EP2758438A1 - Molécules de liaison bispécifiques pour 5t4 et cd3 - Google Patents

Molécules de liaison bispécifiques pour 5t4 et cd3

Info

Publication number
EP2758438A1
EP2758438A1 EP12778970.9A EP12778970A EP2758438A1 EP 2758438 A1 EP2758438 A1 EP 2758438A1 EP 12778970 A EP12778970 A EP 12778970A EP 2758438 A1 EP2758438 A1 EP 2758438A1
Authority
EP
European Patent Office
Prior art keywords
seq
depicted
cdr
region
human
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP12778970.9A
Other languages
German (de)
English (en)
Inventor
Peter Kufer
Roman Kischel
Doris Rau
Tobias Raum
Ralf Lutterbuese
Patrick Hoffmann
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Amgen Research Munich GmbH
Original Assignee
Amgen Research Munich GmbH
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Amgen Research Munich GmbH filed Critical Amgen Research Munich GmbH
Publication of EP2758438A1 publication Critical patent/EP2758438A1/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3007Carcino-embryonic Antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2809Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present invention relates to a bispecific binding molecule comprising a first and a second binding domain wherein the first binding domain is capable of binding to the epitope cluster 4 of 5T4 and the second binding domain is capable of binding to the CD3 receptor complex on T cells.
  • the invention relates to a nucleic acid sequence encoding the bispecific binding molecule, a vector comprising said nucleic acid sequence and a host cell transformed or transfected with said vector.
  • the invention relates to a process for the production of the bispecific binding molecule of the invention, a medical use of said bispecific binding molecule and a kit comprising said bispecific binding molecule.
  • the cell surface antigen defined by monoclonal antibody 5T4 is a 72-kD glycoprotein that is expressed by all types of trophoblasts as early as 9 weeks of development. In adult tissues, the 5T4 expression is limited to a few specialized epithelial cell types but not detected in adult liver, lung, bronchus, heart, testis, ovary, brain or muscle.
  • the antigen is selectively expressed by diverse tumour cell lines, including those of developmental origin. The molecular characteristics, the relatively restricted normal tissue distribution and expression by certain tumour cell types make this antigen worthy for use as a diagnostic marker of malignancy (Hole & Stern, Br J Cancer. (1988) 57(3), 239-46). [0004] .
  • 5T4 Since expression of 5T4 on normal tissues in contrast to cancerous lesions is restricted (AN A, Langdon J, Stern PL, Partidge M. The pattern of expression of 5T4 oncofoetal antigen on normal, dysplastic and malignant oral mucosa. Oral Oncol. 2001 ;37:57-64), 5T4 has been suggested as a suitable target antigen for targeted cancer therapy (Woods AM, Wang WW, Shaw DM, et al. Characterization of the murine 5T4 oncofoetal antigen: a target for immunotherapy in cancer. Biochem J. 2002;366:353-365 and Hole N, Stern PL. Isolation and characterization of 5T4, A tumor-associated antigen. Int J Cancer. 1990;45:179-184).
  • Antibody-based cancer therapies require a target antigen firmly bound to the surface of cancer cells in order to be active. By binding to the surface target, the antibody can directly or indirectly deliver a deadly signal to the cancer cell.
  • the target antigen 5T4 is abundantly present and accessible on specific cancer cells and is absent, shielded or much less abundant on normal cells.
  • the 5T4 antigen was identified as an oncology target more than 20 years ago, no anti-5T4 immunotherapy has been approved for treating cancer. Accordingly, there is a clear need for an effective therapeutic targeting this antigen. [0007] .
  • the present invention provides in a first embodiment a bispecific binding molecule comprising a first and a second binding domain,
  • epitope cluster 4 corresponds to extracellular protein domain of 5T4 formed by amino acid residues 170 to 222 of the human sequence as depicted in SEQ ID NO: 2.
  • the 5T4 onco-fetal antigen is a 72 kDa leucine-rich repeat (LRR) glycoprotein that belongs to the group of type 1 transmembrane proteins. Moreover, the 5T4 antigen is a highly N-glycosylated protein with seven putative consensus N-linked glycosylation sites in the extracellular domain (Shaw et al. Biochem. J. 363 (2002) 137-145).
  • the mature extracellular 5T4 antigen can be structured by its amino acid based elements: An N-terminal serine-rich repeat (SRR; 22 aa), followed by a 36 aa long undefined area containing four cysteines, three adjacent leucine-rich repeats (LLR 1 - 3) of 24, 24, and 23 amino acids in length, respectively, a hydrophilic segment of 45 aa followed by three more leucine-rich repeats (LRR 4 - 5) of 24, 24, and 1 +23 amino acids in length, respectively, a second undefined segment of 53 aa with four more cysteines which is finally followed by a last leucine-rich repeat (LRR 7) of 22+4 amino acids in length (see Figure 1 for an illustration of the structure of 5T4).
  • SRR N-terminal serine-rich repeat
  • LLR 1 - 3 three adjacent leucine-rich repeats
  • LRR 4 - 5 three adjacent leucine-rich repeats
  • LRR 4 - 5 three more leucine-rich repeats
  • N-terminal sequencing of human 5T4 to identify amino acid T35 in the immature protein to be the most N-terminal amino acid of the mature protein after cleavage of the N-terminal leader peptide.
  • a hypothetical structure of 5T4 which was used to design the experiments shown in the examples is depicted in Figure 1 .
  • the redirected lysis of target cells via the recruitment of T cells by bispecific molecules involves cytolytic synapse formation and delivery of perforin and granzymes.
  • the engaged T cells are capable of serial target cell lysis, and are not affected by immune escape mechanisms interfering with peptide antigen processing and presentation, or clonal T cell differentiation.
  • the potency of a compound should increase when the affinity of the compound to the target structure is increased.
  • binders for the membrane distal epitope cluster show significantly higher potency in the lysis of 5T4 expressing target cells than those which bind to 5T4 at an epitope more proximal to the membrane. This is especially surprising since the affinity of the binder for 5T4 was not determined using artificial systems with recombinant antigen expression (e.g. immobilization of a chip) but by Scatchard using viable cells which naturally express the 5T4 antigen.
  • binding domain characterizes in connection with the present invention a domain of a polypeptide which specifically binds/interacts with a given target epitope (capable of binding to/interacting with a given target epitope).
  • An “epitope” is antigenic and thus the term epitope is sometimes also referred to herein as “antigenic structure” or “antigenic determinant”.
  • the binding domain is an "antigen-interaction-site".
  • the term “antigen-interaction-site” defines, in accordance with the present invention, a motif of a polypeptide, which is able to specifically interact with a specific antigen or a specific group of antigens, e.g. the identical antigen in different species. Said binding/interaction is also understood to define a "specific recognition".
  • said polypeptide which specifically binds/Interacts with a given target epitope is an antibody and said domain is a VH and/or VL region of an antibody.
  • epitope refers to a site on an antigen to which a binding domain, such as an antibody or immunoglobulin or derivative of an antibody of immunoglobulin, specifically binds. Epitopes can be formed both from contiguous amino acids or noncontiguous amino acids juxtaposed by tertiary folding of a protein.
  • a “linear epitope” is an epitope where an amino acid primary sequence comprises the epitope recognized.
  • a linear epitope typically includes at least 3, and more usually, at least 5, for example, about 8 to about 10 amino acids in a unique sequence.
  • a “conformational epitope”, in contrast to a linear epitope, is an epitope wherein the primary sequence of the amino acids comprising the epitope is not the sole defining component of the epitope recognized (e.g., an epitope wherein the primary sequence of amino acids is not necessarily recognized by the antibody defining the epitope).
  • a conformational epitope comprises an increased number of amino acids relative to a linear epitope.
  • the binding domain recognizes a 3-dimensional structure of the antigen, preferably a peptide or protein or fragment thereof (in the context of the present invention, the antigen is for one of the binding domains the 5T4 protein).
  • a protein molecule folds to form a three dimensional structure, certain amino acids and/or the polypeptide backbone forming the conformational epitope become juxtaposed enabling the antibody to recognize the epitope.
  • Methods of determining conformation of epitopes include but are not limited to, for example, x-ray crystallography 2-dimensional nuclear magnetic resonance spectroscopy and site- directed spin labeling and electron paramagnetic resonance spectroscopy.
  • the provided example describe a further method to test whether a given binding domain binds to one or more epitope cluster of a given protein
  • epitope cluster denotes the entirety of epitopes lying in a defined contiguous stretch of an antigen.
  • a human 5T4 antigen binding ligand binds to one specific epitope cluster and significantly loses binding capability to the antigen when the respective epitope cluster in the human 5T4 protein is exchanged with the respective epitope cluster of a non-primate (e.g. murine) 5T4 antigen.
  • a non-primate (e.g. murine) 5T4 antigen is described in the appended examples 1 to 3.
  • binding domain is capable of specifically interacting with and/or binding to at least two, preferably at least three, more preferably at least four amino acids of an epitope. Such binding may be exemplified by the specificity of a "lock-and-key- principle”.
  • binding domain exhibits appreciable affinity for a particular protein or antigen and, generally, does not exhibit significant cross-reactivity with other proteins or antigens.
  • “Appreciable” or preferred binding includes binding with an affinity of 10 "6 M (KD) or higher. Preferably, binding is considered specific when binding affinity is about 10 "11 to 10 "8 M, preferably of about 10 "11 to 10 "9 M. If necessary, nonspecific binding can be reduced without substantially affecting specific binding by varying the binding conditions. Whether binding domains specifically react or bind with a target can be tested readily by, inter alia, comparing the reaction of one of said binding domains with a target protein or antigen with the reaction of said binding domains with other proteins or antigens.
  • binding domain is an antibody.
  • the term "does not essentially bind” means that a binding domain of the bispecific binding molecule of the present invention does not bind another protein, i.e., shows a cross- reactivity of less than 30%, preferably 20%, more preferably 10%, particularly preferably less than 9, 8, 7, 6 or 5% with another protein.
  • cross-species specificity denotes a binding domain's ability to bind to the same target molecule in humans and non-human species, preferably in primate species.
  • cross- species specificity or “interspecies specificity” describes interspecies reactivity to the same molecule X (e.g., 5T4 or CD3s) expressed in different species, but not to a molecule other than X (e.g., 5T4 or CD3s).
  • the cross-species specificity of the inventive binding domains is limited to human proteins and their corresponding analogs in primates.
  • human 5T4 amino acid sequence SEQ ID NO: 2
  • cynomolgus SEQ ID NO: 4
  • rhesus SEQ ID NO: 6
  • 5T4 amino acid sequence SEQ ID NO: 6
  • a binding domain against cynomolgus or rhesus 5T4 likely binds human 5T4.
  • a binding domain which binds to human 5T4, in particular to epitope cluster 4 of the extracellular protein domain of 5T4 formed by amino acid residues 170 to 222 of the human sequence as depicted in SEQ ID NO: 1 also binds to cynomolgus 5T4, in particular to epitope cluster 4 of the extracellular protein domain of 5T4 formed by amino acid residues 170 to 222 of the cynomolgus sequence as depicted in SEQ ID NO: 4.
  • Proteins (including fragments thereof, preferably biologically active fragments, and peptides, usually having less than 30 amino acids) comprise one or more amino acids coupled to each other via a covalent peptide bond (resulting in a chain of amino acids).
  • polypeptide as used herein describes a group of molecules, which consist of more than 30 amino acids. Polypeptides may further form multimers such as dimers, trimers and higher oligomers, i.e. consisting of more than one polypeptide molecule. Polypeptide molecules forming such dimers, trimers etc. may be identical or non-identical. The corresponding higher order structures of such multimers are, consequently, termed homo- or heterodimers, homo- or heterotrimers etc.
  • polypeptide and protein also refer to naturally modified polypeptides/proteins wherein the modification is effected e.g. by post-translational modifications like glycosylation, acetylation, phosphorylation and the like. Such modifications are well known in the art.
  • a first binding domain of a bispecific binding molecule is capable of binding to epitope cluster 2 and epitope cluster 4 of 5T4, wherein the epitope cluster 2 corresponds to extracellular protein domain of 5T4 formed by amino acid residues 78 to 138 of the human sequence as depicted in SEQ ID NO: 2.
  • the first binding domain of the bispecific binding molecule is capable of binding to human and Callithrix jacchus, Saguinus oedipus or Saimiri sciureus 5T4 and/or the second binding domain is capable of binding to human and Callithrix jacchus, Saguinus Oedipus or Saimiri sciureus CD3 epsilon.
  • one or both binding domains of the bispecific binding molecule of the invention are cross-species specific for members of the mammalian order of primates.
  • the affinity of the first binding domain for human 5T4 is ⁇ 15 nM (preferably ⁇ 10 nM).
  • Bispecific binding molecules of the invention binding to an epitope in epitope cluster 4 or epitope cluster 2 and 4 showing this high affinity for 5T4 have shown superior cytotoxic activity over bispecific binding molecules binding to an epitope in epitope cluster 7 with affinities in the same range.
  • the first or the second binding domain is derived from an antibody. In another, both binding domains are derived from an antibody.
  • antibody includes embodiments such as monoclonal, chimeric, single chain, humanized and human antibodies, as well as antibody fragments, like, inter alia, Fab fragments.
  • Antibody fragments or derivatives further comprise F(ab') 2 , Fv, scFv fragments or single domain antibodies such as domain antibodies or nanobodies, single variable domain antibodies or immunoglobulin single variable domain comprising merely one variable domain, which might be VHH, VH or VL, that specifically bind an antigen or epitope independently of other V regions or domains; see, for example, Harlow and Lane (1988) and (1999), loc. cit; Kontermann and Dubel, Antibody Engineering, Springer, 2nd ed.
  • immunoglobulin single variable domain encompasses not only an isolated antibody single variable domain polypeptide, but also larger polypeptides that comprise one or more monomers of an antibody single variable domain polypeptide sequence.
  • (antibody) derivatives can be produced by peptidomimetics.
  • techniques described for the production of single chain antibodies see, inter alia, US Patent 4,946,778, Kontermann and Dubel (2010), loc. cit. and Little(2009), loc. cit.
  • transgenic animals may be used to express humanized antibodies specific for polypeptides and fusion proteins of this invention.
  • any technique, providing antibodies produced by continuous cell line cultures can be used.
  • Examples for such techniques include the hybridoma technique (Kohler and Milstein Nature 256 (1975), 495-497), the trioma technique, the human B-cell hybridoma technique (Kozbor, Immunology Today 4 (1983), 72) and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc. (1985), 77-96).
  • antibody comprises antibody constructs, which may be expressed in a host as described herein below, e.g. antibody constructs which may be transfected and/or transduced via, inter alia, viruses or plasmid vectors.
  • antibody as employed in the invention also relates to derivatives or variants of the antibodies described herein which display the same specificity as the described antibodies.
  • antibody variants include humanized variants of non- human antibodies, "affinity matured” antibodies (see, e.g. Hawkins et al. J. Mol. Biol. 254, 889-896 (1992) and Lowman et al., Biochemistry 30, 10832- 10837 (1991 )) and antibody mutants with altered effector function (s) (see, e.g., US Patent 5, 648, 260, Kontermann and Dubel (2010), loc. cit. and Little(2009), loc. cit).
  • an antigen-binding domain refers to a part of an antibody molecule that comprises amino acids responsible for the specific binding between antibody and antigen.
  • the part of the antigen that is specifically recognized and bound by the antibody is referred to as the "epitope" as described herein above.
  • an antigen-binding domain may typically comprise an antibody light chain variable region (VL) and an antibody heavy chain variable region (VH); however, it does not have to comprise both.
  • Fd fragments for example, have two VH regions and often retain some antigen-binding function of the intact antigen-binding domain.
  • antigen-binding fragments of an antibody examples include (1 ) a Fab fragment, a monovalent fragment having the VL, VH, CL and CH1 domains; (2) a F(ab')2 fragment, a bivalent fragment having two Fab fragments linked by a disulfide bridge at the hinge region; (3) a Fd fragment having the two VH and CH 1 domains; (4) a Fv fragment having the VL and VH domains of a single arm of an antibody, (5) a dAb fragment (Ward et al., (1989) Nature 341 :544-546), which has a VH domain; (6) an isolated complementarity determining region (CDR), and (7) a single chain Fv (scFv).
  • a Fab fragment a monovalent fragment having the VL, VH, CL and CH1 domains
  • F(ab')2 fragment a bivalent fragment having two Fab fragments linked by a disulfide bridge at the hinge region
  • a Fd fragment having
  • the two domains of the Fv fragment, VL and VH are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Huston et al. (1988) Proc. Natl. Acad. Sci USA 85:5879-5883).
  • scFv single chain Fv
  • monoclonal antibody refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations and/or post- translation modifications (e.g., isomerizations, amidations) that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen.
  • the monoclonal antibodies are advantageous in that they are synthesized by the hybridoma culture, uncontaminated by other immunoglobulins.
  • the modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al., Nature, 256: 495 (1975), or may be made by recombinant DNA methods (see, e.g., U. S. Patent No. 4,816, 567).
  • the "monoclonal antibodies” may also be isolated from phage antibody libraries using the techniques described in Clackson et al., Nature, 352: 624-628 (1991 ) and Marks et al., J. Mol. Biol., 222: 581 -597 (1991 ), for example.
  • the monoclonal antibodies herein specifically include "chimeric" antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain (s) is (are) identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (U. S. Patent No. 4,816, 567; Morrison et al., Proc. Natl. Acad. Sci. USA, 81 : 6851 -6855 (1984)).
  • Chimeric antibodies of interest herein include "primitized" antibodies comprising variable domain antigen-binding sequences derived from a non-human primate (e.g., Old World Monkey, Ape etc.) and human constant region sequences.
  • “Humanized” forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F (ab') 2 or other antigen-binding subsequences of antibodies) of mostly human sequences, which contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region (also CDR) of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity, and capacity.
  • Fv framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • "humanized antibodies” as used herein may also comprise residues which are found neither in the recipient antibody nor the donor antibody. These modifications are made to further refine and optimize antibody performance.
  • the humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • human antibody includes antibodies having variable and constant regions corresponding substantially to human germline immunoglobulin sequences known in the art, including, for example, those described by Kabat et al. (See Kabat, et al. (1991 ) loc. cit).
  • the human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs, and in particular, CDR3.
  • the human antibody can have at least one, two, three, four, five, or more positions replaced with an amino acid residue that is not encoded by the human germline immunoglobulin sequence.
  • in vitro generated antibody refers to an antibody where all or part of the variable region (e.g., at least one CDR) is generated in a non-immune cell selection (e.g., an in vitro phage display, protein chip or any other method in which candidate sequences can be tested for their ability to bind to an antigen). This term thus preferably excludes sequences generated by genomic rearrangement in an immune cell.
  • a “bispecific” or “Afunctional antibody” is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites. Bispecific antibodies can be produced by a variety of methods including fusion of hybridomas or linking of Fab' fragments.
  • antibodies can be produced using recombinant DNA methods (U.S. Patent 4,816,567).
  • Monoclonal antibodies may also be produced by generation of hybridomas (see e.g., Kohler and Milstein (1975) Nature, 256: 495-499) in accordance with known methods.
  • Hybridomas formed in this manner are then screened using standard methods, such as enzyme-linked immunosorbent assay (ELISA) and surface plasmon resonance (BIACORETM) analysis, to identify one or more hybridomas that produce an antibody that specifically binds with a specified antigen.
  • ELISA enzyme-linked immunosorbent assay
  • BIACORETM surface plasmon resonance
  • Any form of the specified antigen may be used as the immunogen, e.g., recombinant antigen, naturally occurring forms, any variants or fragments thereof, as well as antigenic peptide thereof.
  • One exemplary method of making antibodies includes screening protein expression libraries, e.g., phage or ribosome display libraries.
  • Phage display is described, for example, in Ladner et al., U.S. Patent No. 5,223,409; Smith (1985) Science 228:1315-1317; Clackson et al. (1991 ) Nature, 352: 624-628.
  • the specified antigen can be used to immunize a non-human animal, e.g., a rodent, e.g., a mouse, hamster, or rat.
  • the non-human animal includes at least a part of a human immunoglobulin gene.
  • antigen-specific monoclonal antibodies derived from the genes with the desired specificity may be produced and selected. See, e.g., XENOMOUSETM, Green et al. (1994) Nature Genetics 7:13-21 , US 2003- 0070185, WO 96/34096, and W096/33735.
  • a monoclonal antibody can be obtained from a non-human animal, and then modified, e.g., humanized, deimmunized, chimeric, may be produced using recombinant DNA techniques known in the art.
  • modified e.g., humanized, deimmunized, chimeric
  • a variety of approaches for making chimeric antibodies have been described. See e.g., Morrison et al., Proc. Natl. Acad. ScL U.S.A. 81 :6851 , 1985; Takeda et al., Nature 314:452, 1985, Cabilly et al., U.S. Patent No. 4,816,567; Boss et al., U.S. Patent No.
  • Humanized antibodies may also be produced, for example, using transgenic mice that express human heavy and light chain genes, but are incapable of expressing the endogenous mouse immunoglobulin heavy and light chain genes. Winter describes an exemplary CDR-grafting method that may be used to prepare the humanized antibodies described herein (U.S. Patent No. 5,225,539). All of the CDRs of a particular human antibody may be replaced with at least a portion of a non-human CDR, or only some of the CDRs may be replaced with non-human CDRs. It is only necessary to replace the number of CDRs required for binding of the humanized antibody to a predetermined antigen.
  • Humanized antibodies or fragments thereof can be generated by replacing sequences of the Fv variable domain that are not directly involved in antigen binding with equivalent sequences from human Fv variable domains.
  • Exemplary methods for generating humanized antibodies or fragments thereof are provided by Morrison (1985) Science 229:1202-1207; by Oi et al. (1986) BioTechniques 4:214; and by US 5,585,089; US 5,693,761 ; US 5,693,762; US 5,859,205; and US 6,407,213. Those methods include isolating, manipulating, and expressing the nucleic acid sequences that encode all or part of immunoglobulin Fv variable domains from at least one of a heavy or light chain.
  • nucleic acids may be obtained from a hybridoma producing an antibody against a predetermined target, as described above, as well as from other sources.
  • the recombinant DNA encoding the humanized antibody molecule can then be cloned into an appropriate expression vector.
  • a humanized antibody can be optimized by the introduction of conservative substitutions, consensus sequence substitutions, germline substitutions and/or back mutations.
  • Such altered immunoglobulin molecules can be made by any of several techniques known in the art, (e.g., Teng et al., Proc. Natl. Acad. Sci. U.S.A., 80: 7308-7312, 1983; Kozbor et al, Immunology Today, 4: 7279, 1983; Olsson et al., Meth. Enzymol., 92: 3- 16, 1982, and may be made according to the teachings of EP 239 400).
  • An antibody or fragment thereof may also be modified by specific deletion of human T cell epitopes or "deimmunization" by the methods disclosed in WO 98/52976 and WO 00/34317. Briefly, the heavy and light chain variable domains of an antibody can be analyzed for peptides that bind to MHC Class II; these peptides represent potential T-cell epitopes (as defined in WO 98/52976 and WO 00/34317).
  • peptide threading For detection of potential T-cell epitopes, a computer modeling approach termed "peptide threading" can be applied, and in addition a database of human MHC class II binding peptides can be searched for motifs present in the VH and VL sequences, as described in WO 98/52976 and WO 00/34317. These motifs bind to any of the 18 major MHC class II DR allotypes, and thus constitute potential T cell epitopes.
  • Potential T-cell epitopes detected can be eliminated by substituting small numbers of amino acid residues in the variable domains, or preferably, by single amino acid substitutions. Typically, conservative substitutions are made. Often, but not exclusively, an amino acid common to a position in human germline antibody sequences may be used.
  • the pairing of a VH and VL together forms a single antigen-binding site.
  • the CH domain most proximal to VH is designated as CH1.
  • Each L chain is linked to an H chain by one covalent disulfide bond, while the two H chains are linked to each other by one or more disulfide bonds depending on the H chain isotype.
  • the VH and VL domains consist of four regions of relatively conserved sequences called framework regions (FR1 , FR2, FR3, and FR4), which form a scaffold for three regions of hypervariable sequences (complementarity determining regions, CDRs).
  • the CDRs contain most of the residues responsible for specific interactions of the antibody with the antigen.
  • CDRs are referred to as CDR 1 , CDR2, and CDR3. Accordingly, CDR constituents on the heavy chain are referred to as H1 , H2, and H3, while CDR constituents on the light chain are referred to as L1 , L2, and L3.
  • variable refers to the portions of the immunoglobulin domains that exhibit variability in their sequence and that are involved in determining the specificity and binding affinity of a particular antibody (i.e., the "variable domain(s)"). Variability is not evenly distributed throughout the variable domains of antibodies; it is concentrated in sub-domains of each of the heavy and light chain variable regions. These sub-domains are called “hypervariable” regions or “complementarity determining regions” (CDRs). The more conserved (i.e., non-hypervariable) portions of the variable domains are called the "framework" regions (FRM).
  • CDRs complementarity determining regions
  • variable domains of naturally occurring heavy and light chains each comprise four FRM regions, largely adopting a ⁇ - sheet configuration, connected by three hypervariable regions, which form loops connecting, and in some cases forming part of, the ⁇ -sheet structure.
  • the hypervariable regions in each chain are held together in close proximity by the FRM and, with the hypervariable regions from the other chain, contribute to the formation of the antigen- binding site (see Kabat et al., loc. cit).
  • the constant domains are not directly involved in antigen binding, but exhibit various effector functions, such as, for example, antibody- dependent, cell-mediated cytotoxicity and complement activation.
  • first and the second domain form a molecule that is selected from the group of (scFv) 2 , (single domain mAb) 2 , scFv-single domain mAb, diabody or oligomeres thereof.
  • CDR complementarity determining region
  • CDRL1 , CDRL2 and CDRL3 three make up the binding character of a light chain variable region
  • CDRH1 , CDRH2 and CDRH3 three make up the binding character of a heavy chain variable region
  • CDRs contribute to the functional activity of an antibody molecule and are separated by amino acid sequences that comprise scaffolding or framework regions.
  • the exact definitional CDR boundaries and lengths are subject to different classification and numbering systems. CDRs may therefore be referred to by Kabat, Chothia, contact or any other boundary definitions, including the numbering system described herein. Despite differing boundaries, each of these systems has some degree of overlap in what constitutes the so called "hypervariable regions" within the variable sequences. CDR definitions according to these systems may therefore differ in length and boundary areas with respect to the adjacent framework region. See for example Kabat, Chothia, and/or MacCallum et al., (Kabat et al., loc. cit.; Chothia et al., J. Mol. Biol, 1987, 196: 901 ; and MacCallum et al, J. Mol. Biol, 1996, 262: 732). However, the numbering in accordance with the so-called Kabat system is preferred.
  • amino acid typically refers to an amino acid having its art recognized definition such as an amino acid selected from the group consisting of: alanine (Ala or A); arginine (Arg or R); asparagine (Asn or N); aspartic acid (Asp or D); cysteine (Cys or C); glutamine (Gin or Q); glutamic acid (Glu or E); glycine (Gly or G); histidine (His or H); isoleucine (He or I): leucine (Leu or L); lysine (Lys or K); methionine (Met or M); phenylalanine (Phe or F); pro line (Pro or P); serine (Ser or S); threonine (Thr or T); tryptophan (Trp or W); tyrosine (Tyr or Y); and valine (Val or V), although modified, synthetic, or rare amino acids may be used
  • amino acids can be grouped as having a nonpolar side chain (e.g., Ala, Cys, He, Leu, Met, Phe, Pro, Val); a negatively charged side chain (e.g., Asp, Glu); a positively charged sidechain (e.g., Arg, His, Lys); or an uncharged polar side chain (e.g., Asn, Cys, Gin, Gly, His, Met, Phe, Ser, Thr, Trp, and Tyr).
  • a nonpolar side chain e.g., Ala, Cys, He, Leu, Met, Phe, Pro, Val
  • a negatively charged side chain e.g., Asp, Glu
  • a positively charged sidechain e.g., Arg, His, Lys
  • an uncharged polar side chain e.g., Asn, Cys, Gin, Gly, His, Met, Phe, Ser, Thr, Trp, and Tyr.
  • hypervariable region also known as “complementarity determining regions” or CDRs
  • CDRs complementarity determining regions
  • CDR residues are preferably identified in accordance with the so-called Kabat (numbering) system.
  • framework region refers to the art-recognized portions of an antibody variable region that exist between the more divergent (i.e., hypervariable) CDRs.
  • framework regions are typically referred to as frameworks 1 through 4 (FR1 , FR2, FR3, and FR4) and provide a scaffold for the presentation of the six CDRs (three from the heavy chain and three from the light chain) in three dimensional space, to form an antigen-binding surface.
  • CDRs form a loop structure that can be classified as a canonical structure.
  • canonical structure refers to the main chain conformation that is adopted by the antigen binding (CDR) loops. From comparative structural studies, it has been found that five of the six antigen binding loops have only a limited repertoire of available conformations. Each canonical structure can be characterized by the torsion angles of the polypeptide backbone. Correspondent loops between antibodies may, therefore, have very similar three dimensional structures, despite high amino acid sequence variability in most parts of the loops (Chothia and Lesk, J. Mol.
  • canonical structure may also include considerations as to the linear sequence of the antibody, for example, as catalogued by Kabat (Kabat et al, loc. cit).
  • Kabat numbering scheme system
  • a given antibody sequence may be placed into a canonical class which allows for, among other things, identifying appropriate chassis sequences ⁇ e.g., based on a desire to include a variety of canonical structures in a library).
  • Kabat numbering of antibody amino acid sequences and structural considerations as described by Chothia et al., loc. cit. and their implications for construing canonical aspects of antibody structure, are described in the literature.
  • CDR3 is typically the greatest source of molecular diversity within the antibody- binding site.
  • H3 for example, can be as short as two amino acid residues or greater than 26 amino acids.
  • the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known in the art. For a review of the antibody structure, see Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, eds. Harlow et al., 1988.
  • each subunit structure e.g., a CH, VH, CL, VL, CDR, FR structure
  • comprises active fragments e.g., the portion of the VH, VL, or CDR subunit the binds to the antigen, i.e., the antigen-binding fragment, or, e.g., the portion of the CH subunit that binds to and/or activates, e.g., an Fc receptor and/or complement.
  • the CDRs typically refer to the Kabat CDRs, as described in Sequences of Proteins of immunological Interest, US Department of Health and Human Services (1991 ), eds. Kabat et al.
  • Another standard for characterizing the antigen binding site is to refer to the hypervariable loops as described by Chothia. See, e.g., Chothia, et al. (1987; J. Mol. Biol. 227:799-817; and Tomlinson et al. (1995) EMBO J. 14: 4628-4638. Still another standard is the AbM definition used by Oxford Molecular's AbM antibody modeling software. See, generally, e.g., Protein Sequence and Structure Analysis of Antibody Variable Domains. In: Antibody Engineering Lab Manual (Ed.: Duebel, S. and Kontermann, R., Springer-Verlag, Heidelberg). Embodiments described with respect to Kabat CDRs can alternatively be implemented using similar described relationships with respect to Chothia hypervariable loops or to the AbM-defined loops.
  • the sequence of antibody genes after assembly and somatic mutation is highly varied, and these varied genes are estimated to encode 10 10 different antibody molecules (Immunoglobulin Genes, 2nd ed., eds. Jonio et al., Academic Press, San Diego, CA, 1995). Accordingly, the immune system provides a repertoire of immunoglobulins.
  • the term "repertoire” refers to at least one nucleotide sequence derived wholly or partially from at least one sequence encoding at least one immunoglobulin.
  • the sequence(s) may be generated by rearrangement in vivo of the V, D, and J segments of heavy chains, and the V and J segments of light chains.
  • sequence(s) can be generated from a cell in response to which rearrangement occurs, e.g., in vitro stimulation.
  • part or all of the sequence(s) may be obtained by DNA splicing, nucleotide synthesis, mutagenesis, and other methods, see, e.g., U.S. Patent 5,565,332.
  • a repertoire may include only one sequence or may include a plurality of sequences, including ones in a genetically diverse collection.
  • the first binding domain of a bispecific binding molecule comprises a VL region and a VH region, wherein the VH region comprises a CDR-H3 selected from the group consisting of SEQ ID NOs: 15, 25, 35, 49, 89, 99, 109, 119, 129, 143, 157 and 167.
  • the bispecific binding molecule of the invention comprises a VL region comprising CDR-L1 , CDR-L2 and CDR-L3 and a VH region comprising CDR-H1 , CDR-H2 and CDR-H3 selected from:
  • the first binding domain of the bispecific binding molecule comprises a VL region selected from the group consisting of a VL region as depicted in SEQ ID NO: 12, SEQ ID NO: 22, SEQ I D NO: 32, SEQ I D NO: 46, SEQ ID NO: 86, SEQ ID NO: 96, SEQ ID NO: 106, SEQ ID NO: 116, SEQ ID NO: 126, SEQ ID NO: 140, SEQ ID NO: 154, and SEQ ID NO: 164. [0060] .
  • the first binding domain comprises a VH region selected from the group consisting of a VH region as depicted in VH region as depicted in SEQ ID NO: 11 , SEQ ID NO: 21 , SEQ ID NO: 31 , SEQ ID NO: 45, SEQ I D NO: 85, SEQ ID NO: 95, SEQ ID NO: 105, SEQ I D NO: 115, SEQ I D NO: 125, SEQ ID NO: 139, SEQ ID NO: 153, and SEQ ID NO: 163.
  • the first binding domain of the bispecific binding molecule comprises a VL region and a VH region selected from the group consisting of:
  • the first binding domain comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 10, SEQ ID NO: 20, SEQ ID NO: 30, SEQ ID NO: 39, SEQ ID NO: 44, SEQ ID NO: 84, SEQ ID NO: 94, SEQ I D NO: 104, SEQ ID NO: 114, SEQ ID NO: 124, SEQ ID NO: 133, SEQ ID NO: 138, SEQ ID NO: 147, SEQ ID NO: 152, and SEQ ID NO: 162.
  • the bispecific binding molecule of the present invention is preferably an "isolated" bispecific binding molecule.
  • isolated when used to describe the bispecific binding molecule disclosed herein, means a bispecific binding molecule that has been identified, separated and/or recovered from a component of its production environment.
  • the isolated bispecific binding molecule is free of association with all other components from its production environment. Contaminant components of its production environment, such as that resulting from recombinant transfected cells, are materials that would typically interfere with diagnostic or therapeutic uses for the polypeptide, and may include enzymes, hormones, and other proteinaceous or non-proteinaceous solutes.
  • the bispecific binding molecule will be purified (1 ) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (2) to homogeneity by SDS-PAGE under non-reducing or reducing conditions using Coomassie blue or, preferably, silver stain. Ordinarily, however, an isolated antibody will be prepared by at least one purification step.
  • Amino acid sequence modifications of the bispecific binding molecules described herein are contemplated. For example, it may be desirable to improve the binding affinity and/or other biological properties of the antibody.
  • Amino acid sequence variants of the bispecific binding molecules are prepared by introducing appropriate nucleotide changes into the bispecific binding molecules nucleic acid, or by peptide synthesis.
  • Such modifications include, for example, deletions from, and/or insertions into, and/or substitutions of, residues within the amino acid sequences of the bispecific binding molecules. Any combination of deletion, insertion, and substitution is made to arrive at the final construct, provided that the final construct possesses the desired characteristics.
  • the amino acid changes also may alter post-translational processes of the bispecific binding molecules, such as changing the number or position of glycosylation sites.
  • 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids may be substituted in a CDR, while 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, or 25 amino acids may be substituted in the FRs.
  • substitutions are preferably conservative substitutions as described herein. Additionally or alternatively, 1 , 2, 3, 4, 5, or 6 amino acids may be inserted or deleted in each of the CDRs (of course, dependent on their length), while 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, or 25 amino acids may be inserted or deleted in each of the FRs.
  • a useful method for identification of certain residues or regions of the bispecific binding molecules that are preferred locations for mutagenesis is called "alanine scanning mutagenesis" as described by Cunningham and Wells in Science, 244: 1081 -1085 (1989).
  • a residue or group of target residues within the bispecific binding molecule is/are identified (e.g. charged residues such as arg, asp, his, lys, and glu) and replaced by a neutral or negatively charged amino acid (most preferably alanine or polyalanine) to affect the interaction of the amino acids with epitope.
  • amino acid sequence insertions include amino-and/or carboxyl-terminal fusions ranging in length from one, two, three, four, five, six, seven, eight, nine or ten residues to polypeptides containing a hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues.
  • An insertional variant of the bispecific binding molecule include the fusion to the N-or C-terminus of the antibody to an enzyme or a fusion to a polypeptide which increases the serum half-life of the antibody.
  • variants are an amino acid substitution variant. These variants have preferably at least one, two, three, four, five, six, seven, eight, nine or ten amino acid residues in the bispecific binding molecule replaced by a different residue.
  • the sites of greatest interest for substitutional mutagenesis include the CDRs of the heavy and/or light chain, in particular the hypervariable regions, but FR alterations in the heavy and/or light chain are also contemplated.
  • a CDR sequence encompasses 6 amino acids, it is envisaged that one, two or three of these amino acids are substituted. Similarly, if a CDR sequence encompasses 15 amino acids it is envisaged that one, two, three, four, five or six of these amino acids are substituted.
  • the then-obtained "substituted" sequence is at least 60%, more preferably 65%, even more preferably 70%, particularly preferable 75%, more particularly preferable 80% identical to the "original" CDR sequence. This means that it is dependent of the length of the CDR to which degree it is identical to the "substituted" sequence.
  • a CDR having 5 amino acids is preferably 80% identical to its substituted sequence in order to have at least one amino acid substituted.
  • the CDRs of the bispecific binding molecule may have different degrees of identity to their substituted sequences, e.g., CDRL1 may have 80%, while CDRL3 may have 90%.
  • Preferred substitutions (or replacements) are conservative substitutions.
  • any substitution including non-conservative substitution or one or more from the "exemplary substitutions listed in Table 1 , below is envisaged as long as the bispecific binding molecule retains its capability to bind to 5T4 via the first binding domain and to CD3s via the second binding domain and/or its CDRs have an identity to the then substituted sequence (at least 60%, more preferably 65%, even more preferably 70%, particularly preferable 75%, more particularly preferable 80% identical to the "original" CDR sequence).
  • Substantial modifications in the biological properties of the binding molecule of the present invention are accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain.
  • Naturally occurring residues are divided into groups based on common side-chain properties: (1 ) hydrophobic: norleucine, met, ala, val, leu, ile; (2) neutral hydrophilic: cys, ser, thr; (3) acidic: asp, glu; (4) basic: asn, gin, his, lys, arg; (5) residues that influence chain orientation: gly, pro; and (6) aromatic : trp, tyr, phe. [0075] .
  • Non-conservative substitutions will entail exchanging a member of one of these classes for another class. Any cysteine residue not involved in maintaining the proper conformation of the bispecific binding molecule may be substituted, generally with serine, to improve the oxidative stability of the molecule and prevent aberrant crosslinking.
  • cysteine bond may be added to the antibody to improve its stability (particularly where the antibody is an antibody fragment such as an Fv fragment).
  • a particularly preferred type of substitutional variant involves substituting one or more hypervariable region residues of a parent antibody (e. g. a humanized or human antibody).
  • a parent antibody e. g. a humanized or human antibody
  • the resulting variant (s) selected for further development will have improved biological properties relative to the parent antibody from which they are generated.
  • a convenient way for generating such substitutional variants involves affinity maturation using phage display. Briefly, several hypervariable region sites (e. g. 6-7 sites) are mutated to generate all possible amino acid substitutions at each site.
  • the antibody variants thus generated are displayed in a monovalent fashion from filamentous phage particles as fusions to the gene III product of M13 packaged within each particle. The phage-displayed variants are then screened for their biological activity (e.
  • alanine scanning mutagenesis can be performed to identify hypervariable region residues contributing significantly to antigen binding.
  • Such contact residues and neighboring residues are candidates for substitution according to the techniques elaborated herein.
  • bispecific binding molecule may be linked to one of a variety of nonproteinaceous polymers, e.g., polyethylene glycol, polypropylene glycol, polyoxyalkylenes, or copolymers of polyethylene glycol and polypropylene glycol.
  • nonproteinaceous polymers e.g., polyethylene glycol, polypropylene glycol, polyoxyalkylenes, or copolymers of polyethylene glycol and polypropylene glycol.
  • the bispecific binding molecule also 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.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • macroemulsions for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules.
  • the bispecific binding molecules disclosed herein may also be formulated as immunoliposomes.
  • a “liposome” is a small vesicle composed of various types of lipids, phospholipids and/or surfactant which is useful for delivery of a drug to a mammal.
  • the components of the liposome are commonly arranged in a bilayer formation, similar to the lipid arrangement of biological membranes.
  • Liposomes containing the antibody are prepared by methods known in the art, such as described in Epstein et al., Proc. Natl. Acad. Sci. USA, 82: 3688 (1985); Hwang et al. , Proc. Natl Acad. Sci. USA, 77: 4030 (1980); U. S. Pat. Nos.
  • Liposomes with enhanced circulation time are disclosed in U. S. Patent No. 5,013, 556.
  • Particularly useful liposomes can be generated by the reverse phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter.
  • Fab' fragments of the antibody of the present invention can be conjugated to the liposomes as described in Martin et al. J. Biol. Chem. 257: 286-288 (1982) via a disulfide interchange reaction.
  • a chemotherapeutic agent is optionally contained within the liposome. See Gabizon et al. J. National Cancer Inst. 81 (19) 1484 (1989).
  • the bispecific binding molecule can be produced intracellular ⁇ , in the periplasmic space, or directly secreted into the medium. If the bispecific binding molecule is produced intracellularly, as a first step, the particulate debris, either host cells or lysed fragments, are removed, for example, by centrifugation or ultrafiltration. Carter et al., Bio/Technology 10: 163-167 (1992) describe a procedure for isolating antibodies which are secreted to the periplasmic space of E coli.
  • the bispecific binding molecule composition prepared from the cells can be purified using, for example, hydroxylapatite chromatography, gel electrophoresis, dialysis, and affinity chromatography, with affinity chromatography being the preferred purification technique.
  • the present invention relates to a nucleic acid sequence encoding a bispecific binding molecule of the invention.
  • Said nucleic acid molecule is preferably comprised in a vector which is preferably comprised in a host cell.
  • Said host cell is, e.g. after transformation or transfection with the nucleic acid sequence of the invention, capable of expressing the bispecific binding molecule.
  • the nucleic acid molecule is operatively linked with control sequences.
  • the term "host cell” is intended to refer to a cell into which a nucleic acid encoding the bispecific binding molecule of the invention is introduced by way of transformation, transfection and the like. It should be understood that such terms refer not only to the particular subject cell but to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
  • the term "expression” includes any step involved in the production of a bispecific binding molecule of the invention including, but not limited to, transcription, post- transcriptional modification, translation, post-translational modification, and secretion.
  • control sequences refers to DNA sequences necessary for the expression of an operably linked coding sequence in a particular host organism.
  • the control sequences that are suitable for prokaryotes include a promoter, optionally an operator sequence, and a ribosome binding site.
  • Eukaryotic cells are known to utilize promoters, polyadenylation signals, and enhancers.
  • Nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid sequence.
  • DNA for a presequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide;
  • a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or
  • a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation.
  • "operably linked” means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading phase. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice.
  • Suitable host cells include prokaryotes and eukaryotic host cells including yeasts, fungi, insect cells and mammalian cells.
  • the bispecific binding molecule of the invention can be produced in bacteria.
  • the bispecific binding molecule of the invention preferably the bispecific binding molecule is isolated from the E. coli cell paste in a soluble fraction and can be purified through, e.g., affinity chromatography and/or size exclusion. Final purification can be carried out similar to the process for purifying antibody expressed e. g, in CHO cells.
  • eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for the bispecific binding molecule of the invention.
  • Saccharomyces cerevisiae or common baker's yeast, is the most commonly used among lower eukaryotic host microorganisms.
  • Kluyveromyces hosts such as, e. g. , K. lactis, K. fragilis (ATCC 12424), K. bulgaricus (ATCC 16045), K. wickeramii (ATCC 24178), K.
  • waltii ATCC 56500
  • K. drosophilarum ATCC 36906
  • K. thermotolerans K. marxianus
  • yarrowia EP 402 226
  • Pichia pastoris EP 183 070
  • Candida Trichoderma reesia
  • Neurospora crassa Schwanniomyces such as Schwanniomyces occidentalis
  • filamentous fungi such as, e.g., Neurospora, Penicillium, Tolypocladium, and Aspergillus hosts such as A. nidulans and A. niger.
  • Suitable host cells for the expression of glycosylated bispecific binding molecule of the invention are derived from multicellular organisms.
  • invertebrate cells include plant and insect cells.
  • Numerous baculoviral strains and variants and corresponding permissive insect host cells from hosts such as Spodoptera frugiperda (caterpillar), Aedes aegypti (mosquito), Aedes albopictus (mosquito), Drosophila melanogaster (fruit fly), and Bombyx mori have been identified.
  • a variety of viral strains for transfection are publicly available, e. g.
  • the L-1 variant of Autographa californica NPV and the Bm-5 strain of Bombyx mori NPV, and such viruses may be used as the virus herein according to the present invention, particularly for transfection of Spodoptera frugiperda cells.
  • Plant cell cultures of cotton, corn, potato, soybean, petunia, tomato, Arabidopsis and tobacco can also be utilized as hosts.
  • Cloning and expression vectors useful in the production of proteins in plant cell culture are known to those of skill in the art. See e.g. Hiatt et al., Nature (1989) 342: 76-78, Owen et al. (1992) Bio/Technology 10: 790-794, Artsaenko et al. (1995) The Plant J 8: 745-750, and Fecker et al. (1996) Plant Mol Biol 32: 979-986.
  • vertebrate cells have been greatest in vertebrate cells, and propagation of vertebrate cells in culture (tissue culture) has become a routine procedure.
  • useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651 ) ; human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, Graham et al. , J. Gen Virol. 36 : 59 (1977)); baby hamster kidney cells (BHK, ATCC CCL 10); Chinese hamster ovary cells/-DHFR (CHO, Urlaub et al. , Proc. Natl. Acad. Sci.
  • COS-7 monkey kidney CV1 line transformed by SV40
  • human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, Graham et al. , J. Gen Virol. 36 : 59 (1977)
  • baby hamster kidney cells BHK, ATCC CCL 10
  • mice Sertoli cells TM4, Mather, Biol. Reprod. 23: 243- 251 (1980)); monkey kidney cells (CVI ATCC CCL 70); African green monkey kidney cells (VERO-76, ATCC CRL1587) ; human cervical carcinoma cells (HELA, ATCC CCL 2); canine kidney cells (MDCK, ATCC CCL 34); buffalo rat liver cells (BRL 3A, ATCC CRL 1442); human lung cells (W138, ATCC CCL 75); human liver cells (Hep G2,1413 8065); mouse mammary tumor (MMT 060562, ATCC CCL5 1 ); TRI cells (Mather et al., Annals N. Y Acad. Sci.
  • the bispecific binding molecule of the invention can be produced intracellularly, in the periplasmic space, or directly secreted into the medium. If the bispecific binding molecule is produced intracellularly, as a first step, the particulate debris, either host cells or lysed fragments, are removed, for example, by centrifugation or ultrafiltration. Carter et al., Bio/Technology 10: 163-167 (1992) describe a procedure for isolating antibodies which are secreted to the periplasmic space of E coli.
  • cell paste is thawed in the presence of sodium acetate (pH 3.5), EDTA, and phenylmethylsulfonylfluoride (PMSF) over about 30 min.
  • PMSF phenylmethylsulfonylfluoride
  • Cell debris can be removed by centrifugation.
  • supernatants from such expression systems are generally first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit.
  • a protease inhibitor such as PMSF may be included in any of the foregoing steps to inhibit proteolysis and antibiotics may be included to prevent the growth of adventitious contaminants.
  • the bispecific binding molecule of the invention prepared from the host cells can be purified using, for example, hydroxylapatite chromatography, gel electrophoresis, dialysis, and affinity chromatography, with affinity chromatography being the preferred purification technique.
  • the matrix to which the affinity ligand is attached is most often agarose, but other matrices are available. Mechanically stable matrices such as controlled pore glass or poly (styrenedivinyl) benzene allow for faster flow rates and shorter processing times than can be achieved with agarose. Where the bispecific binding molecule of the invention comprises a CH3 domain, the Bakerbond ABXMresin (J. T. Baker, Phillipsburg, NJ) is useful for purification.
  • compositions comprising a bispecific binding molecule of the invention, or produced according to the process of the invention.
  • said composition is a pharmaceutical composition.
  • pharmaceutical composition relates to a composition for administration to a patient, preferably a human patient.
  • the particular preferred pharmaceutical composition of this invention comprises the bispecific binding molecule of the invention.
  • the pharmaceutical composition comprises suitable formulations of carriers, stabilizers and/or excipients.
  • the pharmaceutical composition comprises a composition for parenteral, transdermal, intraluminal, intraarterial, intrathecal and/or intranasal administration or by direct injection into tissue.
  • said composition is administered to a patient via infusion or injection.
  • Administration of the suitable compositions may be effected by different ways, e.g., by intravenous, intraperitoneal, subcutaneous, intramuscular, topical or intradermal administration.
  • the present invention provides for an uninterrupted administration of the suitable composition.
  • uninterrupted, i.e. continuous administration may be realized by a small pump system worn by the patient for metering the influx of therapeutic agent into the body of the patient.
  • the pharmaceutical composition comprising the bispecific binding molecule of the invention can be administered by using said pump systems.
  • Such pump systems are generally known in the art, and commonly rely on periodic exchange of cartridges containing the therapeutic agent to be infused.
  • the continuous or uninterrupted administration of these bispecific binding molecules of the invention may be intravenuous or subcutaneous by way of a fluid delivery device or small pump system including a fluid driving mechanism for driving fluid out of a reservoir and an actuating mechanism for actuating the driving mechanism.
  • Pump systems for subcutaneous administration may include a needle or a cannula for penetrating the skin of a patient and delivering the suitable composition into the patient's body. Said pump systems may be directly fixed or attached to the skin of the patient independently of a vein, artery or blood vessel, thereby allowing a direct contact between the pump system and the skin of the patient.
  • the pump system can be attached to the skin of the patient for 24 hours up to several days.
  • the pump system may be of small size with a reservoir for small volumes. As a non-limiting example, the volume of the reservoir for the suitable pharmaceutical composition to be administered can be between 0.1 and 50 ml.
  • the continuous administration may be transdermal by way of a patch worn on the skin and replaced at intervals.
  • a patch worn on the skin worn on the skin and replaced at intervals.
  • patch systems for drug delivery suitable for this purpose. It is of note that transdermal administration is especially amenable to uninterrupted administration, as exchange of a first exhausted patch can advantageously be accomplished simultaneously with the placement of a new, second patch, for example on the surface of the skin immediately adjacent to the first exhausted patch and immediately prior to removal of the first exhausted patch. Issues of flow interruption or power cell failure do not arise.
  • compositions may further comprise a pharmaceutically acceptable carrier.
  • suitable pharmaceutical carriers include solutions, e.g. phosphate buffered saline solutions, water, emulsions, such as oil/water emulsions, various types of wetting agents, sterile solutions, liposomes, etc.
  • Compositions comprising such carriers can be formulated by well-known conventional methods.
  • Formulations can comprise carbohydrates, buffer solutions, amino acids and/or surfactants.
  • Carbohydrates may be non-reducing sugars, preferably trehalose, sucrose, octasulfate, sorbitol or xylitol.
  • pharmaceutically acceptable carrier means any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, compatible with pharmaceutical administration.
  • solvents dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, compatible with pharmaceutical administration.
  • the use of such media and agents for pharmaceutically active substances is well known in the art.
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed and include: additional buffering agents; preservatives; co-solvents; antioxidants, including ascorbic acid and methionine; chelating agents such as EDTA; metal complexes (e.g., Zn-protein complexes); biodegradable polymers, such as polyesters; salt- forming counterions, such as sodium, polyhydric sugar alcohols; amino acids, such as alanine, glycine, asparagine, 2-phenylalanine, and threonine; sugars or sugar alcohols, such as trehalose, sucrose, octasulfate, sorbitol or xylitol stachyose, mannose, sorbose, xylose, ribose, myoinisitose, galactose, lactitol, ribitol, myoinisitol, galacti
  • Such formulations may be used for continuous administrations which may be intravenuous or subcutaneous with and/or without pump systems.
  • Amino acids may be charged amino acids, preferably lysine, lysine acetate, arginine, glutamate and/or histidine.
  • Surfactants may be detergents, preferably with a molecular weight of >1 .2 KD and/or a polyether, preferably with a molecular weight of >3 KD.
  • Non-limiting examples for preferred detergents are Tween 20, Tween 40, Tween 60, Tween 80 or Tween 85.
  • Non-limiting examples for preferred polyethers are PEG 3000, PEG 3350, PEG 4000 or PEG 5000.
  • Buffer systems used in the present invention can have a preferred pH of 5-9 and may comprise citrate, succinate, phosphate, histidine and acetate.
  • compositions of the present invention can be administered to the subject at a suitable dose which can be determined e.g. by dose escalating studies by administration of increasing doses of the polypeptide of the invention exhibiting cross-species specificity described herein to non-chimpanzee primates, for instance macaques.
  • a suitable dose which can be determined e.g. by dose escalating studies by administration of increasing doses of the polypeptide of the invention exhibiting cross-species specificity described herein to non-chimpanzee primates, for instance macaques.
  • the polypeptide of the invention exhibiting cross-species specificity described herein can be advantageously used in identical form in preclinical testing in non-chimpanzee primates and as drug in humans.
  • These compositions can also be administered in combination with other proteinaceous and non-proteinaceous drugs.
  • These drugs may be administered simultaneously with the composition comprising the polypeptide of the invention as defined herein or separately before or after administration of said polypeptide in timely defined intervals and doses.
  • the dosage regimen will be determined by the attending physician and clinical factors. As is well known in the medical arts, dosages for any one patient depend upon many factors, including the patient's size, body surface area, age, the particular compound to be administered, sex, time and route of administration, general health, and other drugs being administered concurrently.
  • Preparations for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, and emulsions.
  • non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate.
  • Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media.
  • Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's, or fixed oils.
  • Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and the like. Preservatives and other additives may also be present such as, for example, antimicrobials, anti-oxidants, chelating agents, inert gases and the like.
  • the composition of the present invention might comprise proteinaceous carriers, like, e.g., serum albumin or immunoglobulin, preferably of human origin. It is envisaged that the composition of the invention might comprise, in addition to the polypeptide of the invention defined herein, further biologically active agents, depending on the intended use of the composition.
  • Such agents might be drugs acting on the gastrointestinal system, drugs acting as cytostatica, drugs preventing hyperurikemia, drugs inhibiting immunoreactions (e.g. corticosteroids), drugs modulating the inflammatory response, drugs acting on the circulatory system and/or agents such as cytokines known in the art. It is also envisaged that the bispecific binding molecule of the present invention is applied in a co-therapy, i.e., in combination with another anti-cancer medicament. [0101] .
  • the biological activity of the pharmaceutical composition defined herein can be determined for instance by cytotoxicity assays, as described in the following examples, in WO 99/54440 or by Schlereth et al. (Cancer Immunol. Immunother. 20 (2005), 1 - 12).
  • Effectiveacy refers to the response to therapy by the pharmaceutical composition of the invention, using e.g. standardized NCI response criteria.
  • the success or in vivo efficacy of the therapy using a pharmaceutical composition of the invention refers to the effectiveness of the composition for its intended purpose, i.e. the ability of the composition to cause its desired effect, i.e. depletion of pathologic cells, e.g. tumor cells.
  • the in vivo efficacy may be monitored by established standard methods for the respective disease entities including, but not limited to white blood cell counts, differentials, Fluorescence Activated Cell Sorting, bone marrow aspiration. In addition, various disease specific clinical chemistry parameters and other established standard methods may be used.
  • positron-emission tomography scanning white blood cell counts, differentials, Fluorescence Activated Cell Sorting, bone marrow aspiration, lymph node biopsies/histologies, and various lymphoma specific clinical chemistry parameters (e.g. lactate dehydrogenase) and other established standard methods may be used.
  • pharmacokinetic profile of the drug candidate i.e. a profile of the pharmacokinetic parameters that affect the ability of a particular drug to treat a given condition
  • Pharmacokinetic parameters of the drug influencing the ability of a drug for treating a certain disease entity include, but are not limited to: half-life, volume of distribution, hepatic first-pass metabolism and the degree of blood serum binding.
  • the efficacy of a given drug agent can be influenced by each of the parameters mentioned above.
  • Half-life means the time where 50% of an administered drug are eliminated through biological processes, e.g. metabolism, excretion, etc.
  • hepatic first-pass metabolism is meant the propensity of a drug to be metabolized upon first contact with the liver, i.e. during its first pass through the liver.
  • Volume of distribution means the degree of retention of a drug throughout the various compartments of the body, like e.g. intracellular and extracellular spaces, tissues and organs, etc. and the distribution of the drug within these compartments.
  • “Degree of blood serum binding” means the propensity of a drug to interact with and bind to blood serum proteins, such as albumin, leading to a reduction or loss of biological activity of the drug.
  • Pharmacokinetic parameters also include bioavailability, lag time (Tlag), Tmax, absorption rates, more onset and/or Cmax for a given amount of drug administered.
  • Bioavailability means the amount of a drug in the blood compartment.
  • “Lag time” means the time delay between the administration of the drug and its detection and measurability in blood or plasma.
  • Tmax is the time after which maximal blood concentration of the drug is reached
  • Cmax is the blood concentration maximally obtained with a given drug.
  • the time to reach a blood or tissue concentration of the drug which is required for its biological effect is influenced by all parameters.
  • Pharmacokinetic parameters of bispecific single chain antibodies exhibiting cross-species specificity which may be determined in preclinical animal testing in non-chimpanzee primates as outlined above, are also set forth e.g. in the publication by Schlereth et al. (Cancer Immunol. Immunother. 20 (2005), 1 - 12).
  • toxicity refers to the toxic effects of a drug manifested in adverse events or severe adverse events. These side events might refer to a lack of tolerability of the drug in general and/or a lack of local tolerance after administration. Toxicity could also include teratogenic or carcinogenic effects caused by the drug.
  • safety means the administration of a drug without inducing severe adverse events directly after administration (local tolerance) and during a longer period of application of the drug.
  • Safety can be evaluated e.g. at regular intervals during the treatment and follow-up period. Measurements include clinical evaluation, e.g. organ manifestations, and screening of laboratory abnormalities. Clinical evaluation may be carried out and deviations to normal findings recorded/coded according to NCI-CTC and/or MedDRA standards.
  • Organ manifestations may include criteria such as allergy/immunology, blood/bone marrow, cardiac arrhythmia, coagulation and the like, as set forth e.g. in the Common Terminology Criteria for adverse events v3.0 (CTCAE).
  • Laboratory parameters which may be tested include for instance haematology, clinical chemistry, coagulation profile and urine analysis and examination of other body fluids such as serum, plasma, lymphoid or spinal fluid, liquor and the like.
  • Safety can thus be assessed e.g. by physical examination, imaging techniques (i.e. ultrasound, x-ray, CT scans, Magnetic Resonance Imaging (MRI), other measures with technical devices (i.e. electrocardiogram), vital signs, by measuring laboratory parameters and recording adverse events.
  • imaging techniques i.e. ultrasound, x-ray, CT scans, Magnetic Resonance Imaging (MRI), other measures with technical devices (i.e. electrocardiogram), vital signs, by measuring laboratory parameters and recording adverse events.
  • MRI Magnetic Resonance Imaging
  • adverse events in non-chimpanzee primates in the uses and methods according to the invention may be examined by histopathological and/or histochemical methods.
  • the term “effective dose” or “effective dosage” is defined as an amount sufficient to achieve or at least partially achieve the desired effect.
  • therapeutically effective dose is defined as an amount sufficient to cure or at least partially arrest the disease and its complications in a patient already suffering from the disease. Amounts effective for this use will depend upon the severity of the infection and the general state of the subject's own immune system.
  • patient includes human and other mammalian subjects that receive either prophylactic or therapeutic treatment.
  • the term "effective and non-toxic dose” as used herein refers to a tolerable dose of an inventive bispecific binding molecule which is high enough to cause depletion of pathologic cells, tumor elimination, tumor shrinkage or stabilization of disease without or essentially without major toxic effects. Such effective and non-toxic doses may be determined e.g. by dose escalation studies described in the art and should be below the dose inducing severe adverse side events (dose limiting toxicity, DLT).
  • the appropriate dosage, or therapeutically effective amount, of the bispecific binding molecule of the invention will depend on the condition to be treated, the severity of the condition, prior therapy, and the patient's clinical history and response to the therapeutic agent.
  • the proper dose can be adjusted according to the judgment of the attending physician such that it can be administered to the patient one time or over a series of administrations.
  • the pharmaceutical composition can be administered as a sole therapeutic or in combination with additional therapies such as anti-cancer therapies as needed.
  • compositions of this invention are particularly useful for parenteral administration, i.e., subcutaneously, intramuscularly, intravenously, intra-articular and/or intra-synovial.
  • Parenteral administration can be by bolus injection or continuous infusion.
  • the lyophilized material is first reconstituted in an appropriate liquid prior to administration.
  • the lyophilized material may be reconstituted in, e.g., bacteriostatic water for injection (BWFI), physiological saline, phosphate buffered saline (PBS), or the same formulation the protein had been in prior to lyophilization.
  • BWFI bacteriostatic water for injection
  • PBS phosphate buffered saline
  • the bispecific binding molecule of the invention or produced by a process of the invention is used in the prevention, treatment or amelioration of a disease selected from a proliferative disease, a tumorous disease, or an immunological disorder.
  • An alternative embodiment of the invention provides a method for the treatment or amelioration of a disease selected from a proliferative disease, a tumorous disease, or an immunological disorder comprising the step of administering to a patient in the need thereof the bispecific binding molecule of the invention or produced by a process of the invention.
  • treatment refers to both therapeutic treatment and prophylactic or preventative measures.
  • Treatment includes the application or administration of the formulation to the body, an isolated tissue, or cell from a patient who has a disease/disorder, a symptom of a disease/disorder, or a predisposition toward a disease/disorder, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve, or affect the disease, the symptom of the disease, or the predisposition toward the disease.
  • kits comprising a bispecific binding molecule of the invention, a nucleic acid molecule of the invention, a vector of the invention, or a host cell of the invention.
  • the kit may comprise one or more vials containing the bispecific binding molecule and instructions for use.
  • the kit may also contain means for administering the bispecific binding molecule of the present invention such as a syringe, pump, infusor or the like.
  • the present invention relates to the use of amino acid residues 170 to 222 (epitope cluster 4) and/or 78 to 138 (epitope cluster 2) of the human 5T4 sequence as depicted in SEQ ID NO: 2 for the generation of an antibody, preferably a bispecific binding molecule having preferably the capacity to bind to human 5T4 and CD3 as described herein.
  • the present invention relates to a method for the generation of an antibody, preferably bispecific binding molecule, comprising (a) immunizing an animal with a protein comprising amino acid residues 170 to 222 (epitope cluster 4) and/or 78 to 138 (epitope cluster 2) of the human 5T4 sequence as depicted in SEQ ID NO: 2, (b) obtaining said antibody, and optionally (c) converting said antibody into a bispecific binding molecule having preferably the capacity to bind to human 5T4 and CD3 as described herein.
  • the present invention further contemplates a fragment of 5T4 consisting of of amino acid residues 170 to 222 (epitope cluster 4) or amino acids 78 to 138 (epitope cluster 2) of the human 5T4 sequence as depicted in SEQ ID NO: 2.
  • Figure 1 Hypothetical structure of 5T4. SRR: serine-rich repeat (brown); LRR: leucine-rich repeat (blue); cysteines are depicted by yellow circles; potential glycosylation sites are marked by asterisks. [0128] .
  • Figure 2 Structure model of 5T4 with highlighted segments exchanged in chimeric constructs as designed for epitope clustering. LLR: Leucin rich repeat. SRR: Serine rich repeat. Asterisks: position of carbohydrates. [0129] .
  • Figure 3 Epitope clustering with chimeric 5T4 constructs - Sequence alignment of human and murine 5T4.
  • Figure 4 Murine and human 5T4 as well as seven chimeric human-murine 5T4 constructs expressed on the surface of CHO cells as shown by flow cytometry.
  • the expression of human 5T4 and the chimeric 5T4 molecules hu 5T4 E1 mu, hu 5T4 E2 mu, hu 5T4 E3 mu and hu 5T4 E6 mu on CHO was detected with a monoclonal anti-human 5T4 antibody.
  • the expression of hu 5T4 E4 mu, hu 5T4 E5 mu and hu 5T4 E7 mu was detected with a polyclonal anti-human 5T4 antibody.
  • Murine 5T4 expression was detected with a polyclonal anti-mouse 5T4-antibody. Bound monoclonal antibody was detected with an anti- mouse Fc-gamma-specific antibody conjugated to phycoerythrin. Bound polyclonal antibodies were detected with a PE-labeled anti-sheep antibody.
  • Figure 5 Determination of binding constants of bispecific antibodies on human and macaque 5T4 and on human and macaque CD3 using the Biacore system. Antigen was immobilized in low to intermediate density (100 RU) on CM5 chip. Dilutions of bispecific antibodies were floated over the chip surface and binding determined using BiaEval Software. Respective on- and off-rates and the resulting binding constant (KD) of the respective bispecific antibodies are depicted below every graph. Numbers in brackets indicate imprecise off-rate and KD calculation due to a biphasic off-rate of the respective 5T4 bispecific antibody.
  • FIG. 6 Determination of binding constants of bispecific antibodies directed against epitope 4 on 5T4 antigen positive cells. Cells were incubated with triplicate dilution series of respective bispecific antibodies. Detection was carried out in a strictly monovalent manner using anti-His Fab/Alexa488 as detection antibody. 5T4 antigen positive cells used: NCI-N87 as naturally human 5T4 expressing cell line (left column), transfected CHO cells expressing recombinant 5T4 (middle column) and transfected CHO cells expressing recombinant 5T4 (right column). Hyperbole graphs are shown with Scatchard analysis incorporated in every graph. Respective KD values are included in each diagram.
  • Figure 7 Determination of binding constants of bispecific antibodies directed against epitope 7 on 5T4 antigen positive cells. Cells were incubated with triplicate dilution series of respective bispecific antibodies. Detection was carried out in a strictly monovalent manner using anti-His Fab/Alexa488 as detection antibody. 5T4 antigen positive cells used: NCI-N87 as naturally human 5T4 expressing cell line (left column), transfected CHO cells expressing recombinant 5T4 (middle column) and transfected CHO cells expressing recombinant 5T4 (right column). Hyperbole graphs are shown with Scatchard analysis incorporated in every graph. Respective KD values are included in each diagram.
  • Figure 8 Determination of binding constants of bispecific antibodies directed against epitope E2/4 on 5T4 antigen positive cells. Cells were incubated with triplicate dilution series of respective bispecific antibodies. Detection was carried out in a strictly monovalent manner using anti-His Fab/Alexa488 as detection antibody. 5T4 antigen positive cells used: NCI-N87 as naturally human 5T4 expressing cell line (left column), transfected CHO cells expressing recombinant 5T4 (middle column) and transfected CHO cells expressing recombinant 5T4 (right column). Hyperbole graphs are shown with Scatchard analysis incorporated in every graph. Respective KD values are included in each diagram.
  • Figure 9 Cytotoxic activity of 5T4 bispecific antibodies as measured in an 18-hour 51 chromium release assay. Effector cells: stimulated enriched human CD8 T cells. Target cells: 5T4-positive human tumor cell line HCT1 16. Effector to target cell (E:T) ratio: 10:1. Ranking intervals based on current bispecific antibody experience for cytotoxic activity with stimulated enriched human CD8 T cells (EC50) ⁇ 10 pg/ml (1 ), [10-100 pg/ml] (2), [100-1000 pg/ml] (3), [1000-5000 pg/ml] (4), ⁇ 5000 pg/ml pg/ml (5).
  • FIG. 10 Cytotoxic activity of 5T4 bispecific antibodies as measured in a 48 hour FACS-based cytotoxicity assay. Effector cells: unstimulated human PBMC. Target cells: CHO cells transfected with human 5T4. Effector to target cell (E:T)-ratio: 10:1. Ranking intervals based on current bispecific antibody experience for cytotoxic activity with unstimulated human PBMC (EC50): ⁇ 250 pg/ml (1 ), [250-1000 pg/ml] (2), [1000-5000 pg/ml] (3), [5000-20000 pg/ml] (4), ⁇ 20000 pg/ml pg/ml (5). [0137] .
  • Figure 11 Cytotoxic activity of 5T4 bispecific antibodies as measured in a 48 hour FACS-based cytotoxicity assay. Effector cells: unstimulated human PBMC. Target cells: 5T4- positive human tumor cell line NCI-N87. Effector to target cell (E:T)-ratio: 10:1. Ranking intervals based on current bispecific antibody experience for cytotoxic activity with unstimulated human PBMC (EC50): ⁇ 250 pg/ml (1 ), [250-1000 pg/ml] (2), [1000-5000 pg/ml] (3), [5000-20000 pg/ml] (4), ⁇ 20000 pg/ml pg/ml (5).
  • Figure 12 Cytotoxic activity of 5T4 bispecific antibodies as measured in a 48 hour FACS-based cytotoxicity assay. Effector cells: macaque T cell line 41 19LnPx. Target cells: CHO cells transfected with macaque 5T4. Effector to target cell (E:T) ratio: 10:1 . Ranking intervals based on current bispecific antibody experience for cytotoxic activity with the macaque T cell line 41 19LnPx (EC50): ⁇ 250 pg/ml (1 ), [250-1000 pg/ml] (2), [1000-5000 pg/ml] (3), [5000-20000 pg/ml] (4), ⁇ 20000 pg/ml pg/ml (5). [0139] . Figure 13: Anti-tumor activity of bispecific antibodies in a HCT-1 16 advanced stage tumor model
  • a 5T4-CD3 bispecific antibody (5T4-12 HL x CD3 HL) (0.1 , 0.5 and 2.5 mg/kg/d) was administered by intravenous bolus injection into the lateral tail vein for 14 days.
  • Control animals (Group 2) and animals without T cell injection (Group 1 ) received the vehicle by intravenous bolus injection into the lateral tail vein for 14 days. Growth of tumors was determined by external caliper measurements, and tumor volumes were calculated using a standard hemi-ellipsoid formula: (length [mm] x width [mm] 2 )/2.
  • Figure 14 Anti-tumor activity of bispecific antibodies in a in a NCI-N87 advanced stage tumor model
  • a 5T4-CD3 bispecific antibody (5T4-12 HL x CD3 HL) (0.1 , 0.5 and 2.5 mg/kg/d) was administered by intravenous bolus injection into the lateral tail vein for 21 days.
  • Control animals (Group 2) and animals without T cell injection (Group 1 ) received the vehicle by intravenous bolus injection into the lateral tail vein for 21 days. Growth of tumors was determined by external caliper measurements, and tumor volumes were calculated using a standard hemi-ellipsoid formula: (length [mm] x width [mm] 2 )/2.
  • the amino acid sequence of the respective epitope domains of human 5T4 was changed to the murine sequence.
  • the cDNA constructs were cloned into the mammalian expression vector pEF-DHFR and stably transfected into CHO cells.
  • the expression of human 5T4 and chimeric 5T4 molecules hu 5T4 E1 mu, hu 5T4 E2 mu, hu 5T4 E3 mu and hu 5T4 E6 mu on CHO was verified in a FACS assay using a monoclonal anti-human 5T4 antibody (R&D Systems #MAB49751 ).
  • the expression of hu 5T4 E4 mu, hu 5T4 E5 mu and hu 5T4 E7 mu was detected with a polyclonal anti-human 5T4 antibody (R&D Systems #AF4975).
  • Mu 5T4 expression was demonstrated with a polyclonal anti-mouse 5T4-antibody (R&D Systems #AF5049).
  • the used concentration of the 5T4 antibodies was 5 ⁇ g ml.
  • Bound monoclonal antibodies were detected with an anti-mouse Fc gamma-specific antibody conjugated to R-phycoerythrin (1 : 100; Dianova #1 15-1 16-071 ).
  • Bound polyclonal antibodies were detected with 5 ⁇ g ml of a PE-labeled anti-sheep antibody (Santa Cruz Biotechnology #sc-3746). All antibodies were diluted in PBS with 2% FCS. As negative control, cells were incubated with PBS/2% FCS instead of the first antibody.
  • Clones of the expression plasmids with sequence-verified nucleotide sequences were used for transfection and protein expression in the FreeStyle 293 Expression System (Invitrogen GmbH, Düsseldorf, Germany) according to the manufacturer's protocol. Supernatants containing the expressed proteins were obtained, cells were removed by centrifugation and the supernatants were stored at -20 C.
  • Clones of the expression plasmids with sequence-verified nucleotide sequences were transfected into DHFR deficient CHO cells for eukaryotic expression of the constructs.
  • Eukaryotic protein expression in DHFR deficient CHO cells was performed as described by Kaufman R.J. (1990) Methods Enzymol. 185, 537-566.
  • Gene amplification of the constructs was induced by increasing concentrations of methotrexate (MTX) to a final concentration of 20 nM MTX.
  • MTX methotrexate
  • the cells were grown in roller bottles with nucleoside-free HyQ PF CHO liquid soy medium (with 4.0 mM L-Glutamine with 0.1 % Pluronic F - 68; HyClone) for 7 days before harvest.
  • the cells were removed by centrifugation and the supernatant containing the expressed protein was stored at -20 C.
  • human and macaque 5T4 human and mouse 5T4 sequences as published in GenBank, accession numbers NM_006670 [human]; NM_01 1627 [mouse]
  • coding sequences of human albumin, murine Fcy1 and murine albumin were used for the construction of artificial cDNA sequences encoding soluble fusion proteins of human and macaque 5T4 respectively and human albumin, murine lgG1 Fc and murine albumin respectively as well as soluble proteins comprising only the extracellular domains of 5T4.
  • cDNA fragments were obtained by PCR mutagenesis of the full-length 5T4 cDNAs described above and molecular cloning according to standard protocols.
  • the modified cDNA fragments were designed as to contain first a Kozak site for eukaryotic expression of the constructs followed by the coding sequence of the human and macaque 5T4 proteins respectively comprising the amino acids 1 to 355 corresponding to the signal peptide and extracellular domain of human and macaque 5T4 respectively, followed in frame by the coding sequence of an artificial Ser1 - Gly4-Ser1 -linker, followed in frame by the coding sequence of human serum albumin, followed in frame by the coding sequence of a Flag tag, followed in frame by the coding sequence of a modified histidine tag (SGHHGGHHGGHH) and a stop codon.
  • SGHHGGHHGGHH modified histidine tag
  • the modified cDNA fragments were designed as to contain first a Kozak site for eukaryotic expression of the constructs followed by the coding sequence of the human and macaque 5T4 proteins respectively comprising the amino acids 1 to 355 corresponding to the signal peptide and extracellular domain of human and macaque 5T4 respectively, followed in frame by the coding sequence of an artificial Ser1 -Gly4-Ser1 -linker, followed in frame by the coding sequence of the hinge and Fc gamma portion of murine lgG1 , followed in frame by the coding sequence of a hexahistidine tag and a stop codon.
  • the modified cDNA fragments were designed as to contain first a Kozak site for eukaryotic expression of the constructs followed by the coding sequence of the human and macaque 5T4 proteins respectively comprising the amino acids 1 to 355 corresponding to the signal peptide and extracellular domain of human and macaque 5T4 respectively, followed in frame by the coding sequence of an artificial Ser1 - Gly4-Ser1 -linker, followed in frame by the coding sequence of murine serum albumin, followed in frame by the coding sequence of a Flag tag, followed in frame by the coding sequence of a modified histidine tag (SGHHGGHHGGHH) and a stop codon.
  • SGHHGGHHGGHH modified histidine tag
  • the modified cDNA fragments were designed as to contain first a Kozak site for eukaryotic expression of the constructs followed by the coding sequence of the human and macaque 5T4 proteins respectively comprising the amino acids 1 to 355 corresponding to the signal peptide and extracellular domain of human and macaque 5T4 respectively, followed in frame by the coding sequence of an artificial Ser1 - Gly1 -linker, followed in frame by the coding sequence of a Flag tag, followed in frame by the coding sequence of a modified histidine tag (SGHHGGHHGGHH) and a stop codon.
  • SGHHGGHHGGHH modified histidine tag
  • the cDNA fragments were also designed to introduce restriction sites at the beginning and at the end of the fragments.
  • the introduced restriction sites, EcoRI at the 5' end and Sail at the 3' end, were utilized in the following cloning procedures.
  • the cDNA fragments were cloned via EcoRI and Sail into a plasmid designated pEF-DHFR (pEF-DHFR is described in Griffin et al. Cancer Immunol Immunother 50 (2001 ) 141 -150).
  • the aforementioned procedures were all carried out according to standard protocols (Sambrook, Molecular Cloning; A Laboratory Manual, 3rd edition, Cold Spring Harbour Laboratory Press, Cold Spring Harbour, New York (2001 )).
  • Biacore analysis experiments were performed using recombinant 5T4 fusion proteins with human serum albumin (ALB) to determine 5T4 target binding.
  • ARB human serum albumin
  • recombinant fusion proteins having the N-terminal 27 amino acids of the CD3 epsilon (CD3e) fused to human antibody Fc portion were used. This recombinant protein exists in a human CD3e1 -27 version and in a cynomolgous CD3e version, both bearing the epitope of the CD3 binder in the bispecific antibodies.
  • CM5 Sensor Chips (GE Healthcare) were immobilized with approximately 100 to 150 RU of the respective recombinant antigen using acetate buffer pH4.5 according to the manufacturer's manual.
  • the bispecific antibody samples were loaded in five concentrations: 50 nM, 25 nM, 12.5 nM, 6.25 nM and 3.13 nM diluted in HBS-EP running buffer (GE Healthcare). Flow rate was 30 to 35 ul/min for 3 min, then HBS-EP running buffer was applied for 8 min again at a flow rate of 30 to 35 ul/ml. Regeneration of the chip was performed using 10 mM glycine 0.5 M NaCI pH 2.45. Data sets were analyzed using BiaEval Software ( Figure 5). In general two independent experiments were performed.
  • the cells were resuspended in 150 ⁇ FACS buffer containing 3.5 % formaldehyde, incubated for further 15 min, centrifuged, resuspended in FACS buffer and analyzed using a FACS Cantoll machine and FACS Diva software. Data were generated from two independant sets of experiments. Values were plotted as hyperbole binding curves. Respective Scatchard analysis was calculated to extrapolate maximal binding (Bmax). The concentrations of bispecific antibodies at half-maximal binding were determined reflecting the respective KDs. Values of triplicate measurements were plotted as hyperbolic curves. Maximal binding was determined using Scatchard evaluation and the respective KDs were calculated. Values depicted in table 2 were derived from two independent experiments per 5T4 bispecific antibody. One representative experiment per 5T4 bispecific antibody is shown in Figures 6 - 8.
  • 5T4 bispecific Native human 5T4 Human 5T4 Macaque 5T4 antibody NCI- N87 cells CHO cells CHO cells
  • 5T4-CD3-4 1 .57 ⁇ 0.19 5.49 ⁇ 0.38 5.65 ⁇ 1 .25
  • 5T4-CD3-1 1 0.13 ⁇ 0.14 0.29 ⁇ 0.03 1 .75 ⁇ 0.85
  • Table 2 Affinities (KD) of 5T4 bispecific antibodies from cell based Scatchard analysis (two independent experiments each) The high affinities of the 15 5T4 bispecific antibodies measured by Biacore on recombinant soluble human and macaque 5T4-antigen could be confirmed by Scatchard analysis on CHO cells transfected with human or macaque 5T4. Most importantly, the affinities to native 5T4 expressed on the surface of the human tumor cell line NCI-N87 closely resemble those measured on CHO cells expressing recombinant 5T4 on their surface.
  • Stimulated T cells enriched for CD8 + T cells were obtained as described below.
  • a petri dish (145 mm diameter, Greiner bio-one GmbH, Kremsmijnster) was coated with a commercially available anti-CD3 specific antibody (OKT3, Orthoclone) in a final concentration of 1 ⁇ g ml for 1 hour at 37°C. Unbound protein was removed by one washing step with PBS. 3 - 5 x 10 7 human PBMC were added to the precoated petri dish in 120 ml of RPMI 1640 with stabilized glutamine / 10% FCS / IL-2 20 U/ml (Proleukin®, Chiron) and stimulated for 2 days. On the third day, the cells were collected and washed once with RPMI 1640. IL-2 was added to a final concentration of 20 U/ml and the cells were cultured again for one day in the same cell culture medium as above.
  • a commercially available anti-CD3 specific antibody OKT3, Orthoclone
  • CD8 + cytotoxic T lymphocytes were enriched by depletion of CD4 + T cells and CD56 + NK cells using Dynal-Beads according to the manufacturer ' s protocol.
  • Macaque or human 5T4-transfected CHO target cells were washed twice with PBS and labeled with 1 1 .1 MBq 51 Cr in a final volume of 100 ⁇ RPMI with 50% FCS for 60 minutes at 37°C. Subsequently, the labeled target cells were washed 3 times with 5 ml RPMI and then used in the cytotoxicity assay.
  • the assay was performed in a 96-well plate in a total volume of 200 ⁇ supplemented RPMI (as above) with an E:T ratio of 10:1 .
  • a starting concentration of 0.01 - 1 ⁇ g ml of purified bispecific antibody and threefold dilutions thereof were used.
  • Incubation time for the assay was 18 hours.
  • Cytotoxicity was determined as relative values of released chromium in the supernatant relative to the difference of maximum lysis (addition of Triton-X) and spontaneous lysis (without effector cells). All measurements were carried out in quadruplicates.
  • Measurement of chromium activity in the supernatants was performed in a Wizard 3" gammacounter (Perkin Elmer Life Sciences GmbH, Koln, Germany). Analysis of the results was carried out with Prism 5 for Windows (version 5.0, GraphPad Software Inc., San Diego, California, USA). EC50 values calculated by the analysis program from the sigmoidal dose response curves were used for
  • PBMC Human peripheral blood mononuclear cells
  • PBMC Human peripheral blood mononuclear cells
  • Buffy coats enriched lymphocyte preparations
  • Buffy coats were supplied by a local blood bank and PBMC were prepared on the same day of blood collection.
  • erythrocytes were removed from PBMC via incubation with erythrocyte lysis buffer (155 mM NH 4 CI, 10 mM KHC0 3 , 100 ⁇ EDTA). Platelets were removed via the supernatant upon centrifugation of PBMC at 100 x g.
  • Remaining lymphocytes mainly encompass B and T lymphocytes, NK cells and monocytes.
  • PBMC were kept in culture at 37°C/5% C0 2 in RPMI medium (Gibco) with 10% FCS (Gibco).
  • CD14 MicroBeads and CD56 MicroBeads (20 ⁇ /10 7 cells) were added and incubated for 15 min at 4 - 8°C. The cells were washed with MACS isolation buffer (1 - 2 mL/10 7 cells). After centrifugation (see above), supernatant was discarded and cells resuspended in MACS isolation buffer (500 0 8 cells). CD14/CD56 negative cells were then isolated using LS Columns (Miltenyi Biotec, #130-042-401 ). PBMC w/o CD14+/CD56+ cells were cultured in RPMI complete medium i.e.
  • RPMI1640 Biochrom AG, #FG1215) supplemented with 10% FBS (Biochrom AG, #S01 15), 1 x non essential amino acids (Biochrom AG, #K0293), 10 mM Hepes buffer (Biochrom AG, #L1613), 1 mM sodium pyruvate (Biochrom AG, #L0473) and 100 U/mL penicillin/streptomycin (Biochrom AG, #A2213) at 37°C in an incubator until needed.
  • FBS Biochrom AG, #S01 15
  • 1 x non essential amino acids Biochrom AG, #K0293
  • 10 mM Hepes buffer Biochrom AG, #L1613
  • 1 mM sodium pyruvate Biochrom AG, #L0473
  • 100 U/mL penicillin/streptomycin Biochrom AG, #A2213
  • the fluorescent membrane dye DiOCi 8 (Molecular Probes, #V22886) was used to label human or macaque 5T4-transfected CHO cells or 5T4-expressing human NCI-N87 cells as target cells and distinguish them from effector cells. Briefly, cells were harvested, washed once with PBS and adjusted to 10 6 cell/mL in PBS containing 2 % (v/v) FBS and the membrane dye DiO (5 ⁇ _/10 6 cells). After incubation for 3 min at 37°C, cells were washed twice in complete RPMI medium and the cell number adjusted to 1.25 x 10 5 cells/mL. The vitality of cells was determined using 0.5 % (v/v) isotonic EosinG solution (Roth, #45380).
  • This assay was designed to quantify the lysis of macaque or human 5T4-transfected CHO cells in the presence of serial dilutions of 5T4 bispecific antibodies.
  • Equal volumes of DiO-labeled target cells and effector cells i.e., PBMC w/o CD14 + cells
  • PBMC w/o CD14 + cells PBMC w/o CD14 + cells
  • E:T cell ratio 10:1.
  • 160 ⁇ _ of this suspension were transferred to each well of a 96-well plate.
  • 40 ⁇ _ of serial dilutions of the 5T4 bispecific antibodies and a negative control bispecific (an CD3-based bispecific antibody recognizing an irrelevant target antigen) or RPMI complete medium as an additional negative control were added.
  • the bispecific antibody-mediated cytotoxic reaction proceeded for 48 hours in a 7% C0 2 humidified incubator.
  • PI propidium iodide
  • Target cells were identified as DiO-positive cells. Pl-negative target cells were classified as living target cells. Percentage of cytotoxicity was calculated according to the following formula:
  • n number of events Using GraphPad Prism 5 software (Graph Pad Software, San Diego), the percentage of cytotoxicity was plotted against the corresponding bispecific antibody concentrations. Dose response curves were analyzed with the four parametric logistic regression models for evaluation of sigmoid dose response curves with fixed hill slope and EC50 values were calculated.
  • the macaque T cell line 41 19LnPx (Knappe et al. Blood 95:3256-61 (2000), kindly provided by Prof Fickenscher, Hygiene Institute, Virology, Eriangen-Nuernberg) was used as source of effector cells.
  • Target cell labeling of macaque 5T4-transfected CHO cells and flow cytometry based analysis of cytotoxic activity was performed as described above.
  • the potency of 5T4 bispecific antibodies in redirecting stimulated human effector T cells against the 5T4-positive human tumor cell line HCT1 16 was measured in a 51 - chromium release assay.
  • the potency gap between monomeric and dimeric forms of 5T4 bispecific antibodies was determined in a 51 -chromium release assay using human 5T4-transfected CHO cells as target cells and stimulated human T cells as effector cells.
  • 5T4 bispecific antibodies of the E7 epitope cluster showed poor cytotoxicity against HCT1 16 cells with 2-digit ng/ml EC50 values, although their EC50 values for cytotoxic activity against human 5T4-transfected CHO cells was still within an acceptable 3-digit pg/ml range.
  • the cytotoxic activity of 5T4 bispecific antibodies was also analyzed in a FACS-based cytotoxicity assay using CHO cells transfected with human 5T4 as target cells, and unstimulated human PBMC as effector cells (Figure 10).
  • macaque T cells from cell line 41 19LnPx were induced to efficiently kill macaque 5T4-transfected CHO cells by 5T4 bispecific antibodies of the E2/4 cluster with 1 -digit pg/ml EC50-value, by 5T4 bispecific antibodies of the E4 cluster with 2-digit pg/ml EC50-value and by 5T4 bispecific antibodies of the E7 cluster with 3- to 4- digit pg/ml EC50-values.
  • the human cancer cell lines HCT-1 16 and NCI-N87 were subcutaneously injected in the right dorsal flank of NOD.CB17-Prkdcscid/J mice.
  • mice were randomized into treatment groups.
  • mice were treated with 0.1 , 0.5 and 2.5 mg/kg/day of 5T4-12-CD3 by intravenous bolus injection for 14 (HCT-1 16) and 21 (NCI-N87) days, respectively.
  • Tumors were measured by caliper during the study and progress evaluated by intergroup comparison of tumor volumes.
  • RTV Relative Tumor Volume

Landscapes

  • Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biophysics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cell Biology (AREA)
  • Oncology (AREA)
  • Engineering & Computer Science (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

La présente invention concerne une molécule de liaison bispécifique comprenant un premier et un second domaine de liaison, le premier domaine de liaison pouvant se lier au cluster d'épitopes 4 de T4 et le second domaine de liaison pouvant se lier au complexe du récepteur de CD3 sur les lymphocytes T. En outre, l'invention concerne une séquence d'acides nucléiques codant pour la molécule de liaison bispécifique, un vecteur comprenant ladite séquence d'acides nucléiques et une cellule hôte transformée ou transfectée avec ledit vecteur. En outre, l'invention concerne un procédé de production de la molécule de liaison bispécifique de l'invention, une utilisation médicale de ladite molécule de liaison bispécifique et un kit comprenant ladite molécule de liaison bispécifique.
EP12778970.9A 2011-09-23 2012-09-21 Molécules de liaison bispécifiques pour 5t4 et cd3 Withdrawn EP2758438A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161538716P 2011-09-23 2011-09-23
PCT/EP2012/068673 WO2013041687A1 (fr) 2011-09-23 2012-09-21 Molécules de liaison bispécifiques pour 5t4 et cd3

Publications (1)

Publication Number Publication Date
EP2758438A1 true EP2758438A1 (fr) 2014-07-30

Family

ID=47088803

Family Applications (1)

Application Number Title Priority Date Filing Date
EP12778970.9A Withdrawn EP2758438A1 (fr) 2011-09-23 2012-09-21 Molécules de liaison bispécifiques pour 5t4 et cd3

Country Status (6)

Country Link
EP (1) EP2758438A1 (fr)
JP (1) JP2014533929A (fr)
AU (1) AU2012311492A1 (fr)
CA (1) CA2846432A1 (fr)
MX (1) MX2014003313A (fr)
WO (1) WO2013041687A1 (fr)

Families Citing this family (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6427564B2 (ja) 2013-10-11 2018-11-21 アサナ・バイオサイエンシズ,リミテッド・ライアビリティ・カンパニー タンパク質−ポリマー−薬物共役体
EP3054991B1 (fr) 2013-10-11 2019-04-03 Mersana Therapeutics, Inc. Conjugués de médicament-protéine-polymère
RU2718692C2 (ru) * 2014-05-29 2020-04-13 Мэкроудженикс, Инк. Триспецифичные связывающие молекулы, которые специфически связывают антигены множества злокачественных опухолей, и способы их применения
JP2017520575A (ja) 2014-07-01 2017-07-27 ファイザー・インク 二重特異的ヘテロ二量体ダイアボディおよびその使用
WO2016022939A1 (fr) * 2014-08-08 2016-02-11 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Anticorps monoclonaux humains spécifiques de 5t4 et leurs procédés d'utilisation
WO2016097408A1 (fr) * 2014-12-19 2016-06-23 Biotecnol Limited Protéine de fusion comprenant trois domaines de liaison à 5t4 et cd3
BR112018011781A2 (pt) 2015-12-14 2018-12-04 Macrogenics Inc molécula biespecífica possuindo um ou mais sítios de ligação a epítopo capazes de ligação imunoespecífica a (um) epítopo(s) de pd-1 e um ou mais sítios de ligação a epítopo capazes de ligação imunoespecífica a (um) epítopo(s) de ctla-4, e composição farmacêutica
CR20180365A (es) 2015-12-16 2018-09-28 Amgen Inc PROTEÍNAS DE UNIÓN AL ANTÍGENO BISPECÍFICO DE ANTI-TL1A/ANTI-TNF-a Y SUS USOS
CN110698558B (zh) * 2015-12-24 2021-09-10 凯惠科技发展(上海)有限公司 一种tpbg抗体及其制备方法、其偶联物和应用
JP2019523630A (ja) * 2016-04-22 2019-08-29 アリゲーター・バイオサイエンス・アーベー Cd137に対する新規の二重特異性ポリペプチド
JP2019517539A (ja) 2016-06-07 2019-06-24 マクロジェニクス,インコーポレーテッド 併用療法
US11135307B2 (en) 2016-11-23 2021-10-05 Mersana Therapeutics, Inc. Peptide-containing linkers for antibody-drug conjugates
EP4389226A2 (fr) 2017-02-24 2024-06-26 MacroGenics, Inc. Molécules de liaison bispécifiques capables de se lier à cd137 et à des antigènes tumoraux, et leurs utilisations
WO2018191438A1 (fr) 2017-04-11 2018-10-18 Inhibrx, Inc. Constructions polypeptidiques multispécifiques comprenant une liaison cd3 contrainte et leurs méthodes d'utilisation
CA3070272A1 (fr) 2017-07-20 2019-01-24 Aptevo Research And Development Llc Proteines de liaison a l'antigene se liant a 5t4 et 4-1bb, compositions et procedes associes
EP3717021A1 (fr) 2017-11-27 2020-10-07 Mersana Therapeutics, Inc. Conjugués anticorps-pyrrolobenzodiazépine
KR20200098590A (ko) 2017-12-12 2020-08-20 마크로제닉스, 인크. 이중특이적 cd16-결합 분자 및 질환 치료에서의 그것의 용도
US20220305127A1 (en) 2017-12-21 2022-09-29 Mersana Therapeutics, Inc. Pyrrolobenzodiazepine antibody conjugates
BR112020016485A2 (pt) 2018-02-15 2020-12-15 Macrogenics, Inc. Molécula de ligação, composição farmacêutica e método para o tratamento de uma doença
CA3093745A1 (fr) * 2018-03-12 2019-09-19 Genmab A/S Anticorps
US20210340273A1 (en) * 2018-10-11 2021-11-04 Inhlbrx, inc. 5t4 single domain antibodies and therapeutic compositions thereof
JP2022513400A (ja) 2018-10-29 2022-02-07 メルサナ セラピューティクス インコーポレイテッド ペプチド含有リンカーを有するシステイン操作抗体-薬物コンジュゲート
WO2021048423A1 (fr) 2019-09-12 2021-03-18 Genmab A/S Anticorps bispécifiques se liant à 5t4 et cd3 pour une utilisation dans le traitement du cancer
KR20230157315A (ko) 2021-01-28 2023-11-16 리제너론 파마슈티칼스 인코포레이티드 사이토카인 방출 증후군을 치료하기 위한 조성물 및 방법
WO2023201226A1 (fr) 2022-04-11 2023-10-19 Regeneron Pharmaceuticals, Inc. Compositions et méthodes permettant la destruction de cellule tumorale universelle

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002038612A2 (fr) 2000-11-13 2002-05-16 Oxford Biomedica (Uk) Limited Polypeptide
WO2003002143A1 (fr) 2001-06-28 2003-01-09 Active Biotech Ab Superantigene modifie genetiquement pour therapie humaine
US20060286634A1 (en) 1997-06-04 2006-12-21 Oxford Biomedica (Uk) Limited Vector
WO2007106744A2 (fr) 2006-03-10 2007-09-20 Wyeth Anticorps anti-5t4 et leurs utilisations

Family Cites Families (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
GB8308235D0 (en) 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4544545A (en) 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
JPS6147500A (ja) 1984-08-15 1986-03-07 Res Dev Corp Of Japan キメラモノクロ−ナル抗体及びその製造法
EP0173494A3 (fr) 1984-08-27 1987-11-25 The Board Of Trustees Of The Leland Stanford Junior University Récepteurs chimériques par liaison et expression de l'ADN
GB8422238D0 (en) 1984-09-03 1984-10-10 Neuberger M S Chimeric proteins
US4879231A (en) 1984-10-30 1989-11-07 Phillips Petroleum Company Transformation of yeasts of the genus pichia
GB8607679D0 (en) 1986-03-27 1986-04-30 Winter G P Recombinant dna product
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
GB8610600D0 (en) 1986-04-30 1986-06-04 Novo Industri As Transformation of trichoderma
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
WO1988007089A1 (fr) 1987-03-18 1988-09-22 Medical Research Council Anticorps alteres
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
EP0402226A1 (fr) 1989-06-06 1990-12-12 Institut National De La Recherche Agronomique Vecteurs de transformation de la levure yarrowia
US5013556A (en) 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
CA2103059C (fr) 1991-06-14 2005-03-22 Paul J. Carter Methode de production d'anticorps humanises
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US6214388B1 (en) 1994-11-09 2001-04-10 The Regents Of The University Of California Immunoliposomes that optimize internalization into target cells
KR20050085971A (ko) 1995-04-27 2005-08-29 아브게닉스, 인크. 면역화된 제노마우스 유래의 인간 항체
AU2466895A (en) 1995-04-28 1996-11-18 Abgenix, Inc. Human antibodies derived from immunized xenomice
JP4436457B2 (ja) 1995-08-18 2010-03-24 モルフォシス アイピー ゲーエムベーハー 蛋白質/(ポリ)ペプチドライブラリー
EP2314625B1 (fr) 1996-12-03 2014-05-07 Amgen Fremont Inc. Mammifères transgèniques obtenus par génie génétique contenant des loci des immunoglobulines humaines qui comprennent plusieurs régions de VH et de Vkappa, et anticorps aussi obtenus
JP2002512624A (ja) 1997-05-21 2002-04-23 バイオベーション リミテッド 非免疫原性タンパク質の製造方法
IL138857A0 (en) 1998-04-21 2001-10-31 Micromet Ges For Biomedizinisc Cd19xcd3 specific polypeptides and uses thereof
EP1051432B1 (fr) 1998-12-08 2007-01-24 Biovation Limited Modification de l'immunogenicite de proteines
AU1290001A (en) * 1999-11-18 2001-05-30 Oxford Biomedica (Uk) Limited Antibodies
US20060088522A1 (en) * 2004-09-10 2006-04-27 Wyeth Humanized anti-5T4 antibodies and anti-5T4/calicheamicin conjugates
EP2277918B1 (fr) * 2005-05-13 2012-11-14 Oxford BioMedica (UK) Limited Antigène peptide mhc classe I et II dérivé d'une tumeur antigène 5t4
JP5490714B2 (ja) * 2007-11-28 2014-05-14 メディミューン,エルエルシー タンパク質製剤

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060286634A1 (en) 1997-06-04 2006-12-21 Oxford Biomedica (Uk) Limited Vector
WO2002038612A2 (fr) 2000-11-13 2002-05-16 Oxford Biomedica (Uk) Limited Polypeptide
WO2003002143A1 (fr) 2001-06-28 2003-01-09 Active Biotech Ab Superantigene modifie genetiquement pour therapie humaine
WO2007106744A2 (fr) 2006-03-10 2007-09-20 Wyeth Anticorps anti-5t4 et leurs utilisations

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
FORSBERG G. ET AL.: "IDENTIFICATION OF FRAMEWORK RESIDUES IN A SECRETED RECOMBINANT ANTIBODY FRAGMENT THAT CONTROL PRODUCTION LEVEL AND LOCALIZATION IN ESCHERICHIA COLI", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 272, no. 19, 1997, pages 12430 - 12436, XP002389356
N. HOLE, P.L. STERN: "A 72 kD trophoblast glycoprotein defined by a monoclonal antibody", BR. J. CANCER, vol. 57, 1988, pages 239 - 246, XP002653671 *
See also references of WO2013041687A1 *

Also Published As

Publication number Publication date
AU2012311492A1 (en) 2014-03-06
MX2014003313A (es) 2014-07-09
WO2013041687A1 (fr) 2013-03-28
CA2846432A1 (fr) 2013-03-28
JP2014533929A (ja) 2014-12-18

Similar Documents

Publication Publication Date Title
US20220064336A1 (en) Binding molecules for bcma and cd3
WO2013041687A1 (fr) Molécules de liaison bispécifiques pour 5t4 et cd3
US10752694B2 (en) Binding molecules for BCMA and CD3
US10941207B2 (en) Fusion protein comprising three binding domains to 5T4 and CD3
WO2016135239A1 (fr) Protéine de fusion comprenant trois domaines de liaison à her2

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20140423

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

TPAB Information related to observations by third parties deleted

Free format text: ORIGINAL CODE: EPIDOSDTIPA

TPAC Observations filed by third parties

Free format text: ORIGINAL CODE: EPIDOSNTIPA

17Q First examination report despatched

Effective date: 20150605

TPAC Observations filed by third parties

Free format text: ORIGINAL CODE: EPIDOSNTIPA

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20151016