCCTTC TTCCTCGTGAGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAGG GGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACCG CTTCACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA VHCH1[9D11]_VHCL CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 195 [CD3]_Fcknob_PGLALA CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC pCON1057 CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA (9D11 TGGATGGGCATCATTAACCCAAGCGGTGGCCCTACCTCCTACGCGC inverted AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC format, 2 + 1: TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA pCON1057 + GTGTACTACTGTGCACGCGGTGACTTCGCTTGGCTGGACTATTGGG pCON1051 + GTCAAGGCACCCTCGTAACGGTTTCTTCTGCTAGCACAAAGGGCCC pCON1063 + CAGCGTGTTCCCTCTGGCCCCTAGCAGCAAGAGCACATCTGGCGGA pETR12940) ACAGCCGCCCTGGGCTGCCTCGTGAAGGACTACTTTCCCGAGCCTG TGACCGTGTCCTGGAACTCTGGCGCCCTGACAAGCGGCGTGCACAC CTTTCCAGCCGTGCTGCAGAGCAGCGGCCTGTACTCTCTGAGCAGC GTGGTCACCGTGCCTAGCAGCAGCCTGGGCACCCAGACCTACATCT GCAACGTGAACCACAAGCCCAGCAACACCAAAGTGGACAAGAAGGT GGAGCCCAAGAGCTGTGATGGCGGAGGAGGGTCCGGAGGCGGAGGA TCCGAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGGTGCAGCCTG GCGGATCTCTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAG CACCTACGCCATGAACTGGGTGCGCCAGGCCCCTGGCAAAGGCCTG GAATGGGTGTCCCGGATCAGAAGCAAGTACAACAACTACGCCACCT ACTACGCCGACAGCGTGAAGGGCCGGTTCACCATCAGCCGGGACGA CAGCAAGAACACCCTGTACCTGCAGATGAACAGCCTGCGGGCCGAG GACACCGCCGTGTACTATTGTGTGCGGCACGGCAACTTCGGCAACA GCTATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCACCCTCGTGAC CGTGTCAAGCGCTAGTGTGGCCGCTCCCTCCGTGTTTATCTTTCCC CCATCCGATGAACAGCTGAAAAGCGGCACCGCCTCCGTCGTGTGTC TGCTGAACAATTTTTACCCTAGGGAAGCTAAAGTGCAGTGGAAAGT GGATAACGCACTGCAGTCCGGCAACTCCCAGGAATCTGTGACAGAA CAGGACTCCAAGGACAGCACCTACTCCCTGTCCTCCACCCTGACAC TGTCTAAGGCTGATTATGAGAAACACAAAGTCTACGCCTGCGAAGT CACCCATCAGGGCCTGAGCTCGCCCGTCACAAAGAGCTTCAACAGG GGAGAGTGTGACAAGACCCACACCTGTCCCCCTTGTCCTGCCCCTG AAGCTGCTGGCGGCCCTTCTGTGTTCCTGTTCCCCCCAAAGCCCAA GGACACCCTGATGATCAGCCGGACCCCCGAAGTGACCTGCGTGGTG GTGGATGTGTCCCACGAGGACCCTGAAGTGAAGTTCAATTGGTACG TGGACGGCGTGGAAGTGCACAACGCCAAGACAAAGCCGCGGGAGGA GCAGTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTG CACCAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCA ACAAAGCCCTCGGCGCCCCCATCGAGAAAACCATCTCCAAAGCCAA AGGGCAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATGCCGG GATGAGCTGACCAAGAACCAGGTCAGCCTGTGGTGCCTGGTCAAAG GCTTCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCA GCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGAC GGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGT GGCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCT GCACAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA 9D11_Fchole_PGLALA_HYRF CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCG 196 pCON1051 CTTCCGTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTC CTATTACATGCACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAA TGGATGGGCATCATTAACCCAAGCGGTGGCCCTACCTCCTACGCGC AGAAATTCCAGGGTCGCGTCACGATGACCCGTGACACTAGCACCTC TACCGTTTATATGGAGCTGTCCAGCCTGCGTTCTGAAGATACTGCA GTGTACTACTGTGCACGCGGTGACTTCGCTTGGCTGGACTATTGGG GTCAAGGCACCCTCGTAACGGTTTCTTCTGCTAGCACCAAGGGCCC CTCCGTGTTCCCCCTGGCCCCCAGCAGCAAGAGCACCAGCGGCGGC ACAGCCGCTCTGGGCTGCCTGGTCAAGGACTACTTCCCCGAGCCCG TGACCGTGTCCTGGAACAGCGGAGCCCTGACCTCCGGCGTGCACAC CTTCCCCGCCGTGCTGCAGAGTTCTGGCCTGTATAGCCTGAGCAGC GTGGTCACCGTGCCTTCTAGCAGCCTGGGCACCCAGACCTACATCT GCAACGTGAACCACAAGCCCAGCAACACCAAGGTGGACAAGAAGGT GGAGCCCAAGAGCTGCGACAAAACTCACACATGCCCACCGTGCCCA GCACCTGAAGCTGCAGGGGGACCGTCAGTCTTCCTCTTCCCCCCAA AACCCAAGGACACCCTCATGATCTCCCGGACCCCTGAGGTCACATG CGTGGTGGTGGACGTGAGCCACGAAGACCCTGAGGTCAAGTTCAAC TGGTACGTGGACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGC GGGAGGAGCAGTACAACAGCACGTACCGTGTGGTCAGCGTCCTCAC CGTCCTGCACCAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAG GTCTCCAACAAAGCCCTCGGCGCCCCCATCGAGAAAACCATCTCCA AAGCCAAAGGGCAGCCCCGAGAACCACAGGTGTGCACCCTGCCCCC ATCCCGGGATGAGCTGACCAAGAACCAGGTCAGCCTCTCGTGCGCA GTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCA ATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGGA CTCCGACGGCTCCTTCTTCCTCGTGAGCAAGCTCACCGTGGACAAG AGCAGGTGGCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATG AGGCTCTGCACAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCC GGGTAAA 9D11_LC GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAG 197 pCON1063 GCGAACCGGCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCA CTCCAACGGCTACAACTATCTCGATTGGTACCTGCAAAAACCGGGT CAGAGCCCTCAGCTGCTGATCTACCTGGGCTCTAACCGCGCTTCCG GTGTACCGGACCGTTTCAGCGGCTCTGGATCCGGCACCGATTTCAC GTTGAAAATCAGCCGTGTTGAAGCAGAAGACGTGGGCGTTTATTAC TGTATGCAGGCAAGCATTATGAACCGGACTTTTGGTCAAGGCACCA AGGTCGAAATTAAACGTACGGTGGCTGCACCATCTGTCTTCATCTT CCCGCCATCTGATGAGCAGTTGAAATCTGGAACTGCCTCTGTTGTG TGCCTGCTGAATAACTTCTATCCCAGAGAGGCCAAAGTACAGTGGA AGGTGGATAACGCCCTCCAATCGGGTAACTCCCAGGAGAGTGTCAC AGAGCAGGACAGCAAGGACAGCACCTACAGCCTCAGCAGCACCCTG ACGCTGAGCAAAGCAGACTACGAGAAACACAAAGTCTACGCCTGCG AAGTCACCCATCAGGGCCTGAGCTCGCCCGTCACAAAGAGCTTCAA CAGGGGAGAGTGT VLCH1C[CD3] CAGGCCGTCGTGACCCAGGAACCCAGCCTGACAGTGTCTCCTGGCG 198 pETR12940 GCACCGTGACCCTGACATGTGGCAGTTCTACAGGCGCCGTGACCAC CAGCAACTACGCCAACTGGGTGCAGGAAAAGCCCGGCCAGGCCTTC AGAGGACTGATCGGCGGCACCAACAAGAGAGCCCCTGGCACCCCTG CCAGATTCAGCGGATCTCTGCTGGGAGGAAAGGCCGCCCTGACACT GTCTGGCGCCCAGCCAGAAGATGAGGCCGAGTACTACTGCGCCCTG TGGTACAGCAACCTGTGGGTGTTCGGCGGAGGCACCAAGCTGACAG TGCTGAGCAGCGCTTCCACCAAAGGCCCTTCCGTGTTTCCTCTGGC TCCTAGCTCCAAGTCCACCTCTGGAGGCACCGCTGCTCTCGGATGC CTCGTGAAGGATTATTTTCCTGAGCCTGTGACAGTGTCCTGGAATA GCGGAGCACTGACCTCTGGAGTGCATACTTTCCCCGCTGTGCTGCA GTCCTCTGGACTGTACAGCCTGAGCAGCGTGGTGACAGTGCCCAGC AGCAGCCTGGGCACCCAGACCTACATCTGCAACGTGAACCACAAGC CCAGCAACACCAAGGTGGACAAGAAGGTGGAACCCAAGTCTTGT VHCL[CD3]_Fcknob_PGLALA GAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGGTGCAGCCTGGCG 199 pETR13378 GATCTCTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCAC (9D11 CTACGCCATGAACTGGGTGCGCCAGGCCCCTGGCAAAGGCCTGGAA CrossMab TGGGTGTCCCGGATCAGAAGCAAGTACAACAACTACGCCACCTACT format, 1 + 1: ACGCCGACAGCGTGAAGGGCCGGTTCACCATCAGCCGGGACGACAG pETR13378 + CAAGAACACCCTGTACCTGCAGATGAACAGCCTGCGGGCCGAGGAC pCON1051 + ACCGCCGTGTACTATTGTGTGCGGCACGGCAACTTCGGCAACAGCT pCON1063+ ATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCACCCTCGTGACCGT pETR12940) GTCATCTGCTAGCGTGGCCGCTCCCTCCGTGTTTATCTTTCCCCCA TCCGATGAACAGCTGAAAAGCGGCACCGCCTCCGTCGTGTGTCTGC TGAACAATTTTTACCCTAGGGAAGCTAAAGTGCAGTGGAAAGTGGA TAACGCACTGCAGTCCGGCAACTCCCAGGAATCTGTGACAGAACAG GACTCCAAGGACAGCACCTACTCCCTGTCCTCCACCCTGACACTGT CTAAGGCTGATTATGAGAAACACAAAGTCTACGCCTGCGAAGTCAC CCATCAGGGCCTGAGCTCGCCCGTCACAAAGAGCTTCAACAGGGGA GAGTGTGACAAGACCCACACCTGTCCCCCTTGTCCTGCCCCTGAAG CTGCTGGCGGCCCTTCTGTGTTCCTGTTCCCCCCAAAGCCCAAGGA CACCCTGATGATCAGCCGGACCCCCGAAGTGACCTGCGTGGTGGTG GATGTGTCCCACGAGGACCCTGAAGTGAAGTTCAATTGGTACGTGG ACGGCGTGGAAGTGCACAACGCCAAGACAAAGCCGCGGGAGGAGCA GTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCAC CAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACA AAGCCCTCGGCGCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGG GCAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATGCCGGGAT GAGCTGACCAAGAACCAGGTCAGCCTGTGGTGCCTGGTCAAAGGCT TCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCC GGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGC TCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGC AGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCA CAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA 16D5 GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCG 200 inverted GTTCCCTGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAA 2 + 1 with CGCGTGGATGAGCTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAG N100A in TGGGTTGGTCGTATCAAGTCTAAAACTGACGGTGGCACCACGGATT CDR H3 ACGCGGCTCCAGTTAAAGGTCGTTTTACCATTTCCCGCGACGATAG pETR14096 CAAAAACACTCTGTATCTGCAGATGAACTCTCTGAAAACTGAAGAC (pETR14096 + ACCGCAGTCTACTACTGTACTACCCCGTGGGAATGGTCTTGGTACG pCON983 + ATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCCGCTAGCAC pETR13197) AAAGGGCCCTAGCGTGTTCCCTCTGGCCCCCAGCAGCAAGAGCACA AGCGGCGGAACAGCCGCCCTGGGCTGCCTCGTGAAGGACTACTTCC CCGAGCCCGTGACAGTGTCTTGGAACAGCGGAGCCCTGACAAGCGG CGTGCACACTTTCCCTGCCGTGCTGCAGAGCAGCGGCCTGTACTCC CTGAGCAGCGTGGTCACCGTGCCTAGCAGCAGCCTGGGCACCCAGA CCTACATCTGCAACGTGAACCACAAGCCCAGCAACACCAAAGTGGA CAAGAAGGTGGAGCCCAAGAGCTGTGATGGCGGAGGAGGGTCCGGA GGCGGAGGATCCGAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGG TGCAGCCTGGCGGATCTCTGAGACTGAGCTGTGCCGCCAGCGGCTT CACCTTCAGCACCTACGCCATGAACTGGGTGCGCCAGGCCCCTGGC AAAGGCCTGGAATGGGTGTCCCGGATCAGAAGCAAGTACAACAACT ACGCCACCTACTACGCCGACAGCGTGAAGGGCCGGTTCACCATCAG CCGGGACGACAGCAAGAACACCCTGTACCTGCAGATGAACAGCCTG CGGGCCGAGGACACCGCCGTGTACTATTGTGTGCGGCACGGCAACT TCGGCGCCAGCTATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCAC CCTCGTGACCGTGTCAAGCGCTAGTACCAAGGGCCCCAGCGTGTTC CCCCTGGCACCCAGCAGCAAGAGCACATCTGGCGGAACAGCCGCTC TGGGCTGTCTGGTGAAAGACTACTTCCCCGAGCCCGTGACCGTGTC TTGGAACTCTGGCGCCCTGACCAGCGGCGTGCACACCTTTCCAGCC GTGCTGCAGAGCAGCGGCCTGTACTCCCTGTCCTCCGTGGTCACCG TGCCCTCTAGCTCCCTGGGAACACAGACATATATCTGTAATGTCAA TCACAAGCCTTCCAACACCAAAGTCGATAAGAAAGTCGAGCCCAAG AGCTGCGACAAAACTCACACATGCCCACCGTGCCCAGCACCTGAAG CTGCAGGGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGA CACCCTCATGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTG GACGTGAGCCACGAAGACCCTGAGGTCAAGTTCAACTGGTACGTGG ACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCA GTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCAC CAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACA AAGCCCTCGGCGCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGG GCAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATGCCGGGAT GAGCTGACCAAGAACCAGGTCAGCCTGTGGTGCCTGGTCAAAGGCT TCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCC GGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGC TCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGC AGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCA CAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA 16D5 GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCG 201 inverted GTTCCCTGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAA 2 + 1 with CGCGTGGATGAGCTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAG S100aA in TGGGTTGGTCGTATCAAGTCTAAAACTGACGGTGGCACCACGGATT CDR H3 ACGCGGCTCCAGTTAAAGGTCGTTTTACCATTTCCCGCGACGATAG pETR14097 CAAAAACACTCTGTATCTGCAGATGAACTCTCTGAAAACTGAAGAC (pETR14097 + ACCGCAGTCTACTACTGTACTACCCCGTGGGAATGGTCTTGGTACG pCON983 + ATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCCGCTAGCAC pETR13197) AAAGGGCCCTAGCGTGTTCCCTCTGGCCCCCAGCAGCAAGAGCACA AGCGGCGGAACAGCCGCCCTGGGCTGCCTCGTGAAGGACTACTTCC CCGAGCCCGTGACAGTGTCTTGGAACAGCGGAGCCCTGACAAGCGG CGTGCACACTTTCCCTGCCGTGCTGCAGAGCAGCGGCCTGTACTCC CTGAGCAGCGTGGTCACCGTGCCTAGCAGCAGCCTGGGCACCCAGA CCTACATCTGCAACGTGAACCACAAGCCCAGCAACACCAAAGTGGA CAAGAAGGTGGAGCCCAAGAGCTGTGATGGCGGAGGAGGGTCCGGA GGCGGAGGATCCGAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGG TGCAGCCTGGCGGATCTCTGAGACTGAGCTGTGCCGCCAGCGGCTT CACCTTCAGCACCTACGCCATGAACTGGGTGCGCCAGGCCCCTGGC AAAGGCCTGGAATGGGTGTCCCGGATCAGAAGCAAGTACAACAACT ACGCCACCTACTACGCCGACAGCGTGAAGGGCCGGTTCACCATCAG CCGGGACGACAGCAAGAACACCCTGTACCTGCAGATGAACAGCCTG CGGGCCGAGGACACCGCCGTGTACTATTGTGTGCGGCACGGCAACT TCGGCAACGCCTATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCAC CCTCGTGACCGTGTCAAGCGCTAGTACCAAGGGCCCCAGCGTGTTC CCCCTGGCACCCAGCAGCAAGAGCACATCTGGCGGAACAGCCGCTC TGGGCTGTCTGGTGAAAGACTACTTCCCCGAGCCCGTGACCGTGTC TTGGAACTCTGGCGCCCTGACCAGCGGCGTGCACACCTTTCCAGCC GTGCTGCAGAGCAGCGGCCTGTACTCCCTGTCCTCCGTGGTCACCG TGCCCTCTAGCTCCCTGGGAACACAGACATATATCTGTAATGTCAA TCACAAGCCTTCCAACACCAAAGTCGATAAGAAAGTCGAGCCCAAG AGCTGCGACAAAACTCACACATGCCCACCGTGCCCAGCACCTGAAG CTGCAGGGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGA CACCCTCATGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTG GACGTGAGCCACGAAGACCCTGAGGTCAAGTTCAACTGGTACGTGG ACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCA GTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCAC CAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACA AAGCCCTCGGCGCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGG GCAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATGCCGGGAT GAGCTGACCAAGAACCAGGTCAGCCTGTGGTGCCTGGTCAAAGGCT TCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCC GGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGC TCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGC AGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCA CAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA CD3 light CAGGCCGTCGTGACCCAGGAACCCAGCCTGACAGTGTCTCCTGGCG 202 chain fused GCACCGTGACCCTGACATGTGGCAGTTCTACAGGCGCCGTGACCAC to CH1; CAGCAACTACGCCAACTGGGTGCAGGAAAAGCCCGGCCAGGCCTTC Fc_PGLALA; AGAGGACTGATCGGCGGCACCAACAAGAGAGCCCCTGGCACCCCTG pETR13862 CCAGATTCAGCGGATCTCTGCTGGGAGGAAAGGCCGCCCTGACACT (Kappa- GTCTGGCGCCCAGCCAGAAGATGAGGCCGAGTACTACTGCGCCCTG lambda TGGTACAGCAACCTGTGGGTGTTCGGCGGAGGCACCAAGCTGACAG antibody with TGCTGAGCAGCGCTAGCACCAAGGGCCCATCGGTCTTCCCCCTGGC CD3 common ACCCTCCTCCAAGAGCACCTCTGGGGGCACAGCGGCCCTGGGCTGC light chain CTGGTCAAGGACTACTTCCCCGAACCGGTGACGGTGTCGTGGAACT fused to CH1 + CAGGCGCCCTGACCAGCGGCGTGCACACCTTCCCGGCTGTCCTACA Fc_PGLALA. GTCCTCAGGACTCTACTCCCTCAGCAGCGTGGTGACCGTGCCCTCC VHs fused to AGCAGCTTGGGCACCCAGACCTACATCTGCAACGTGAATCACAAGC kappa or CCAGCAACACCAAGGTGGACAAGAAAGTTGAGCCCAAATCTTGTGA lambda CAAAACTCACACATGCCCACCGTGCCCAGCACCTGAAGCTGCAGGG constant chain GGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGACACCCTCA pETR13859 + TGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTGGACGTGAG pETR13860 + CCACGAAGACCCTGAGGTCAAGTTCAACTGGTACGTGGACGGCGTG pETR13862) GAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCAGTACAACA GCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGACTG GCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCCTC GGCGCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCC GAGAACCA CAGGTGTACACCCTGCCCCCATCCCGGGATGAGCTGACCAAGAACC AGGTCAGCCTGACCTGCCTGGTCAAAGGCTTCTATCCCAGCGACAT CGCCGTGGAGTGGGAGAGCAATGGGCAGCCGGAGAACAACTACAAG ACCACGCCTCCCGTGCTGGACTCCGACGGCTCCTTCTTCCTCTACA GCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAGGGGAACGTCTT CTCATGCTCCGTGATGCATGAGGCTCTGCACAACCACTACACGCAG AAGAGCCTCTCCCTGTCTCCGGGTAAA 16D5 VH GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCG 203 fused to GTTCCCTGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAA constant CGCGTGGATGAGCTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAG kappa TGGGTTGGTCGTATCAAGTCTAAAACTGACGGTGGCACCACGGATT chain; ACGCGGCTCCAGTTAAAGGTCGTTTTACCATTTCCCGCGACGATAG pETR13859 CAAAAACACTCTGTATCTGCAGATGAACTCTCTGAAAACTGAAGAC ACCGCAGTCTACTACTGTACTACCCCGTGGGAATGGTCTTGGTACG ATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCCGCTAGCGT GGCCGCTCCCTCCGTGTTCATCTTCCCACCTTCCGACGAGCAGCTG AAGTCCGGCACCGCTTCTGTCGTGTGCCTGCTGAACAACTTCTACC CCCGCGAGGCCAAGGTGCAGTGGAAGGTGGACAACGCCCTGCAGTC CGGCAACAGCCAGGAATCCGTGACCGAGCAGGACTCCAAGGACAGC ACCTACTCCCTGTCCTCCACCCTGACCCTGTCCAAGGCCGACTACG AGAAGCACAAGGTGTACGCCTGCGAAGTGACCCACCAGGGCCTGTC TAGCCCCGTGACCAAGTCTTTCAACCGGGGCGAGTGC CD3 VH GAAGTGCAGCTGCTGGAATCCGGCGGAGGACTGGTGCAGCCTGGCG 204 fused to GATCTCTGAGACTGTCTTGTGCCGCCTCCGGCTTCACCTTCTCCAC constant CTACGCCATGAACTGGGTGCGACAGGCTCCTGGCAAGGGCCTGGAA lambda TGGGTGTCCCGGATCAGATCCAAGTACAACAACTACGCCACCTACT chain; ACGCCGACTCCGTGAAGGGCCGGTTCACCATCTCTCGGGACGACTC pETR13860 CAAGAACACCCTGTACCTGCAGATGAACTCCCTGCGGGCCGAGGAC ACCGCCGTGTACTATTGTGTGCGGCACGGCAACTTCGGCAACTCCT ATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCACCCTCGTGACCGT GTCATCTGCTAGCCCCAAGGCTGCCCCCAGCGTGACCCTGTTTCCC CCCAGCAGCGAGGAACTGCAGGCCAACAAGGCCACCCTGGTCTGCC TGATCAGCGACTTCTACCCAGGCGCCGTGACCGTGGCCTGGAAGGC CGACAGCAGCCCCGTGAAGGCCGGCGTGGAGACCACCACCCCCAGC AAGCAGAGCAACAACAAGTACGCCGCCAGCAGCTACCTGAGCCTGA CCCCCGAGCAGTGGAAGAGCCACAGGTCCTACAGCTGCCAGGTGAC CCACGAGGGCAGCACC GTGGAGAAAACCGTGGCCCCCACCGAGTGCAGC VHCH1[36F2]_VHCL CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCC 246 [CD3]_Fcknob_PGLALA GTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATG pCON1056 CACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATT pETR12940 AACCCAAGCGGTGGCTCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTC ACGATGACCCATGACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGC CTGCGTTCTGAAGATACTGCAGTGTACTACTGTGCACGCTCTTTCTTCACT GGTTTCCATCTGGACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCT GCTAGCACAAAGGGCCCCAGCGTGTTCCCTCTGGCCCCTAGCAGCAAGAGC ACATCTGGCGGAACAGCCGCCCTGGGCTGCCTCGTGAAGGACTACTTTCCC GAGCCTGTGACCGTGTCCTGGAACTCTGGCGCCCTGACAAGCGGCGTGCAC ACCTTTCCAGCCGTGCTGCAGAGCAGCGGCCTGTACTCTCTGAGCAGCGTG GTCACCGTGCCTAGCAGCAGCCTGGGCACCCAGACCTACATCTGCAACGTG AACCACAAGCCCAGCAACACCAAAGTGGACAAGAAGGTGGAGCCCAAG AGCTGTGATGGCGGAGGAGGGTCCGGAGGCGGAGGATCCGAGGTGCAGCTG CTGGAATCTGGCGGCGGACTGGTGCAGCCTGGCGGATCTCTGAGACTGAGC TGTGCCGCCAGCGGCTTCACCTTCAGCACCTACGCCATGAACTGGGTGCGC CAGGCCCCTGGCAAAGGCCTGGAATGGGTGTCCCGGATCAGAAGCAAGTAC AACAACTACGCCACCTACTACGCCGACAGCGTGAAGGGCCGGTTCACCATC AGCCGGGACGACAGCAAGAACACCCTGTACCTGCAGATGAACAGCCTGCGG GCCGAGGACACCGCCGTGTACTATTGTGTGCGGCACGGCAACTTCGGCAAC AGCTATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCACCCTCGTGACCGTG TCAAGCGCTAGTGTGGCCGCTCCCTCCGTGTTTATCTTTCCCCCATCCGAT GAACAGCTGAAAAGCGGCACCGCCTCCGTCGTGTGTCTGCTGAACAATTTT TACCCTAGGGAAGCTAAAGTGCAGTGGAAAGTGGATAACGCACTGCAGTCC CTGGGAGGAAAGGCCGCCCTGACACTGTCTGGCGCCCAGCCAGAAGATGAG GCCGAGTACTACTGCGCCCTGTGGTACAGCAACCTGTGGGTGTTCGGCGGA GGCACCAAGCTGACAGTGCTGAGCAGCGCTTCCACCAAAGGCCCTTCCGTG TTTCCTCTGGCTCCTAGCTCCAAGTCCACCTCTGGAGGCACCGCTGCTCTC GGATGCCTCGTGAAGGATTATTTTCCTGAGCCTGTGACAGTGTCCTGGAAT AGCGGAGCACTGACCTCTGGAGTGCATACTTTCCCCGCTGTGCTGCAGTCC TCTGGACTGTACAGCCTGAGCAGCGTGGTGACAGTGCCCAGCAGCAGCCTG GGCACCCAGACCTACATCTGCAACGTGAACCACAAGCCCAGCAACACCAAG GTGGACAAGAAGGTGGAACCCAAGTCTTGT Name Sequence Seq ID No K53A CAGACCGTCGTGACCCAGGAACCCAGCCTGACAGTGTCTCCTGGCGGCACC 205 nt GTGACCCTGACATGTGGCAGTTCTACAGGCGCCGTGACCACCAGCAACTAC GCCAACTGGGTGCAGCAGAAGCCAGGCCAGGCTCCCAGAGGACTGATCGGC GGCACCAACGCCAGAGCCCCTGGCACCCCTGCCAGATTCAGCGGATCTCTG CTGGGAGGAAAGGCCGCCCTGACACTGTCTGGCGTGCAGCCTGAAGATGAG GCCGAGTACTACTGCGCCCTGTGGTACAGCAACCTGTGGGTGTTCGGCGGA GGCACCAAGCTGACAGTCCTA S93A CAGACCGTCGTGACCCAGGAACCCAGCCTGACAGTGTCTCCTGGCGGCACC 206 nt GTGACCCTGACATGTGGCAGTTCTACAGGCGCCGTGACCACCAGCAACTAC GCCAACTGGGTGCAGCAGAAGCCAGGCCAGGCTCCCAGAGGACTGATCGGC GGCACCAACAAGAGAGCCCCTGGCACCCCTGCCAGATTCAGCGGATCTCTG CTGGGAGGAAAGGCCGCCCTGACACTGTCTGGCGTGCAGCCTGAAGATGAG GCCGAGTACTACTGCGCCCTGTGGTACGCCAACCTGTGGGTGTTCGGCGGA GGCACCAAGCTGACAGTCCTA S35H GAGGTGCAATTGGTGGAAAGCGGAGGCGGCCTCGTGAAGCCTGGCGGATCT 207 nt CTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCAACGCCTGGATG CACTGGGTGCGCCAGGCCCCTGGAAAAGGACTCGAGTGGGTGGGACGGATC AAGAGCAAGACCGATGGCGGCACCACCGACTATGCCGCCCCTGTGAAGGGC CGGTTCACCATCAGCAGGGACGACAGCAAGAACACCCTGTACCTGCAGATG AACAGCCTGAAAACCGAGGACACCGCCGTGTACTACTGCACCACCCCCTGG GAGTGGTCTTGGTACGACTATTGGGGCCAGGGCACCCTCGTGACCGTGTCC TCTGCTAGC G49S GAGGTGCAATTGGTGGAAAGCGGAGGCGGCCTCGTGAAGCCTGGCGGATCT 208 nt CTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCAACGCCTGGATG AGCTGGGTGCGCCAGGCCCCTGGAAAAGGACTCGAGTGGGTGTCCCGGATC AAGAGCAAGACCGATGGCGGCACCACCGACTATGCCGCCCCTGTGAAGGGC CGGTTCACCATCAGCAGGGACGACAGCAAGAACACCCTGTACCTGCAGATG AACAGCCTGAAAACCGAGGACACCGCCGTGTACTACTGCACCACCCCCTGG GAGTGGTCTTGGTACGACTATTGGGGCCAGGGCACCCTCGTGACCGTGTCC TCTGCTAGC R50S GAGGTGCAATTGGTGGAAAGCGGAGGCGGCCTCGTGAAGCCTGGCGGATCT 209 nt CTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCAACGCCTGGATG AGCTGGGTGCGCCAGGCCCCTGGAAAAGGACTCGAGTGGGTGGGATCTATC AAGAGCAAGACCGACGGCGGCACCACCGACTATGCCGCCCCTGTGAAGGGC CGGTTCACCATCAGCAGGGACGACAGCAAGAACACCCTGTACCTGCAGATG AACAGCCTGAAAACCGAGGACACCGCCGTGTACTACTGCACCACCCCCTGG GAGTGGTCTTGGTACGACTATTGGGGCCAGGGCACCCTCGTGACCGTGTCC TCT GCTAGC W96Y GAGGTGCAATTGGTGGAAAGCGGAGGCGGCCTCGTGAAGCCTGGCGGATCT 210 nt CTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCAACGCCTGGATG AGCTGGGTGCGCCAGGCCCCTGGAAAAGGACTCGAGTGGGTGGGACGGATC AAGAGCAAGACCGATGGCGGCACCACCGACTATGCCGCCCCTGTGAAGGGC CGGTTCACCATCAGCAGGGACGACAGCAAGAACACCCTGTACCTGCAGATG AACAGCCTGAAAACCGAGGACACCGCCGTGTACTACTGCACCACCCCCTAC GAGTGGTCTTGGTACGACTACTGGGGCCAGGGCACCCTCGTGACCGTGTCA TCT GCTAGC W98Y GAGGTGCAATTGGTGGAAAGCGGAGGCGGCCTCGTGAAGCCTGGCGGATCT 211 nt CTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCAACGCCTGGATG AGCTGGGTGCGCCAGGCCCCTGGAAAAGGACTCGAGTGGGTGGGACGGATC AAGAGCAAGACCGATGGCGGCACCACCGACTATGCCGCCCCTGTGAAGGGC CGGTTCACCATCAGCAGGGACGACAGCAAGAACACCCTGTACCTGCAGATG AACAGCCTGAAAACCGAGGACACCGCCGTGTACTACTGCACCACCCCCTGG GAGTACTCTTGGTACGACTACTGGGGCCAGGGCACCCTCGTGACCGTGTCA TCT GCTAGC 90D7 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCC 212 nt GTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATG CACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATT AACCCAAGCGGTGGCTCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTC ACGATGACCCGTGACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGC CTGCGTTCTGAAGATACTGCAGTGTACTACTGTGCACGCAACTACACTATC GTTGTTTCTCCGTTCGACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCT TCTGCTAGC 90C1 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCC 213 nt GTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATG CACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATT AACCCAAGCGGTGGCTCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTC ACGATGACCCGTGACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGC CTGCGTTCTGAAGATACTGCAGTGTACTACTGTGCACGCAACTACTTCATC GGTTCTGTTGCTATGGACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCT TCTGCTAGC 5E8 VH CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCC 214 nt GTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATG CACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATT AACCCAAGCGGTGGCTCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTC ACGATGACCCGTGACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGC CTGCGTTCTGAAGATACTGCAGTGTACTACTGTGCACGCGGTCTGACTTAC TCTATGGACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCTGCTAGC 5E8 VL GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAGGCGAA 215 nt CCGGCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCACTCCAACGGC TACAACTATCTCGATTGGTACCTGCAAAAACCGGGTCAGAGCCCTCAGCTG CTGATCTACCTGGGCTCTAACCGCGCTTCCGGTGTACCGGACCGTTTCAGC GGCTCTGGATCCGGCACCGATTTCACGTTGAAAATCAGCCGTGTTGAAGCA GAAGACGTGGGCGTTTATTACTGTATGCAGGCACTGCAGATTCCAAACACT TTTGGTCAAGGCACCAAGGTCGAAATTAAACGTACG 12A4 VH GAGGTGCAATTGTTGGAGTCTGGGGGAGGCTTGGTACAGCCTGGGGGGTCC 216 nt CTGAGACTCTCCTGTGCAGCCTCCGGATTCACCTTTAGCAGTTATGCCATG AGCTGGGTCCGCCAGGCTCCAGGGAAGGGGCTGGAGTGGGTCTCAGCTATT AGTGGTAGTGGTGGTAGCACATACTACGCAGACTCCGTGAAGGGCCGGTTC ACCATCTCCAGAGACAATTCCAAGAACACGCTGTATCTGCAGATGAACAGC CTGAGAGCCGAGGACACGGCCGTATATTACTGTGCGAAATACGCTTACGCT CTGGACTACTGGGGCCAAGGAACCCTGGTCACCGTCTCGAGTGCTAGC 12A4 VL GAAATCGTGTTAACGCAGTCTCCAGGCACCCTGTCTTTGTCTCCAGGGGAA 217 nt AGAGCCACCCTCTCTTGCAGGGCCAGTCAGAGTGTTAGCAGCAGCTACTTA GCCTGGTACCAGCAGAAACCTGGCCAGGCTCCCAGGCTCCTCATCTATGGA GCATCCAGCAGGGCCACTGGCATCCCAGACAGGTTCAGTGGCAGTGGATCC GGGACAGACTTCACTCTCACCATCAGCAGACTGGAGCCTGAAGATTTTGCA GTGTATTACTGTCAGCAGCATGGCAGCAGCAGCACGTTCGGCCAGGGGACC AAAGTGGAAATCAAACGTACG 7A3 VH CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCC 218 nt GTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATG CACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATT AACCCAAGCGGTGGCTCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTC ACGATGACCCGTGACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGC CTGCGTTCTGAAGATACTGCAGTGTACTACTGTGCACGCGGTGACTTCTCT GCTGGTCGTCTGATGGACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCT TCTGCTAGC 7A3 VL GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAGGCGAA 219 nt CCGGCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCACTCCAACGGC TACAACTATCTCGATTGGTACCTGCAAAAACCGGGTCAGAGCCCTCAGCTG CTGATCTACCTGGGCTCTAACCGCGCTTCCGGTGTACCGGACCGTTTCAGC GGCTCTGGATCCGGCACCGATTTCACGTTGAAAATCAGCCGTGTTGAAGCA GAAGACGTGGGCGTTTATTACTGTATGCAGGCACTGCAGACCCCACCAATT ACCTTTGGTCAAGGCACCAAGGTCGAAATTAAACGTACG 6E10 VH CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCC 220 nt GTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATG CACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATT AACCCAAGCGGTGGCTCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTC ACGATGACCCGTGACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGC CTGCGTTCTGAAGATACTGCAGTGTACTACTGTGCACGCGGTGACTACAAC GCTTTCGACTATTGGGGTCACGGCACCCTCGTAACGGTTTCTTCTGCTAGC 6E10 VL GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAGGCGAA 221 nt CCGGCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCACTCCAACGGC TACAACTATCTCGATTGGTACCTGCAAAAACCGGGTCAGAGCCCTCAGCTG CTGATCTACCTGGGCTCTAACCGCGCTTCCGGTGTACCGGACCGTTTCAGC GGCTCTGGATCCGGCACCGATTTCACGTTGAAAATCAGCCGTGTTGAAGCA GAAGACGTGGGCGTTTATTACTGTATGCAGGCATGGCATAGCCCAACTTTT GGTCAAGGCACCAAGGTCGAAATTAAACGTACG 12F9 VH CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCC 222 nt GTTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATG CACTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATT AACCCAAGCGGTGGCTCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTC ACGATGACCCGTGACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGC CTGCGTTCTGAAGATACTGCAGTGTACTACTGTGCACGCGGTGCTACTTAC ACTATGGACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCTGCTAGC 12F9 VL GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAGGCGAA 223 nt CCGGCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCACTCCAACGGC TACAACTATCTCGATTGGTACCTGCAAAAACCGGGTCAGAGCCCTCAGCTG CTGATCTACCTGGGCTCTAACCGCGCTTCCGGTGTACCGGACCGTTTCAGC GGCTCTGGATCCGGCACCGATTTCACGTTGAAAATCAGCCGTGTTGAAGCA GAAGACGTGGGCGTTTATTACTGTATGCAGGCACTGCAGACCCCAATTACT TTTGGTCAAGGCACCAAGGTCGAAATTAAACGTACG pETR11646 CAGGTGCAGCTGCAGCAGTCTGGCGCCGAGCTCGTGAAACCTGGCGCCTCC 224 Mov19 VH- GTGAAGATCAGCTGCAAGGCCAGCGGCTACAGCTTCACCGGCTACTTCATG CH1-Fchole AACTGGGTCAAGCAGAGCCACGGCAAGAGCCTGGAATGGATCGGCAGAATC PG/LALA CACCCCTACGACGGCGACACCTTCTACAACCAGAACTTCAAGGACAAGGCC ACCCTGACCGTGGACAAGAGCAGCAACACCGCCCACATGGAACTGCTGAGC CTGACCAGCGAGGACTTCGCCGTGTACTACTGCACCAGATACGACGGCAGC CGGGCCATGGATTATTGGGGCCAGGGCACCACCGTGACAGTGTCCAGCGCT AGCACCAAGGGCCCCTCCGTGTTCCCCCTGGCCCCCAGCAGCAAGAGCACC AGCGGCGGCACAGCCGCTCTGGGCTGCCTGGTCAAGGACTACTTCCCCGAG CCCGTGACCGTGTCCTGGAACAGCGGAGCCCTGACCTCCGGCGTGCACACC TTCCCCGCCGTGCTGCAGAGTTCTGGCCTGTATAGCCTGAGCAGCGTGGTC ACCGTGCCTTCTAGCAGCCTGGGCACCCAGACCTACATCTGCAACGTGAAC CACAAGCCCAGCAACACCAAGGTGGACAAGAAGGTGGAGCCCAAGAGCTGC GACAAAACTCACACATGCCCACCGTGCCCAGCACCTGAAGCTGCAGGGGGA CCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGACACCCTCATGATCTCC CGGACCCCTGAGGTCACATGCGTGGTGGTGGACGTGAGCCACGAAGACCCT GAGGTCAAGTTCAACTGGTACGTGGACGGCGTGGAGGTGCATAATGCCAAG ACAAAGCCGCGGGAGGAGCAGTACAACAGCACGTACCGTGTGGTCAGCGTC CTCACCGTCCTGCACCAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAG GTCTCCAACAAAGCCCTCGGCGCCCCCATCGAGAAAACCATCTCCAAAGCC AAAGGGCAGCCCCGAGAACCACAGGTGTGCACCCTGCCCCCATCCCGGGAT GAGCTGACCAAGAACCAGGTCAGCCTCTCGTGCGCAGTCAAAGGCTTCTAT CCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCCGGAGAACAAC TACAAGACCACGCCTCCCGTGCTGGACTCCGACGGCTCCTTCTTCCTCGTG AGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAGGGGAACGTCTTCTCA TGCTCCGTGATGCATGAGGCTCTGCACAACCACTACACGCAGAAGAGCCTC TCCCTGTCTCCGGGTAAA pETR11647 CAGGTGCAGCTGCAGCAGTCTGGCGCCGAGCTCGTGAAACCTGGCGCCTCC 225 Mov19 VH- GTGAAGATCAGCTGCAAGGCCAGCGGCTACAGCTTCACCGGCTACTTCATG CH1-CD3 AACTGGGTCAAGCAGAGCCACGGCAAGAGCCTGGAATGGATCGGCAGAATC VH-CL- CACCCCTACGACGGCGACACCTTCTACAACCAGAACTTCAAGGACAAGGCC Fcknob ACCCTGACCGTGGACAAGAGCAGCAACACCGCCCACATGGAACTGCTGAGC PG/LALA CTGACCAGCGAGGACTTCGCCGTGTACTACTGCACCAGATACGACGGCAGC CGGGCCATGGATTATTGGGGCCAGGGCACCACCGTGACAGTGTCCAGCGCT AGCACAAAGGGCCCCAGCGTGTTCCCTCTGGCCCCTAGCAGCAAGAGCACA TCTGGCGGAACAGCCGCCCTGGGCTGCCTCGTGAAGGACTACTTTCCCGAG CCTGTGACCGTGTCCTGGAACTCTGGCGCCCTGACAAGCGGCGTGCACACC TTTCCAGCCGTGCTGCAGAGCAGCGGCCTGTACTCTCTGAGCAGCGTGGTC ACCGTGCCTAGCAGCAGCCTGGGCACCCAGACCTACATCTGCAACGTGAAC CACAAGCCCAGCAACACCAAAGTGGACAAGAAGGTGGAGCCCAAGAGCTGT GATGGCGGAGGAGGGTCCGGAGGCGGAGGATCCGAAGTGCAGCTGGTGGAA AGCGGCGGAGGCCTGGTGCAGCCTAAGGGCTCTCTGAAGCTGAGCTGTGCC GCCAGCGGCTTCACCTTCAACACCTACGCCATGAACTGGGTGCGCCAGGCC CCTGGCAAAGGCCTGGAATGGGTGGCCCGGATCAGAAGCAAGTACAACAAT TACGCCACCTACTACGCCGACAGCGTGAAGGACCGGTTCACCATCAGCCGG GACGACAGCCAGAGCATCCTGTACCTGCAGATGAACAACCTGAAAACCGAG GACACCGCCATGTACTACTGCGTGCGGCACGGCAACTTCGGCAACAGCTAT GTGTCTTGGTTTGCCTACTGGGGCCAGGGCACCCTCGTGACAGTGTCTGCT GCTAGCGTGGCCGCTCCCTCCGTGTTTATCTTTCCCCCATCCGATGAACAG CTGAAAAGCGGCACCGCCTCCGTCGTGTGTCTGCTGAACAATTTTTACCCT AGGGAAGCTAAAGTGCAGTGGAAAGTGGATAACGCACTGCAGTCCGGCAAC TCCCAGGAATCTGTGACAGAACAGGACTCCAAGGACAGCACCTACTCCCTG TCCTCCACCCTGACACTGTCTAAGGCTGATTATGAGAAACACAAAGTCTAC GCCTGCGAAGTCACCCATCAGGGCCTGAGCTCGCCCGTCACAAAGAGCTTC AACAGGGGAGAGTGTGACAAGACCCACACCTGTCCCCCTTGTCCTGCCCCT GAAGCTGCTGGCGGCCCTTCTGTGTTCCTGTTCCCCCCAAAGCCCAAGGAC ACCCTGATGATCAGCCGGACCCCCGAAGTGACCTGCGTGGTGGTGGATGTG TCCCACGAGGACCCTGAAGTGAAGTTCAATTGGTACGTGGACGGCGTGGAA GTGCACAACGCCAAGACAAAGCCGCGGGAGGAGCAGTACAACAGCACGTAC CGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGACTGGCTGAATGGCAAG GAGTACAAGTGCAAGGTCTCCAACAAAGCCCTCGGCGCCCCCATCGAGAAA ACCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCACAGGTGTACACCCTG CCCCCATGCCGGGATGAGCTGACCAAGAACCAGGTCAGCCTGTGGTGCCTG GTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGG CAGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGC TCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAG GGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACCACTAC ACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA pETR11644 GACATCGAGCTGACCCAGAGCCCTGCCTCTCTGGCCGTGTCTCTGGGACAG 226 Mov19 LC AGAGCCATCATCAGCTGCAAGGCCAGCCAGAGCGTGTCCTTTGCCGGCACC TCTCTGATGCACTGGTATCACCAGAAGCCCGGCCAGCAGCCCAAGCTGCTG ATCTACAGAGCCAGCAACCTGGAAGCCGGCGTGCCCACAAGATTTTCCGGC AGCGGCAGCAAGACCGACTTCACCCTGAACATCCACCCCGTGGAAGAAGAG GACGCCGCCACCTACTACTGCCAGCAGAGCAGAGAGTACCCCTACACCTTC GGCGGAGGCACCAAGCTGGAAATCAAGCGTACGGTGGCTGCACCATCTGTC TTCATCTTCCCGCCATCTGATGAGCAGTTGAAATCTGGAACTGCCTCTGTT GTGTGCCTGCTGAATAACTTCTATCCCAGAGAGGCCAAAGTACAGTGGAAG GTGGATAACGCCCTCCAATCGGGTAACTCCCAGGAGAGTGTCACAGAGCAG GACAGCAAGGACAGCACCTACAGCCTCAGCAGCACCCTGACGCTGAGCAAA GCAGACTACGAGAAACACAAAGTCTACGCCTGCGAAGTCACCCATCAGGGC CTGAGCTCGCCCGTCACAAAGAGCTTCAACAGGGGAGAGTGT Variant Sequence Seq ID No 16D5 GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCGGTTCCC 261 VH_D52dE TGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAACGCGTGGATGAG CTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAGTGGGTTGGTCGTATCAAG TCTAAAACTGAGGGTGGCACCACGGATTACGCGGCTCCAGTTAAAGGTCGTT TTACCATTTCCCGCGACGATAGCAAAAACACTCTGTATCTGCAGATGAACTC TCTGAAAACTGAAGACACCGCAGTCTACTACTGTACTACCCCGTGGGAATGG TCTTGGTACGATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCC 16D5 GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCGGTTCCC 262 VH_D52dQ TGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAACGCGTGGATGAG CTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAGTGGGTTGGTCGTATCAAG TCTAAAACTCAGGGTGGCACCACGGATTACGCGGCTCCAGTTAAAGGTCGTT TTACCATTTCCCGCGACGATAGCAAAAACACTCTGTATCTGCAGATGAACTC TCTGAAAACTGAAGACACCGCAGTCTACTACTGTACTACCCCGTGGGAATGG TCTTGGTACGATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCC CD3_VH GAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGGTGCAGCCTGGCGGATCTC 263 N100A TGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCACCTACGCCATGAA CTGGGTGCGCCAGGCCCCTGGCAAAGGCCTGGAATGGGTGTCCCGGATCAGA AGCAAGTACAACAACTACGCCACCTACTACGCCGACAGCGTGAAGGGCCGGT TCACCATCAGCCGGGACGACAGCAAGAACACCCTGTACCTGCAGATGAACAG CCTGCGGGCCGAGGACACCGCCGTGTACTATTGTGTGCGGCACGGCAACTTC GGCGCCAGCTATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCACCCTCGTGA CCGTGTCAAGC CD3_VH GAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGGTGCAGCCTGGCGGATCTC 264 S100aA TGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTCAGCACCTACGCCATGAA CTGGGTGCGCCAGGCCCCTGGCAAAGGCCTGGAATGGGTGTCCCGGATCAGA AGCAAGTACAACAACTACGCCACCTACTACGCCGACAGCGTGAAGGGCCGGT TCACCATCAGCCGGGACGACAGCAAGAACACCCTGTACCTGCAGATGAACAG CCTGCGGGCCGAGGACACCGCCGTGTACTATTGTGTGCGGCACGGCAACTTC GGCAACGCCTATGTGTCTTGGTTTGCCTACTGGGGCCAGGGCACCCTCGTGA CCGTGTCAAGC 16D5 GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCGGTTCCC 265 [VHCH1]- TGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAACGCGTGGATGAG CD3[VHCH1- CTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAGTGGGTTGGTCGTATCAAG N100A]- TCTAAAACTGACGGTGGCACCACGGATTACGCGGCTCCAGTTAAAGGTCGTT Fcknob_PGLALA TTACCATTTCCCGCGACGATAGCAAAAACACTCTGTATCTGCAGATGAACTC TCTGAAAACTGAAGACACCGCAGTCTACTACTGTACTACCCCGTGGGAATGG TCTTGGTACGATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCCGCTA GCACAAAGGGCCCTAGCGTGTTCCCTCTGGCCCCCAGCAGCAAGAGCACAAG CGGCGGAACAGCCGCCCTGGGCTGCCTCGTGAAGGACTACTTCCCCGAGCCC GTGACAGTGTCTTGGAACAGCGGAGCCCTGACAAGCGGCGTGCACACTTTCC CTGCCGTGCTGCAGAGCAGCGGCCTGTACTCCCTGAGCAGCGTGGTCACCGT GCCTAGCAGCAGCCTGGGCACCCAGACCTACATCTGCAACGTGAACCACAAG CCCAGCAACACCAAAGTGGACAAGAAGGTGGAGCCCAAGAGCTGTGATGGCG GAGGAGGGTCCGGAGGCGGAGGATCCGAGGTGCAGCTGCTGGAATCTGGCGG CGGACTGGTGCAGCCTGGCGGATCTCTGAGACTGAGCTGTGCCGCCAGCGGC TTCACCTTCAGCACCTACGCCATGAACTGGGTGCGCCAGGCCCCTGGCAAAG GCCTGGAATGGGTGTCCCGGATCAGAAGCAAGTACAACAACTACGCCACCTA CTACGCCGACAGCGTGAAGGGCCGGTTCACCATCAGCCGGGACGACAGCAAG AACACCCTGTACCTGCAGATGAACAGCCTGCGGGCCGAGGACACCGCCGTGT ACTATTGTGTGCGGCACGGCAACTTCGGCGCCAGCTATGTGTCTTGGTTTGC CTACTGGGGCCAGGGCACCCTCGTGACCGTGTCAAGCGCTAGTACCAAGGGC CCCAGCGTGTTCCCCCTGGCACCCAGCAGCAAGAGCACATCTGGCGGAACAG CCGCTCTGGGCTGTCTGGTGAAAGACTACTTCCCCGAGCCCGTGACCGTGTC TTGGAACTCTGGCGCCCTGACCAGCGGCGTGCACACCTTTCCAGCCGTGCTG CAGAGCAGCGGCCTGTACTCCCTGTCCTCCGTGGTCACCGTGCCCTCTAGCT CCCTGGGAACACAGACATATATCTGTAATGTCAATCACAAGCCTTCCAACAC CAAAGTCGATAAGAAAGTCGAGCCCAAGAGCTGCGACAAAACTCACACATGC CCACCGTGCCCAGCACCTGAAGCTGCAGGGGGACCGTCAGTCTTCCTCTTCC CCCCAAAACCCAAGGACACCCTCATGATCTCCCGGACCCCTGAGGTCACATG CGTGGTGGTGGACGTGAGCCACGAAGACCCTGAGGTCAAGTTCAACTGGTAC GTGGACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCAGT ACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGACTG GCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCCTCGGCGCC CCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCACAGG TGTACACCCTGCCCCCATGCCGGGATGAGCTGACCAAGAACCAGGTCAGCCT GTGGTGCCTGGTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGGGAG AGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACT CCGACGGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTG GCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAAC CACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA 16D5- GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCGGTTCCC 266 Fchole- TGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAACGCGTGGATGAG PGLALA CTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAGTGGGTTGGTCGTATCAAG TCTAAAACTGACGGTGGCACCACGGATTACGCGGCTCCAGTTAAAGGTCGTT TTACCATTTCCCGCGACGATAGCAAAAACACTCTGTATCTGCAGATGAACTC TCTGAAAACTGAAGACACCGCAGTCTACTACTGTACTACCCCGTGGGAATGG TCTTGGTACGATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCCGCTA GCACCAAGGGCCCCTCCGTGTTCCCCCTGGCCCCCAGCAGCAAGAGCACCAG CGGCGGCACAGCCGCTCTGGGCTGCCTGGTCAAGGACTACTTCCCCGAGCCC GTGACCGTGTCCTGGAACAGCGGAGCCCTGACCTCCGGCGTGCACACCTTCC CCGCCGTGCTGCAGAGTTCTGGCCTGTATAGCCTGAGCAGCGTGGTCACCGT GCCTTCTAGCAGCCTGGGCACCCAGACCTACATCTGCAACGTGAACCACAAG CCCAGCAACACCAAGGTGGACAAGAAGGTGGAGCCCAAGAGCTGCGACAAAA CTCACACATGCCCACCGTGCCCAGCACCTGAAGCTGCAGGGGGACCGTCAGT CTTCCTCTTCCCCCCAAAACCCAAGGACACCCTCATGATCTCCCGGACCCCT GAGGTCACATGCGTGGTGGTGGACGTGAGCCACGAAGACCCTGAGGTCAAGT TCAACTGGTACGTGGACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCG GGAGGAGCAGTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTG CACCAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAG CCCTCGGCGCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCCG AGAACCACAGGTGTGCACCCTGCCCCCATCCCGGGATGAGCTGACCAAGAAC CAGGTCAGCCTCTCGTGCGCAGTCAAAGGCTTCTATCCCAGCGACATCGCCG TGGAGTGGGAGAGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCC CGTGCTGGACTCCGACGGCTCCTTCTTCCTCGTGAGCAAGCTCACCGTGGAC AAGAGCAGGTGGCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGG CTCTGCACAACCGCTTCACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA CD3-CLC CAGGCCGTCGTGACCCAGGAACCCAGCCTGACAGTGTCTCCTGGCGGCACCG 267 TGACCCTGACATGTGGCAGTTCTACAGGCGCCGTGACCACCAGCAACTACGC CAACTGGGTGCAGGAAAAGCCCGGCCAGGCCTTCAGAGGACTGATCGGCGGC ACCAACAAGAGAGCCCCTGGCACCCCTGCCAGATTCAGCGGATCTCTGCTGG GAGGAAAGGCCGCCCTGACACTGTCTGGCGCCCAGCCAGAAGATGAGGCCGA GTACTACTGCGCCCTGTGGTACAGCAACCTGTGGGTGTTCGGCGGAGGCACC AAGCTGACAGTCCTAGGTCAACCCAAGGCTGCCCCCAGCGTGACCCTGTTCC CCCCCAGCAGCGAGGAACTGCAGGCCAACAAGGCCACCCTGGTCTGCCTGAT CAGCGACTTCTACCCAGGCGCCGTGACCGTGGCCTGGAAGGCCGACAGCAGC CCCGTGAAGGCCGGCGTGGAGACCACCACCCCCAGCAAGCAGAGCAACAACA AGTACGCCGCCAGCAGCTACCTGAGCCTGACCCCCGAGCAGTGGAAGAGCCA CAGGTCCTACAGCTGCCAGGTGACCCACGAGGGCAGCACCGTGGAGAAAACC GTGGCCCCCACCGAGTGCAGC 16D5 GAGGTGCAATTGGTTGAATCTGGTGGTGGTCTGGTAAAACCGGGCGGTTCCC 268 [VHCH1]- TGCGTCTGAGCTGCGCGGCTTCCGGATTCACCTTCTCCAACGCGTGGATGAG CD3[VHCH1- CTGGGTTCGCCAGGCCCCGGGCAAAGGCCTCGAGTGGGTTGGTCGTATCAAG S100aA]- TCTAAAACTGACGGTGGCACCACGGATTACGCGGCTCCAGTTAAAGGTCGTT Fcknob_PGLALA TTACCATTTCCCGCGACGATAGCAAAAACACTCTGTATCTGCAGATGAACTC TCTGAAAACTGAAGACACCGCAGTCTACTACTGTACTACCCCGTGGGAATGG TCTTGGTACGATTATTGGGGCCAGGGCACGCTGGTTACGGTGTCTTCCGCTA GCACAAAGGGCCCTAGCGTGTTCCCTCTGGCCCCCAGCAGCAAGAGCACAAG CGGCGGAACAGCCGCCCTGGGCTGCCTCGTGAAGGACTACTTCCCCGAGCCC GTGACAGTGTCTTGGAACAGCGGAGCCCTGACAAGCGGCGTGCACACTTTCC CTGCCGTGCTGCAGAGCAGCGGCCTGTACTCCCTGAGCAGCGTGGTCACCGT GCCTAGCAGCAGCCTGGGCACCCAGACCTACATCTGCAACGTGAACCACAAG CCCAGCAACACCAAAGTGGACAAGAAGGTGGAGCCCAAGAGCTGTGATGGCG GAGGAGGGTCCGGAGGCGGAGGATCCGAGGTGCAGCTGCTGGAATCTGGCGG CGGACTGGTGCAGCCTGGCGGATCTCTGAGACTGAGCTGTGCCGCCAGCGGC TTCACCTTCAGCACCTACGCCATGAACTGGGTGCGCCAGGCCCCTGGCAAAG GCCTGGAATGGGTGTCCCGGATCAGAAGCAAGTACAACAACTACGCCACCTA CTACGCCGACAGCGTGAAGGGCCGGTTCACCATCAGCCGGGACGACAGCAAG AACACCCTGTACCTGCAGATGAACAGCCTGCGGGCCGAGGACACCGCCGTGT ACTATTGTGTGCGGCACGGCAACTTCGGCAACGCCTATGTGTCTTGGTTTGC CTACTGGGGCCAGGGCACCCTCGTGACCGTGTCAAGCGCTAGTACCAAGGGC CCCAGCGTGTTCCCCCTGGCACCCAGCAGCAAGAGCACATCTGGCGGAACAG CCGCTCTGGGCTGTCTGGTGAAAGACTACTTCCCCGAGCCCGTGACCGTGTC TTGGAACTCTGGCGCCCTGACCAGCGGCGTGCACACCTTTCCAGCCGTGCTG CAGAGCAGCGGCCTGTACTCCCTGTCCTCCGTGGTCACCGTGCCCTCTAGCT CCCTGGGAACACAGACATATATCTGTAATGTCAATCACAAGCCTTCCAACAC CAAAGTCGATAAGAAAGTCGAGCCCAAGAGCTGCGACAAAACTCACACATGC CCACCGTGCCCAGCACCTGAAGCTGCAGGGGGACCGTCAGTCTTCCTCTTCC CCCCAAAACCCAAGGACACCCTCATGATCTCCCGGACCCCTGAGGTCACATG CGTGGTGGTGGACGTGAGCCACGAAGACCCTGAGGTCAAGTTCAACTGGTAC GTGGACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCAGT ACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGACTG GCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCCTCGGCGCC CCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCACAGG TGTACACCCTGCCCCCATGCCGGGATGAGCTGACCAAGAACCAGGTCAGCCT GTGGTGCCTGGTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGGGAG AGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACT CCGACGGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTG GCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAAC CACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA 9D11 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCCG 269 [VHCH1]- TTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATGCA CD3[VHCL- CTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATTAAC N100A]- CCAAGCGGTGGCCCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTCACGA Fcknob_PGLALA TGACCCGTGACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGCCTGCG TTCTGAAGATACTGCAGTGTACTACTGTGCACGCGGTGACTTCGCTTGGCTG GACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCTGCTAGCACAAAGG GCCCCAGCGTGTTCCCTCTGGCCCCTAGCAGCAAGAGCACATCTGGCGGAAC AGCCGCCCTGGGCTGCCTCGTGAAGGACTACTTTCCCGAGCCTGTGACCGTG TCCTGGAACTCTGGCGCCCTGACAAGCGGCGTGCACACCTTTCCAGCCGTGC TGCAGAGCAGCGGCCTGTACTCTCTGAGCAGCGTGGTCACCGTGCCTAGCAG CAGCCTGGGCACCCAGACCTACATCTGCAACGTGAACCACAAGCCCAGCAAC ACCAAAGTGGACAAGAAGGTGGAGCCCAAGAGCTGTGATGGCGGAGGAGGGT CCGGAGGCGGAGGATCCGAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGGT GCAGCCTGGCGGATCTCTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTC AGCACCTACGCCATGAACTGGGTGCGCCAGGCCCCTGGCAAAGGCCTGGAAT GGGTGTCCCGGATCAGAAGCAAGTACAACAACTACGCCACCTACTACGCCGA CAGCGTGAAGGGCCGGTTCACCATCAGCCGGGACGACAGCAAGAACACCCTG TACCTGCAGATGAACAGCCTGCGGGCCGAGGACACCGCCGTGTACTATTGTG TGCGGCACGGCAACTTCGGCGCCAGCTATGTGTCTTGGTTTGCCTACTGGGG CCAGGGCACCCTCGTGACCGTGTCAAGCGCTAGTGTGGCCGCTCCCTCCGTG TTTATCTTTCCCCCATCCGATGAACAGCTGAAAAGCGGCACCGCCTCCGTCG TGTGTCTGCTGAACAATTTTTACCCTAGGGAAGCTAAAGTGCAGTGGAAAGT GGATAACGCACTGCAGTCCGGCAACTCCCAGGAATCTGTGACAGAACAGGAC TCCAAGGACAGCACCTACTCCCTGTCCTCCACCCTGACACTGTCTAAGGCTG ATTATGAGAAACACAAAGTCTACGCCTGCGAAGTCACCCATCAGGGCCTGAG CTCGCCCGTCACAAAGAGCTTCAACAGGGGAGAGTGTGACAAGACCCACACC TGTCCCCCTTGTCCTGCCCCTGAAGCTGCTGGCGGCCCTTCTGTGTTCCTGT TCCCCCCAAAGCCCAAGGACACCCTGATGATCAGCCGGACCCCCGAAGTGAC CTGCGTGGTGGTGGATGTGTCCCACGAGGACCCTGAAGTGAAGTTCAATTGG TACGTGGACGGCGTGGAAGTGCACAACGCCAAGACAAAGCCGCGGGAGGAGC AGTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGA CTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCCTCGGC GCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCAC AGGTGTACACCCTGCCCCCATGCCGGGATGAGCTGACCAAGAACCAGGTCAG CCTGTGGTGCCTGGTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGG GAGAGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGG ACTCCGACGGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAG GTGGCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCAC AACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA 9D11- CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCCG 270 Fchole TTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATGCA CTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATTAAC CCAAGCGGTGGCCCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTCACGA TGACCCGTGACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGCCTGCG TTCTGAAGATACTGCAGTGTACTACTGTGCACGCGGTGACTTCGCTTGGCTG GACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCTGCTAGCACCAAGG GCCCCTCCGTGTTCCCCCTGGCCCCCAGCAGCAAGAGCACCAGCGGCGGCAC AGCCGCTCTGGGCTGCCTGGTCAAGGACTACTTCCCCGAGCCCGTGACCGTG TCCTGGAACAGCGGAGCCCTGACCTCCGGCGTGCACACCTTCCCCGCCGTGC TGCAGAGTTCTGGCCTGTATAGCCTGAGCAGCGTGGTCACCGTGCCTTCTAG CAGCCTGGGCACCCAGACCTACATCTGCAACGTGAACCACAAGCCCAGCAAC ACCAAGGTGGACAAGAAGGTGGAGCCCAAGAGCTGCGACAAAACTCACACAT GCCCACCGTGCCCAGCACCTGAAGCTGCAGGGGGACCGTCAGTCTTCCTCTT CCCCCCAAAACCCAAGGACACCCTCATGATCTCCCGGACCCCTGAGGTCACA TGCGTGGTGGTGGACGTGAGCCACGAAGACCCTGAGGTCAAGTTCAACTGGT ACGTGGACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCA GTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGAC TGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCCTCGGCG CCCCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCACA GGTGTGCACCCTGCCCCCATCCCGGGATGAGCTGACCAAGAACCAGGTCAGC CTCTCGTGCGCAGTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGGG AGAGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGGA CTCCGACGGCTCCTTCTTCCTCGTGAGCAAGCTCACCGTGGACAAGAGCAGG TGGCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACA ACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA 9D11_LC GATATTGTTATGACTCAATCTCCACTGTCTCTGCCGGTGACTCCAGGCGAAC 271 [N95Q] CGGCGAGCATTTCTTGCCGTTCCAGCCAGTCTCTGCTGCACTCCAACGGCTA CAACTATCTCGATTGGTACCTGCAAAAACCGGGTCAGAGCCCTCAGCTGCTG ATCTACCTGGGCTCTAACCGCGCTTCCGGTGTACCGGACCGTTTCAGCGGCT CTGGATCCGGCACCGATTTCACGTTGAAAATCAGCCGTGTTGAAGCAGAAGA CGTGGGCGTTTATTACTGTATGCAGGCAAGCATTATGCAGCGGACTTTTGGT CAAGGCACCAAGGTCGAAATTAAACGTACGGTGGCTGCACCATCTGTCTTCA TCTTCCCGCCATCTGATGAGCAGTTGAAATCTGGAACTGCCTCTGTTGTGTG CCTGCTGAATAACTTCTATCCCAGAGAGGCCAAAGTACAGTGGAAGGTGGAT AACGCCCTCCAATCGGGTAACTCCCAGGAGAGTGTCACAGAGCAGGACAGCA AGGACAGCACCTACAGCCTCAGCAGCACCCTGACGCTGAGCAAAGCAGACTA CGAGAAACACAAAGTCTACGCCTGCGAAGTCACCCATCAGGGCCTGAGCTCG CCCGTCACAAAGAGCTTCAACAGGGGAGAGTGT CD3_VLCH1 CAGGCCGTCGTGACCCAGGAACCCAGCCTGACAGTGTCTCCTGGCGGCACCG 272 TGACCCTGACATGTGGCAGTTCTACAGGCGCCGTGACCACCAGCAACTACGC CAACTGGGTGCAGGAAAAGCCCGGCCAGGCCTTCAGAGGACTGATCGGCGGC ACCAACAAGAGAGCCCCTGGCACCCCTGCCAGATTCAGCGGATCTCTGCTGG GAGGAAAGGCCGCCCTGACACTGTCTGGCGCCCAGCCAGAAGATGAGGCCGA GTACTACTGCGCCCTGTGGTACAGCAACCTGTGGGTGTTCGGCGGAGGCACC AAGCTGACAGTGCTGAGCAGCGCTTCCACCAAAGGCCCTTCCGTGTTTCCTC TGGCTCCTAGCTCCAAGTCCACCTCTGGAGGCACCGCTGCTCTCGGATGCCT CGTGAAGGATTATTTTCCTGAGCCTGTGACAGTGTCCTGGAATAGCGGAGCA CTGACCTCTGGAGTGCATACTTTCCCCGCTGTGCTGCAGTCCTCTGGACTGT ACAGCCTGAGCAGCGTGGTGACAGTGCCCAGCAGCAGCCTGGGCACCCAGAC CTACATCTGCAACGTGAACCACAAGCCCAGCAACACCAAGGTGGACAAGAAG GTGGAACCCAAGTCTTGT 9D11 CAGGTGCAATTGGTTCAATCTGGTGCTGAAGTAAAAAAACCGGGCGCTTCCG 273 [VHCH1]- TTAAAGTGAGCTGCAAAGCATCCGGATACACCTTCACTTCCTATTACATGCA CD3[VHCH1- CTGGGTTCGTCAAGCCCCGGGCCAGGGTCTGGAATGGATGGGCATCATTAAC S100aA]- CCAAGCGGTGGCCCTACCTCCTACGCGCAGAAATTCCAGGGTCGCGTCACGA Fcknob_PGLALA TGACCCGTGACACTAGCACCTCTACCGTTTATATGGAGCTGTCCAGCCTGCG TTCTGAAGATACTGCAGTGTACTACTGTGCACGCGGTGACTTCGCTTGGCTG GACTATTGGGGTCAAGGCACCCTCGTAACGGTTTCTTCTGCTAGCACAAAGG GCCCCAGCGTGTTCCCTCTGGCCCCTAGCAGCAAGAGCACATCTGGCGGAAC AGCCGCCCTGGGCTGCCTCGTGAAGGACTACTTTCCCGAGCCTGTGACCGTG TCCTGGAACTCTGGCGCCCTGACAAGCGGCGTGCACACCTTTCCAGCCGTGC TGCAGAGCAGCGGCCTGTACTCTCTGAGCAGCGTGGTCACCGTGCCTAGCAG CAGCCTGGGCACCCAGACCTACATCTGCAACGTGAACCACAAGCCCAGCAAC ACCAAAGTGGACAAGAAGGTGGAGCCCAAGAGCTGTGATGGCGGAGGAGGGT CCGGAGGCGGAGGATCCGAGGTGCAGCTGCTGGAATCTGGCGGCGGACTGGT GCAGCCTGGCGGATCTCTGAGACTGAGCTGTGCCGCCAGCGGCTTCACCTTC AGCACCTACGCCATGAACTGGGTGCGCCAGGCCCCTGGCAAAGGCCTGGAAT GGGTGTCCCGGATCAGAAGCAAGTACAACAACTACGCCACCTACTACGCCGA CAGCGTGAAGGGCCGGTTCACCATCAGCCGGGACGACAGCAAGAACACCCTG TACCTGCAGATGAACAGCCTGCGGGCCGAGGACACCGCCGTGTACTATTGTG TGCGGCACGGCAACTTCGGCAACGCCTATGTGTCTTGGTTTGCCTACTGGGG CCAGGGCACCCTCGTGACCGTGTCAAGCGCTAGTGTGGCCGCTCCCTCCGTG TTTATCTTTCCCCCATCCGATGAACAGCTGAAAAGCGGCACCGCCTCCGTCG TGTGTCTGCTGAACAATTTTTACCCTAGGGAAGCTAAAGTGCAGTGGAAAGT GGATAACGCACTGCAGTCCGGCAACTCCCAGGAATCTGTGACAGAACAGGAC TCCAAGGACAGCACCTACTCCCTGTCCTCCACCCTGACACTGTCTAAGGCTG ATTATGAGAAACACAAAGTCTACGCCTGCGAAGTCACCCATCAGGGCCTGAG CTCGCCCGTCACAAAGAGCTTCAACAGGGGAGAGTGTGACAAGACCCACACC TGTCCCCCTTGTCCTGCCCCTGAAGCTGCTGGCGGCCCTTCTGTGTTCCTGT TCCCCCCAAAGCCCAAGGACACCCTGATGATCAGCCGGACCCCCGAAGTGAC CTGCGTGGTGGTGGATGTGTCCCACGAGGACCCTGAAGTGAAGTTCAATTGG TACGTGGACGGCGTGGAAGTGCACAACGCCAAGACAAAGCCGCGGGAGGAGC AGTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGA CTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCCTCGGC GCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCAC AGGTGTACACCCTGCCCCCATGCCGGGATGAGCTGACCAAGAACCAGGTCAG CCTGTGGTGCCTGGTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGG GAGAGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGG ACTCCGACGGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAG GTGGCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCAC AACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA

9) Additonal Common Light Chain Protein and Nucleotide Sequences SEQ ID CH2527 (VL_7-46(13)/VH_3-23(12) NO. Heavy chain EVQLLESGGGLVQPGGSLRLSCAASGFTFSTYAMNWVRQ 123 “CH2527 (VH_3- APGKGLEWVSRIRSKYNNYATYYADSVKGRFTISRDDSK 23(12))” NTLYLQMNSLRAEDTAVYYCVRHGNFGNSYVSWFAYWGQ GTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKD YFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVT VPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTC PPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVD VSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVS VLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQP REPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWE SNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGN VFSCSVMHEALHNHYTQKSLSLSPGK Light chain QTVVTQEPSLTVSPGGTVTLTCGSSTGAVTTSNYANWVQ 124 “CH2527 (VL_7- EKPGQAFRGLIGGTNKRAPGTPARFSGSLLGGKAALTLS 46(13))” GAQPEDEAEYYCALWYSNLWVFGGGTKLTVLGQPKAAPS VTLFPPSSEELQANKATLVCLISDFYPGAVTVAWKADSS PVKAGVETTTPSKQSNNKYAASSYLSLTPEQWKSHRSYS CQVTHEGSTVEKTVAPTECS VH EVQLLESGGGLVQPGGSLRLSCAASGFTFSTYAMNWVRQ 36 “CH2527 (VH_3- APGKGLEWVSRIRSKYNNYATYYADSVKGRFTISRDDSK 23(12))” NTLYLQMNSLRAEDTAVYYCVRHGNFGNSYVSWFAYWGQ GTLVTVSS VH CDR H1 TYAMN 37 “CH2527 (VH_3- 23(12))” VH CDR H2 RIRSKYNNYATYYADSVKG 38 “CH2527 (VH_3- 23(12))” VH CDR H3 HGNFGNSYVSWFAY 39 “CH2527 (VH_3- 23(12))” VL QTVVTQEPSLTVSPGGTVTLTCGSSTGAVTTSNYANWVQ 125 “CH2527 (VL_7- EKPGQAFRGLIGGTNKRAPGTPARFSGSLLGGKAALTLS 46(13))” GAQPEDEAEYYCALWYSNLWVFGGGTKLTVL VL CDR L1 GSSTGAVTTSNYAN 32 “CH2527 (VL_7- 46(13))” VL CDR L2 GTNKRAP 33 “CH2527 (VL_7- 46(13))” VL CDR L3 ALWYSNLWV 34 “CH2527 (VL_7- 46(13))”

10) DNA sequences humanized CD3CH2527 (CDR/VH/VL) CH2527 (VL_7-46(13)/VH_3-23(12) SEQ ID NO. Heavy chain ATGGGATGGAGCTGTATCATCCTCTTCTTGGTAGC 126 “CH2527 (VH_3- AACAGCTACCGGTGTGCATTCCGAGGTGCAGCTGC 23(12))” TGGAATCTGGCGGCGGACTGGTGCAGCCTGGCGGA TCTCTGAGACTGAGCTGTGCCGCCAGCGGCTTCAC CTTCAGCACCTACGCCATGAACTGGGTGCGCCAGG CCCCTGGCAAAGGCCTGGAATGGGTGTCCCGGATC AGAAGCAAGTACAACAACTACGCCACCTACTACGC CGACAGCGTGAAGGGCCGGTTCACCATCAGCCGGG ACGACAGCAAGAACACCCTGTACCTGCAGATGAAC AGCCTGCGGGCCGAGGACACCGCCGTGTACTATTG TGTGCGGCACGGCAACTTCGGCAACAGCTATGTGT CTTGGTTTGCCTACTGGGGCCAGGGCACCCTCGTG ACCGTGTCATCTGCTAGCACCAAGGGCCCATCGGT CTTCCCCCTGGCACCCTCCTCCAAGAGCACCTCTG GGGGCACAGCGGCCCTGGGCTGCCTGGTCAAGGAC TACTTCCCCGAACCGGTGACGGTGTCGTGGAACTC AGGCGCCCTGACCAGCGGCGTGCACACCTTCCCGG CTGTCCTACAGTCCTCAGGACTCTACTCCCTCAGC AGCGTGGTGACCGTGCCCTCCAGCAGCTTGGGCAC CCAGACCTACATCTGCAACGTGAATCACAAGCCCA GCAACACCAAGGTGGACAAGAAAGTTGAGCCCAAA TCTTGTGACAAAACTCACACATGCCCACCGTGCCC AGCACCTGAACTCCTGGGGGGACCGTCAGTCTTCC TCTTCCCCCCAAAACCCAAGGACACCCTCATGATC TCCCGGACCCCTGAGGTCACATGCGTGGTGGTGGA CGTGAGCCACGAAGACCCTGAGGTCAAGTTCAACT GGTACGTGGACGGCGTGGAGGTGCATAATGCCAAG ACAAAGCCGCGGGAGGAGCAGTACAACAGCACGTA CCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGG ACTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTC TCCAACAAAGCCCTCCCAGCCCCCATCGAGAAAAC CATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCAC AGGTGTACACCCTGCCCCCATCCCGGGATGAGCTG ACCAAGAACCAGGTCAGCCTGACCTGCCTGGTCAA AGGCTTCTATCCCAGCGACATCGCCGTGGAGTGGG AGAGCAATGGGCAGCCGGAGAACAACTACAAGACC ACGCCTCCCGTGCTGGACTCCGACGGCTCCTTCTT CCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGT GGCAGCAGGGGAACGTCTTCTCATGCTCCGTGATG CATGAGGCTCTGCACAACCACTACACGCAGAAGAG CCTCTCCCTGTCTCCGGGTAAATGA Light chain ATGGGATGGAGCTGTATCATCCTCTTCTTGGTAGC 127 “CH2527 (VL_7- AACAGCTACCGGTGTGCATTCTCAGGCCGTCGTGA 46(13))” CCCAGGAACCCAGCCTGACAGTGTCTCCTGGCGGC ACCGTGACCCTGACATGTGGCAGTTCTACAGGCGC CGTGACCACCAGCAACTACGCCAACTGGGTGCAGG AAAAGCCCGGCCAGGCCTTCAGAGGACTGATCGGC GGCACCAACAAGAGAGCCCCTGGCACCCCTGCCAG ATTCAGCGGATCTCTGCTGGGAGGAAAGGCCGCCC TGACACTGTCTGGCGCCCAGCCAGAAGATGAGGCC GAGTACTACTGCGCCCTGTGGTACAGCAACCTGTG GGTGTTCGGCGGAGGCACCAAGCTGACAGTCCTAG GTCAACCCAAGGCTGCCCCCAGCGTGACCCTGTTC CCCCCCAGCAGCGAGGAACTGCAGGCCAACAAGGC CACCCTGGTCTGCCTGATCAGCGACTTCTACCCAG GCGCCGTGACCGTGGCCTGGAAGGCCGACAGCAGC CCCGTGAAGGCCGGCGTGGAGACCACCACCCCCAG CAAGCAGAGCAACAACAAGTACGCCGCCAGCAGCT ACCTGAGCCTGACCCCCGAGCAGTGGAAGAGCCAC AGGTCCTACAGCTGCCAGGTGACCCACGAGGGCAG CACCGTGGAGAAAACCGTGGCCCCCACCGAGTGCA GCTGA VH ATGGGATGGAGCTGTATCATCCTCTTCTTGGTAGC 128 “CH2527 (VH_3- AACAGCTACCGGTGTGCATTCCGAGGTGCAGCTGC 23(12))” TGGAATCTGGCGGCGGACTGGTGCAGCCTGGCGGA TCTCTGAGACTGAGCTGTGCCGCCAGCGGCTTCAC CTTCAGCACCTACGCCATGAACTGGGTGCGCCAGG CCCCTGGCAAAGGCCTGGAATGGGTGTCCCGGATC AGAAGCAAGTACAACAACTACGCCACCTACTACGC CGACAGCGTGAAGGGCCGGTTCACCATCAGCCGGG ACGACAGCAAGAACACCCTGTACCTGCAGATGAAC AGCCTGCGGGCCGAGGACACCGCCGTGTACTATTG TGTGCGGCACGGCAACTTCGGCAACAGCTATGTGT CTTGGTTTGCCTACTGGGGCCAGGGCACCCTCGTG ACCGTGTCATCT VH CDR H1 ACCTACGCCATGAAC 129 “CH2527 (VH_3- 23(12))” VH CDR H2 CGGATCAGAAGCAAGTACAACAACTACGCCACCTA 130 “CH2527 (VH_3- CTACGCCGACAGCGTGAAGGGC 23(12))” VH CDR H3 CACGGCAACTTCGGCAACAGCTATGTGTCTTGGTT 131 “CH2527 (VH_3- TGCCTAC 23(12))” VL ATGGGATGGAGCTGTATCATCCTCTTCTTGGTAGC 132 “CH2527 (VL_7- AACAGCTACCGGTGTGCATTCTCAGGCCGTCGTGA 46(13))” CCCAGGAACCCAGCCTGACAGTGTCTCCTGGCGGC ACCGTGACCCTGACATGTGGCAGTTCTACAGGCGC CGTGACCACCAGCAACTACGCCAACTGGGTGCAGG AAAAGCCCGGCCAGGCCTTCAGAGGACTGATCGGC GGCACCAACAAGAGAGCCCCTGGCACCCCTGCCAG ATTCAGCGGATCTCTGCTGGGAGGAAAGGCCGCCC TGACACTGTCTGGCGCCCAGCCAGAAGATGAGGCC GAGTACTACTGCGCCCTGTGGTACAGCAACCTGTG GGTGTTCGGCGGAGGCACCAAGCTGACAGTCCTA VL CDR L1 GGCAGTTCTACAGGCGCCGTGACCACCAGCAACTA 133 “CH2527 (VL_7- CGCCAAC 46(13))” VL CDR L2 GGCACCAACAAGAGAGCCCCT 134 “CH2527 (VL_7- 46(13))” VL CDR L3 GCCCTGTGGTACAGCAACCTGTGGGTG 135 “CH2527 (VL_7- 46(13))”

Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, the descriptions and examples should not be construed as limiting the scope of the invention. The disclosures of all patent and scientific literature cited herein are expressly incorporated in their entirety by reference.

Claims

1. A T cell activating bispecific antigen binding molecule comprising a first and a second antigen binding moiety, wherein the first antigen binding moiety comprises a first light chain and wherein the first antigen binding moiety is capable of specific binding to an activating T cell antigen, wherein the second antigen binding moiety comprises a second light chain and wherein the second antigen binding moiety is capable of specific binding to a target cell antigen, wherein the amino acid sequence of the first and the second light chain is identical.

2. The T cell activating bispecific antigen binding molecule of claim 1, wherein the first antigen binding moiety is a Fab.

3. The T cell activating bispecific antigen binding molecule of claim 1 or 2, wherein the second antigen binding moiety is a Fab.

4. The T cell activating bispecific antigen binding molecule of any one of claims 1 to 3, further comprising a third antigen binding moiety capable of specific binding to a target cell antigen.

5. The T cell activating bispecific antigen binding molecule of claim 1, further comprising an Fc domain composed of a first and a second subunit capable of stable association.

6. The T cell activating bispecific antigen binding molecule of any one of claims 1 to 5, wherein the first and the second light chain comprises the light chain CDRs of SEQ ID NO: 32, SEQ ID NO: 33 and SEQ ID NO: 34.

7. The T cell activating bispecific antigen binding molecule of any one of claims 1 to 6, wherein the first and the second light chain comprise a kappa constant light chain domain.

8. The T cell activating bispecific antigen binding molecule of any one of claims 1 to 6, wherein the first and second light chain comprise a lambda constant light chain domain.

9. The T cell activating bispecific antigen binding molecule of claim 8, wherein the first and second light chain is a human or humanized lambda light chain.

10. The T cell activating bispecific antigen binding molecule of claim 9, wherein the first and second light chain is a human or humanized lambda 7 light chain.

11. The T cell activating bispecific antigen binding molecule of any of claims 1 to 6 and 8 to 10, wherein the first and second light chain comprise the amino acid sequence of SEQ ID NO: 31.

12. The T cell activating bispecific antigen binding molecule of claim 11, wherein the first and second light chain comprise the amino acid sequence of SEQ ID NO: 35.

13. The T cell activating bispecific antigen binding molecule of any of claims 1 to 8, wherein said antigen binding moiety capable of specific binding to an activating T cell antigen comprises a heavy chain comprising the heavy chain CDRs of SEQ ID NO: 37, SEQ ID NO: 38 and SEQ ID NO: 39.

14. The T cell activating bispecific antigen binding molecule of any of claims 1 to 13, comprising not more than one antigen binding moiety capable of specific binding to an activating T cell antigen.

15. The T cell activating bispecific antigen binding molecule of any of claims 1 to 13, wherein the first and the second antigen binding moiety are fused to each other, optionally via a peptide linker.

16. The T cell activating bispecific antigen binding molecule of any one of claims 2 to 13, wherein the second antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety.

17. The T cell activating bispecific antigen binding molecule of any one of claims 2 to 13, wherein the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding moiety.

18. The T cell activating bispecific antigen binding molecule of any one of claims 2 to 13 and 9, wherein the second antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or the second subunit of the Fc domain.

19. The T cell activating bispecific antigen binding molecule of any one of claims 2 to 13, wherein the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or second subunit of the Fc domain.

20. The T cell activating bispecific antigen binding molecule of any one of claims 2 to 13, wherein the first and the second antigen binding moiety are each fused at the C-terminus of the Fab heavy chain to the N-terminus of one of the subunits of the Fc domain.

21. The T cell activating bispecific antigen binding molecule of claim 4, wherein the first, second and third antigen binding moiety are each a Fab molecule comprising an identical VLCL light chain.

22. The T cell activating bispecific antigen binding molecule of claim 4 or 21, wherein the first, second and third antigen binding moiety are each a Fab molecule comprising the light chain CDRs of SEQ ID NO: 32, SEQ ID NO: 33 and SEQ ID NO: 34.

23. The T cell activating bispecific antigen binding molecule of claim 4 or 21, wherein the first, second and third antigen binding moiety are each a Fab molecule comprising a light chain comprising the amino acid sequence of SEQ ID NO: 31.

24. The T cell activating bispecific antigen binding molecule of any one of claims 4 or 21 to 23, wherein the third antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the first or second subunit of the Fc domain.

25. The T cell activating bispecific antigen binding molecule of any one of claims 4 or 21 to 23, wherein the second and the third antigen binding moiety are each fused at the C-terminus of the Fab heavy chain to the N-terminus of one of the subunits of the Fc domain, and the first antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the second antigen binding moiety.

26. The T cell activating bispecific antigen binding molecule of claim 25, wherein the second and the third antigen binding moiety and the Fc domain are part of an immunoglobulin molecule, particularly an IgG class immunoglobulin.

27. The T cell activating bispecific antigen binding molecule of any one of claims 4 or 21 to 23, wherein the first and the third antigen binding moiety are each fused at the C-terminus of the Fab heavy chain to the N-terminus of one of the subunits of the Fc domain, and the second antigen binding moiety is fused at the C-terminus of the Fab heavy chain to the N-terminus of the Fab heavy chain of the first antigen binding moiety.

28. The T cell activating bispecific antigen binding molecule of claim 27, wherein the first and the third antigen binding moiety and the Fc domain are part of an immunoglobulin molecule, particularly an IgG class immunoglobulin.

29. The T cell activating bispecific antigen binding molecule of any one of claims 5 to 28, wherein the Fc domain is an IgG, specifically an IgG1 or IgG4, Fc domain.

30. The T cell activating bispecific antigen binding molecule of any one of claims 5 to 25, wherein the Fc domain is a human Fc domain.

31. The T cell activating bispecific antigen binding molecule of any one of claims 5 to 23, wherein the Fc domain comprises a modification promoting the association of the first and the second subunit of the Fc domain.

32. The T cell activating bispecific antigen binding molecule of claim 31, wherein in the CH3 domain of the first subunit of the Fc domain an amino acid residue is replaced with an amino acid residue having a larger side chain volume, thereby generating a protuberance within the CH3 domain of the first subunit which is positionable in a cavity within the CH3 domain of the second subunit, and in the CH3 domain of the second subunit of the Fc domain an amino acid residue is replaced with an amino acid residue having a smaller side chain volume, thereby generating a cavity within the CH3 domain of the second subunit within which the protuberance within the CH3 domain of the first subunit is positionable.

33. The T cell activating bispecific antigen binding molecule of any one of claims 5 to 32, wherein the Fc domain exhibits reduced binding affinity to an Fc receptor and/or reduced effector function, as compared to a native IgG1 Fc domain.

34. The T cell activating bispecific antigen binding molecule of any one of claims 5 to 32, wherein the Fc domain comprises one or more amino acid substitution that reduces binding to an Fc receptor and/or effector function.

35. The T cell activating bispecific antigen binding molecule of claim 34, wherein said one or more amino acid substitution is at one or more position selected from the group of L234, L235, and P329.

36. The T cell activating bispecific antigen binding molecule of claim 34, wherein each subunit of the Fc domain comprises three amino acid substitutions that reduce binding to an activating Fc receptor and/or effector function wherein said amino acid substitutions are L234A, L235A and P329G.

37. The T cell activating bispecific antigen binding molecule of any one of claims 33 to 36, wherein the Fc receptor is an Fcγ receptor.

38. The T cell activating bispecific antigen binding molecule of any one of claims 33 to 37, wherein the effector function is antibody-dependent cell-mediated cytotoxicity (ADCC).

39. The T cell activating bispecific antigen binding molecule of any one of the preceding claims, wherein the activating T cell antigen is CD3.

40. The T cell activating bispecific antigen binding molecule of any one of the preceding claims, wherein the target cell antigen is selected from the group consisting of: Folate Receptor 1 (FolR1), Mucin-1 (MUC1), and B Cell Maturation Antigen (BCMA).

41. The T cell activating bispecific antigen binding molecule of any one of the preceding claims, wherein the first and the second light chain is a humanized light chain.

42. The T cell activating bispecific antigen binding molecule of any one of the preceding claims, wherein the first and the second light chain are capable of use in more than two antigen binding moieties having different antigen specificity.

43. The T cell activating bispecific antigen binding molecule of any of the preceding claims, wherein the target cell antigen is not BCMA.

44. A light chain comprising the amino acid sequence of SEQ ID NO: 32, SEQ ID NO: 33 and SEQ ID NO: 34 for use in a T cell activating bispecific antigen binding molecule.

45. The light chain of claim 44 which comprises a kappa constant light chain domain.

46. The light chain of claim 44 which comprises a lambda constant light chain domain.

47. The light chain of claim 46 which is a human or humanized lambda light chain.

48. The light chain of claim 46 which is a human or humanized lambda 7 light chain.

49. The light chain of any one of claims 46 to 48 comprising the amino acid sequence of SEQ ID NO: 31.

50. The light chain of claim 49 comprising the amino acid sequence of SEQ ID NO: 35.

51. A light chain comprising the amino acid sequence of SEQ ID NO: 32, SEQ ID NO: 33 and SEQ ID NO: 34 for use in a library for production of T cell activating bispecific antigen binding molecule.

52. The light chain of claim 51 which comprises a kappa constant light chain domain.

53. The light chain of claim 51 which comprises a lambda constant light chain domain.

54. The light chain of claim 53 which is a human or humanized lambda light chain.

55. The light chain of claim 54 which is a human or humanized lambda 7 light chain.

56. The light chain of any one of claims 53 to 55 comprising the amino acid sequence of SEQ ID NO: 31.

57. The light chain of claim 56 comprising the amino acid sequence of SEQ ID NO: 35.

58. An isolated polypeptide comprising the amino acid sequence of SEQ ID NO: 31.

59. An isolated polypeptide comprising the amino acid sequence of SEQ ID NO: 35.

60. An isolated polynucleotide encoding the T cell activating bispecific antigen binding molecule of any one of claims 1 to 41 or a fragment thereof or the isolated polypeptide of claim 58 or 59 or a fragment thereof.

61. A polypeptide encoded by the isolated polynucleotide of claim 60.

62. A vector, particularly an expression vector, comprising the isolated polynucleotide of claim 60.

63. A host cell comprising the isolated polynucleotide of claim 60 or the vector of claim 62.

64. A method of producing the T cell activating bispecific antigen binding molecule of any one of claims 1 to 43, comprising the steps of a) culturing the host cell of claim 63 under conditions suitable for the expression of the T cell activating bispecific antigen binding molecule and b) recovering the T cell activating bispecific antigen binding molecule.

65. A T cell activating bispecific antigen binding molecule produced by the method of claim 64.

66. A pharmaceutical composition comprising the T cell activating bispecific antigen binding molecule of any one of claims 1 to 43 and a pharmaceutically acceptable carrier.

67. A method of treating a disease in an individual, comprising administering to said individual a therapeutically effective amount of a composition comprising the T cell activating bispecific antigen binding molecule of any one of claims 1 to 43 or the light chain of any one of claims 44 to 57 in a pharmaceutically acceptable form.

68. The method of claim 67, wherein said disease is cancer.

69. A method for inducing lysis of a target cell, comprising contacting a target cell with the T cell activating bispecific antigen binding molecule of any one of claims 1 to 43 in the presence of a T cell.

70. A method for identifying a variable heavy chain for use in a bispecific antigen binding molecule specific for a T cell activation antigen and a target cell antigen, comprising the step of screening a combinatorial library comprising variable heavy chains with a light chain comprising the amino acid sequence of SEQ ID NO: 32, SEQ ID NO: 33 and SEQ ID NO: 34.

71. The method of claim 70, wherein the light chain comprises a kappa constant light chain domain.

72. The method of claim 70, wherein the light chain comprises a lambda constant light chain domain.

73. The method of claim 72, wherein the light chain is a human or humanized lambda light chain.

74. The light chain of claim 73 which is a human or humanized lambda 7 light chain.

75. The method of any one of claims 72 to 74, wherein the light chain comprises the amino acid sequence of SEQ ID NO: 31.

76. The method of claim 75, wherein the light chain comprises the amino acid sequence of SEQ ID NO: 35.

Patent History
Publication number: 20170253670
Type: Application
Filed: May 19, 2017
Publication Date: Sep 7, 2017
Applicant: Hoffmann-La Roche Inc. (Little Falls, NJ)
Inventors: Christian Klein (Bonstetten), Ekkehard Moessner (Kreuzlingen), Ralf Hosse (Cham), Peter Bruenker (Hittnau), Pablo Umana (Wollerau), Christiane Neumann (Niederweningen)
Application Number: 15/600,015
Classifications
International Classification: C07K 16/46 (20060101); C12N 15/73 (20060101); A61K 39/385 (20060101);