WO2006020258A2 - Novel tetravalent bispecific antibody - Google Patents

Novel tetravalent bispecific antibody Download PDF

Info

Publication number
WO2006020258A2
WO2006020258A2 PCT/US2005/025472 US2005025472W WO2006020258A2 WO 2006020258 A2 WO2006020258 A2 WO 2006020258A2 US 2005025472 W US2005025472 W US 2005025472W WO 2006020258 A2 WO2006020258 A2 WO 2006020258A2
Authority
WO
WIPO (PCT)
Prior art keywords
antigen
binding protein
binding
diabody
specific
Prior art date
Application number
PCT/US2005/025472
Other languages
French (fr)
Other versions
WO2006020258A3 (en
Inventor
Zhenping Zhu
Original Assignee
Imclone Systems Incorporated
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 Imclone Systems Incorporated filed Critical Imclone Systems Incorporated
Priority to EP05773142A priority Critical patent/EP1786918A4/en
Priority to JP2007521716A priority patent/JP2008512352A/en
Publication of WO2006020258A2 publication Critical patent/WO2006020258A2/en
Publication of WO2006020258A3 publication Critical patent/WO2006020258A3/en

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
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • C07K2317/60Immunoglobulins 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
    • C07K2317/60Immunoglobulins 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
    • C07K2317/626Diabody or triabody
    • 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
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • 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
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • 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
    • C07K2317/77Internalization into the cell
    • 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 is directed to novel tetravalent antibodies, which are preferably bi-specific.
  • the tetravalent bispecific antibodies can be efficiently expressed in prokaryotic and eukaryotic cells, and are useful in therapeutic and diagnostic methods.
  • the invention further relates to combination therapies using the antibodies with anti-angiogenic or anti-neoplastic drugs.
  • Bispecific antibodies are immunoglobulin (Ig)-based molecules that bind to two different epitopes on either the same or distinct antigens. Both laboratory and early clinical studies have demonstrated that BsAb may have significant applications in cancer therapy by either targeting tumor cells with cytotoxic agents such as effector cells, radionuclides, drugs and toxins (Weiner et al. (1997) Cancer Immunol. Immunother. 45:190- 2.; van Spriel et al. (2000) Immuol. Today 21:391-7; Segal et al. (2000) J. Immunol.
  • a diabody is a form of scFv dimer which is constructed with a short linker (5- 10 amino acid residues) between the variable heavy (VH) and the variable light (VL) domains.
  • the short linker prevents intrachain pairing of VH and the VL domains, but allows interchain pairing of VH and the VL domains, such that a bivalent antibody fragment with a molecular size of approximately 55 to 60 kDa is formed.
  • a bispecific diabody can be produced by coexpression of two "cross-over" scFv fragments in which the VH and the VL domains for
  • NYOl 924757 vl each of the two binding sites are present on different polypeptide chains (Holliger et al. (1993); Zhu et al. (1996)). Interchain pairing of the two "cross-over" polypeptide chains results in the formation of a scFv dimer, or bispecific diabody, which is a divalent antibody molecule that is monovalent for each of its target antigens.
  • the invention provides a novel tetravalent bispecific antibody (BsAb) molecule.
  • the BsAb comprises two identical diabodies (DAB), each having two antibody binding sites, preferably non-identical.
  • DAB diabodies
  • One of the diabody chains includes an immunoglobulin hinge region and at least one heavy chain constant domain that is capable of self-association to form a dimer (e.g., a C H 2 and/or a C R 3 domain).
  • Di-diabodies offer several advantages as compared to previously described (BsAbs). Di-diabodies can be produced that are bispecific and bivalent. Di-diabodies have no constraints regarding selection of antigen-binding sites, and Fc constant domains and associated functions can be retained. The proteins are substantially homogeneous and can be produced bacterial, mammalian, or other cells without further processing.
  • bispecific di-diabodies for each of two different antigens are comparable to those of natural antibodies (i.e., full, monospecific divalent IgG molecules naturally produced by a host) for each antigen
  • a single pharmaceutical di-diabody preparation can be used in place of a combination of two monospecific antibodies.
  • the di-diabodies can also be monospecific (i.e., four identical antibody binding sites).
  • the di-diabodies of the invention are efficiently produced in mammalian cells, and can be purified by conventional protein A chromatography in a single step.
  • Di-diabodies of the invention comprise at least one IgG constant domain that is capable of self association, such as, for example, C H 2 or CH3 of IgG, IgA, or IgD, or C H 2, CH3, or CR4 of IgE or IgM. Accordingly, di-diabodies possess the immunological functions associated with such domains that are present, such as Fc receptor binding, complement mediated cytotoxicity (CMC), or antibody dependent cell-mediated cytoxicity (ADCC).
  • CMC complement mediated cytotoxicity
  • ADCC antibody dependent cell-mediated cytoxicity
  • di-diabodies of the invention bind to and block activation of a growth factor receptor such as, but not limited to, VEGFR-I, VEGFR-2,
  • the di-diabodies bind to and block activation of two such receptors, hi other embodiments, di-diabodies are capable of simultaneous binding to a growth factor receptor on a target cell, and to a cell surface antigen of an immune system effector cell.
  • Di-diabodies of the invention are used to neutralize growth factor receptors, and to reduce tumor growth and/or inhibit angiogenesis associated with activation of such receptors. Further, the di-diabodies can be administered with anti-neoplastic agents such as chemotherapeutic agents or radiation.
  • Figure 1 is a schematic diagram depicting expression constructs for various antibody fragments, hi the di-diabody, the first of two scFv polypeptide chains was fused to an IgG hinge region and C H 3 domain.
  • a diabody results from association of the first and second scFv chains.
  • a tetrameric structure is formed by two diabodies associated by the hinge and C H 3 domains.
  • G 4 S a 5 amino-acid linker "GGGGS”.
  • E the 13 amino-acid E-tag.
  • Figure 2 shows SDS-PAGE analysis of the purified antibody fragments.
  • Panel A non-reducing conditions
  • Panel B reducing conditions
  • lane 1 diabody (DAB) pi CIl
  • lane 2 DAB 612
  • lane 3 DAB plCl 1/612
  • lane 4 Di-diabody (B); lane 5, di-diabody (M).
  • molecular weight standards run under non-reducing conditions (from top to bottom: 176.5, 113.7, 80.9, 63.8, 49.5, 37.4, 28.0, and 19.6 kDa, respectively).
  • Figure 3 shows bispecific and dose-dependent binding of a di-diabody specific for KDR and FIt-I .
  • Top panel cross-linking assay using FIt-I -coated support and soluble KDR-AP.
  • Middle and bottom panels various concentrations of antibody fragments were bound to a KDR or FIt-I coated support, then incubated with a secondary HRP conjugated reagent. Following incubation with a peroxidase, OD450nm was read. Data shown represent the mean ⁇ SD of triplicate samples.
  • Figure 4 shows inhibition of binding of KDR and FIt-I to immobilized VEGF by the bispecific antibody fragments.
  • Various concentrations of antibody fragments were incubated with a fixed amount of KDR-AP or FIt-I -AP fusion proteins.
  • Bound KDR-AP or Flt-l-AP was quantified by incubation with a peroxidase substrate and OD405nm measurement. Data shown represent the mean ⁇ SD of triplicate samples.
  • Figure 5 shows inhibition of VEGF-induced migration of human leukemia cells by the bispecific antibody fragments.
  • HEL or U937 cells on supports were incubated with various concentrations of antibodies, then placed into wells containing VEGF. Migrated cells were collected and counted. Data shown are the mean ⁇ SD of triplicate determinations.
  • Figure 6 shows the stability of di-diabody preparations incubated in PBS or mouse serum.
  • the di-diabodies were incubated with PBS or serum at 37 0 C for up to 72 hours.
  • Data are expressed as percentage of antigen binding activity of each antibody preparation retained after 37°C incubation in comparison to that of samples incubated at 4 0 C in PBS.
  • Data shown represent the mean ⁇ SD of triplicate samples.
  • Figure 7 depicts construction and production of a di-diabody having binding sites specific for EGFR and IGFR.
  • Panels (A) and (B) are schematic diagrams showing the expression constructs.
  • L Linker (Arg-Thr-Val-Ala-Ala); H: IgGl hinge region.
  • Purified di- diabody preparations were analyzed by SDS-PAGE under non-reducing (C) or reducing (D) conditions.
  • Lane 1 IMC-11F8 IgG
  • lane 2 the di-diabody.
  • Also shown on the left side are molecular weight standards.
  • Figure 8 shows bispecific and dose-dependent binding of the EGFR x IGFR di-diabody.
  • A Receptor cross-linking assay. Antibody preparations were first incubated with a biotin-labeled IGFR in solution and then transferred to a microtiter plate coated with EGFR, followed by incubation with streptoavidin-HRP to measure the plate-bound biotin activity.
  • B and
  • C Dose-dependent binding of the di-diabody to immobilized EGFR and IGFR.
  • Figure 9 shows antiproliferative activity of the EGFR x IGFR di-diabody.
  • Various amounts of the antibodies were incubated with DiFi cells in culture for 4 days followed by incubation with MTT for 4 h. The plates were washed and incubated with HCl/isopropanol at RT for 10 min, followed by optical density reading at 570nm. Data shown represent the mean ⁇ SD of triplicate samples.
  • Figure 10 shows inhibition of EGF and IGF-stimulated activation of receptors and downstream signaling molecules, Akt and MAPK p44/p42. MCF-7 cells were incubated
  • FIG 11 shows IGFR internalization and degradation in tumor cells.
  • Tumor cells were plated and incubated overnight in serum-free medium.
  • IGF-I, EGF (50 nM), IGF plus EGF (I + E) or various antibodies was then added and incubated at 37°C for up to 4 h.
  • Cells were washed in ice-cold PBS, lysed, and electrophoresed using 4-12% Tris-Glycine gels. Proteins were transferred to nitrocellulose membranes and were detected by Western blotting using antibody C-20 (rabbit anti-IGFR) and IMC-11F8 (human anti-EGFR), followed by an anti-rabbit or anti-human antibody-HRP conjugate. The signals were visualized with the ECL reagent.
  • A The di-diabody induced IGFR degradation in a dose- dependent manner in MCF-7 cells.
  • B The di-diabody (100 nM) induced IGFR degradation as efficiently as EVIC-A12 and the combination of IMC-A12 and IMC-11F8 after 4 h incubation at 37°C.
  • C The Di-diabody (100 nM) induced IGFR degradation in BxPC3 cells in a time-dependent manner when incubated at 37 0 C.
  • DAb the Di-diabody; A + F, IMC-A12 plus IMC-11F8; 1121, the control antibody, MC-1121.
  • FIG 12 shows di-diabody mediated ADCC in the presence of human PBMC.
  • Tumor cells were incubated in the wells of 96-well plates with IMC-11F8, IMC- A12, the Di-diabody, or a normal human IgG at 37°C for 30 min, followed by addition of PBMC at an effector/tumor cell ratios of 100/1 and incubation at 37°C for an additional 4 h. After centrifugation at 1500 rpm for 10 min, 100 ⁇ l of supernatant was transferred to 96 well flat bottom plates, followed by the addition of 100 ⁇ l/well LDH assay reagent and reading of the absorbance at 490nm. The percentages of specific cell lysis were calculated as described in the Materials and Methods. Data represent the means of triplicate determinations.
  • Figure 13 shows inhibition of growth of human tumor xenografts in nude mice by the di-diabody.
  • Female athymic nu/nu mice were injected subcutaneously on the lateral dorsal surface with BxPC3 (2 x 10 6 /mouse) or HT-29 (5 x 10 6 /mouse) tumor cells.
  • NYOl 924757 vl tumors reached approximately 200-300 mm 3 mice were randomized by tumor size and divided into treatment groups. The mice were treated by IMC-11F8 (or the equivalent IMC- C225), IMC-A12, IMC-11F8 (or IMC-C225) plus IMC-A12, the di-diabody, or the control articles (saline or normal human IgG). Each antibody was administered by intraperitoneal injections at 40 mg/kg twice a week. Tumor volume and body weight of each animal was measured twice a week. Data represent the mean ⁇ SE of tumor sizes from 10 to 12 animals in each group.
  • the invention provides an antigen-binding protein which is a tetravalent BsAb molecule, comprising two identical diabodies.
  • Each diabody is a heterodimer of two polypeptide chains and is preferably bispecific.
  • one of the polypeptides of the diabody is fused, via a hinge region, to the N-terminus of an immunoglobulin constant domain that is capable of stable self association (e.g., C H 2 and/or C R 3 if IgGl).
  • di-diabody Coexpression of the two polypeptide chains in a single host results in the formation of bispecific diabody-constant domain fusion, which in turn dimerizes via both the hinge and the constant domain to form a bispecific tetravalent protein (a "di-diabody").
  • the di-diabody can be efficiently expressed in E.coli and mammalian cells, and is more efficient in binding to its targets and in blocking target-associated biological activities than the corresponding diabody.
  • Antibody specificity refers to selective recognition of the antibody for a particular epitope of an antigen. Natural antibodies, for example, are monospecific. Bispecific antibodies (BsAbs) are antibodies which have two different antigen-binding specificities or sites. Where an antigen-binding protein has more than one specificity, the recognized epitopes may be associated with a single antigen or with more than one antigen.
  • a natural antibody molecule is composed of two identical heavy chains and two identical light chains. Each light chain is covalently linked to a heavy chain by an interchain disulfide bond. The two heavy chains are further linked to one another by multiple disulfide bonds at the hinge region. The individual chains fold into domains having similar sizes (about 110-125 amino acids) and structures, but different functions.
  • the light chain comprises one variable domain (V L ) and one constant domain (C L ).
  • V L variable domain
  • C L constant domain
  • NYOl 9247 5 7 vl comprises one variable domain (V H ) and, depending on the class or isotype of antibody, three or four constant domains (C H I, C H 2, C H 3 and C H 4).
  • V H variable domain
  • C H I constant domains
  • the isotypes are IgA, IgD, IgE, IgG, and IgM, with IgA and IgG further subdivided into subclasses or subtypes.
  • the portion of an antibody consisting of V L and V H domains is designated "Fv" and constitutes the antigen-binding site.
  • a single chain Fv is an engineered protein containing a VL domain and a V H domain on one polypeptide chain, wherein the N terminus of one domain and the C terminus of the other domain are joined by a flexible linker.
  • Fab refers to the portion of the antibody consisting of V L , V H , C L and C H I domains.
  • variable domains show considerable amino acid sequence variablity from one antibody to the next, particularly at the location of the antigen binding site.
  • Three regions, called “hypervariable” or “complementarity-determining regions” (CDRs) are found in each of V L and V H -
  • Fc is the designation for the portion of an antibody which comprises paired heavy chain constant domains, hi an IgG antibody, for example, the Fc comprises C R 2 and CH3 domains.
  • the Fc of an IgA or an IgM antibody further comprises a C H 4 domain.
  • the Fc is associated with Fc receptor binding, activation of complement-mediated cytotoxicity and antibody-dependent cellular-cytotoxicity.
  • complex formation requires Fc constant domains.
  • the "hinge” region separates the Fab and Fc portions of the antibody, providing for mobility of Fabs relative to each other and relative to Fc, as well as including multiple disulfide bonds for covalent linkage of the two heavy chains.
  • Diabodies contain two polypeptide chains which when associated, form two antibody binding sites.
  • Each polypeptide chain contains an immunoglobulin heavy chain variable (V H ) domain and an immunoglobulin light chain variable (V L ) domain, joined by short peptide linker (5-10 amino acids).
  • V H immunoglobulin heavy chain variable
  • V L immunoglobulin light chain variable
  • short peptide linker 5-10 amino acids
  • the order of the V H and V L domains of one polypeptide chain is reversed with respect to the other (i.e., V ⁇ -linker-V L and V L -linker-V ⁇ )
  • V ⁇ -linker-V L and V L -linker-V ⁇ formation of heterodimers of the two non-identical chains is promoted by the combination of short peptide linkers, which inhibit intrachain V H -VL association, and the order of the variable domains of the two polypeptides. Accordingly, in the heterodimer, VH of the first polypeptide is paired with V
  • V H domains are identical and the VL domains are identical
  • two heterodimer contains two identical binding sites.
  • two different V H domains and two different corresponding VL domains may be employed such that the heterodimer contains two different binding sites and is bispecific (i.e., VB 1 - linker-Vu and V L1 -linker- VH 2 ; see Fig. 1).
  • the bispecific di-diabody that results from linking two such bispecific diabodies, is thus bivalent for each of the two different binding sites.
  • Di-diabodies of the invention which are bispecific have a combination of desirable features.
  • they are essentially homogeneous.
  • mispairing of antibody heavy and light chains is greatly reduced or eliminated.
  • a typical bispecific antibody requires the use of two different heavy chains to provide two specificities.
  • Four combinations are possible when the heavy chains are arranged into an IgG type molecule. Two of those consist of mispaired heavy chains such that the product is monospecific.
  • mispairing is substantially eliminated. Pairing of two diabody polypeptides by heterodimerization is favored, and dimerization of the bispecific diabodies is by a different interaction. Accordingly, the products are bispecific.
  • a second advantage of the di-diabodies is that they are bivalent for each binding specificity.
  • a feature of a natural antibody which is missing from a dimeric BsAb is that the natural antibody is bivalent for the antibody binding site that it comprises.
  • a dimeric BsAb is monovalent for each of the two binding sites that it comprises. This is significant for antibody function because bivalency allows for cooperativity of binding and a significant increase in binding avidity with resepect to a molecule comprising a single antigen-binding site.
  • a third advantage of the di-diabodies is that heavy chain constant domains which constitute the Fc region (e.g., CH2 and/or C H 3 for an IgGl molecule) of a natural antibody and which provide other antibody functions can be present. Furthermore, the multiple binding domains are separated from the constant domains such that functions provided by the constant domains are not impaired. Constant domain functions include binding to certain accessory molecules (e.g., binding to cell surface and soluble Fc receptors, J chain association for IgA and IgM, S protein for IgA), activation of the complement
  • NYOl 924757 vl pathway (complement mediated cytoxicity, CMC), recognition of antibody bound to target cells by several different leukocyte populations (antibody-dependent cell-mediated cytoxicity, ADCC) and opsonization (enhancement of phagocytosis).
  • CMC complement mediated cytoxicity
  • ADCC antibody-dependent cell-mediated cytoxicity
  • opsonization enhancement of phagocytosis
  • the Fc heavy chain constant domain(s) can confer increased serum half-life.
  • a fourth advantage of proteins of the invention is that there is no requirement for processing in vitro to obtain the complete product. Though rearranged in an artificial manner, each of the domains has a natural character which allows expression in a biological system. For example, di-diabodies can be expressed in prokaryotic and eukaryotic expression systems. The proteins that are produced are substantially bispecific. Although multimeric forms of the bispecific di-diabody may be expressed to some degree (see Examples), these are easily removed from preparations by chromatography if necessary.
  • the present invention is also applicable to production of monospecific tetravalent antigen-binding proteins.
  • all four binding sites of the di-diabody have the same specificity.
  • An antigen binding site for use in a di-diabody can be obtained by a variety of methods.
  • the amino acid sequences of the V H and VL portions of a selected binding domain correspond to a naturally-occurring antibody or are chosen or modified to obtained desired immunogenic or binding characteristics.
  • V H and V L domains can be obtained directly from a monoclonal antibody which has the desired binding characteristics.
  • V H and V L domains can be from libraries of V gene sequences from a mammal of choice. Elements of such libraries express random combinations of V H and V L domains and are screened with any desired antigen to identify those elements which have desired binding characteristics. Particularly preferred is a human V gene library. Methods for such screening are known in the art.
  • V H and V L domains from a selected non-human source may be incorporated into chimeric di-diabodies.
  • a di-diabody with one or more functional human constant domains wherein the V H and V L domains have been selected from a non-human source.
  • human constant domains are preferred.
  • a di-diabody can be made that is "humanized.”
  • Humanized variable domains are constructed in which amino acid sequences which comprise one or more
  • NYOl 9247S7 vl complementarity determining regions (CDRs) of non-human origin are grafted to human framework regions (FRs).
  • CDRs complementarity determining regions
  • FRs human framework regions
  • a humanized construct is particularly valuable for elimination of adverse immunogenic characteristics, for example, where an antigen binding domain from a non-human source is desired to be used for treatment in a human.
  • Variable domains have a high degree of structural homology, allowing easy identification of amino acid residues within variable domains which corresponding to CDRs and FRs.
  • CDRs are most easily grafted onto different FRs by first amplifying individual FR sequences using overlapping primers which include desired CDR sequences, and joining the resulting gene segments in subsequent amplification reactions. Grafting of a CDR onto a different variable domain can further involve the substitution of amino acid residues which are adjacent to the CDR in the amino acid sequence or packed against the CDR in the folded variable domain structure which affect the conformation of the CDR.
  • Humanized variable domains of the invention therefore include human domains which comprise one or more non-human CDRs as well as such domains in which additional substitutions or replacements have been made to preserve or enhance binding characteristics.
  • Di-diabodies of the invention may also employ variable domains which have been made less immunogenic by replacing surface-exposed residues to make the di-diabody appear as self to the immune system (Padlan, E.A. (1991) MoI. Immunol. 28, 489-498). Antibodies have been modified by this process with no loss of affinity (Roguska et al. (1994) Proc. Natl. Acad. Sci. USA 91, 969-973). Because the internal packing of amino acid residues in the vicinity of the antigen binding site remains unchanged, affinity is preserved.
  • the invention contemplates binding domains which are essentially human.
  • Human binding domains are obtained from phage display libraries wherein combinations of human heavy and light chain variable domains are displayed on the surface of filamentous phage (See, e.g., McCafferty et al. (1990) Nature 348, 552-554; Aujame et al. (1997) Human Antibodies 8, 155-168).
  • Combinations of variable domains are typically displayed on filamentous phage in the form of Fabs or scFvs.
  • the library is screened for phage bearing combinations of variable domains having desired antigen binding characteristics.
  • Preferred variable domain combinations display high affinity for a selected antigen and little cross- reactivity to other related antigens.
  • human binding domains can be obtained from transgenic animals into which unrearranged human Ig gene segments have been introduced and in which the endogenous mouse Ig genes have been inactivated (reviewed in Bruggemann and Taussig (1997) Curr. Opin. Biotechnol. 8, 455-458).
  • Preferred transgenic animals contain very large contiguous Ig gene fragments that are over 1 Mb in size (Mendez et al. (1997) Nature Genet. 15, 146-156) but human Mabs of moderate affinity can be raised from transgenic animals containing smaller gene loci (See, e.g., Wagner et al. (1994) Eur. J. Immunol. 42, 2672-2681; Green et al. (1994) Nature Genet. 7, 13-21).
  • binding domains of the invention include those for which binding characteristics have been improved by direct mutation or by methods of affinity maturation. Affinity and specificity may be modified or improved by mutating CDRs and screening for antigen binding sites having the desired characteristics (See, e.g., Yang et al. (1995) J. MoI.
  • CDRs are mutated in a variety of ways. One way is to randomize individual residues or combinations of residues so that in a population of otherwise identical antigen binding sites, all twenty amino acids, or a subset thereof, are found at particular positions. Alternatively, mutations are induced over a range of CDR residues by error prone PCR methods (See, e.g., Hawkins et al. (1992) J. MoI. Bio. 226, 889-896). Phage display vectors containing heavy and light chain variable region genes are propagated in mutator strains of E. coli (See, e.g., Low et al. (1996) J. MoI. Bio. 250, 359-368). These methods of mutagenesis are illustrative of the many methods known to one of skill in the art.
  • Each variable domain of the antibodies of the present invention may be a complete immunoglobulin heavy or light chain variable domain, or it may be a functional equivalent or a mutant or derivative of a naturally occurring domain, or a synthetic domain constructed, for example, in vitro using a technique such as one described in WO 93/11236 (Medical Research Council / Griffiths et al.). For instance, it is possible to incorporate domains corresponding to antibody variable domains which are missing one or more amino acids.
  • the important characterizing feature is the ability of each variable domain to associate with a complementary variable domain to form an antigen binding site.
  • Antigen-binding proteins of the invention have binding sites for any epitope, antigenic site or protein.
  • di-diabodies that are useful for treatment of disease.
  • Preferred di-diabodies neutralize receptor proteins, such as receptors which are involved in angiogenesis and/or oncogenesis.
  • Neutralizing a receptor means inactivating the intrinsic kinase activity of the receptor to transduce a signal.
  • a reliable assay for receptor neutralization is the inhibition of receptor phosphorylation.
  • the present invention is not limited by any particular mechanism of receptor neutralization. Some possible mechanisms include preventing binding of the ligand to the extracellular binding domain of the receptor, and preventing dimerization or oligomerization of receptor. Other mechanisms cannot, however, be ruled out.
  • Neutralization of activation of a receptor in a sample of endothelial or non- endothelial cells, such as tumor cells may be performed in vitro or in vivo.
  • Neutralizing activation of a receptor in a sample of receptor expressing cells comprises contacting the cells with an antibody of the invention.
  • the cells are contacted with the antibody before, simultaneously with, or after, adding VEGF to the cell sample.
  • NYOl 9247 5 7 vl invention is contacted with a receptor by administration to a mammal.
  • Methods of administration to a mammal include, for example, oral, intravenous, intraperitoneal, subcutaneous, or intramuscular administration.
  • VEGF receptors include, but are not limited to VEGF receptors (e.g., VEGFR-2/KDR/Flk-l, VEGFR2/F1M, VEGFR3/Flt-4), epidermal growth factor receptor (EGFR), insulin-like growth factor receptor (IGFR) and the like.
  • VEGF receptors e.g., VEGFR-2/KDR/Flk-l, VEGFR2/F1M, VEGFR3/Flt-4
  • EGFR epidermal growth factor receptor
  • IGFR insulin-like growth factor receptor
  • Additional non- limiting examples of receptor tyrosine kinases include Flt-4, HER2/neu, Tek and Tie2.
  • FGF fibroblast growth factor
  • PDGF platelet derived growth factor
  • NGF nerve growth factor
  • FGF-R fibroblast growth factor
  • PDGF-R platelet derived growth factor receptor
  • NGFR nerve growth factor receptor
  • MSP-R macrophage-stimulating protein receptor
  • Receptors of interest include human proteins and homologues from other mammals.
  • Di-diabodies can incorporate immunoglobulin binding domains from any source.
  • antibodies are known for the above listed receptors and are sources of V H and V L domains for use in di-diabodies of the present invention.
  • binding domains specific for KDR include IMC-ICl 1 (nucleotide and amino acids sequences of V H : SEQ ID NOS:1 and 2; nucleotide and amino acid sequences of V L : SEQ ID NOS:3 and 4) (see, WO 00/44777), EVIC-2C6 (nucleotide and amino acids sequences of V H : SEQ ID NOS: 5 and 6; nucleotide and amino acid sequences of V L : SEQ ID NOS:7 and 8) (see, WO 03/075840), and IMC-1121 (nucleotide and amino acids sequences of V H : SEQ ID NOS: 5 and 6; nucleotide and amino acid sequences of V L :
  • binding domains specific for FIt-I include 6.12 (nucleotide and amino acids sequences of V H : SEQ ID NOS: 11 and 12; nucleotide and amino acid sequences of V L : SEQ ID NOS:13 and 14) and BVIC-18F1 (nucleotide and amino acids sequences of V H : SEQ ID NOS :27 and 28; nucleotide and amino acid sequences of V L : SEQ ID NOS :29 and 30).
  • Binding domains specific for EGFR include, for example, ERBITUX ® (Cetuximab; IMC-C225) (nucleotide and amino acids sequences of V H : SEQ ID NOS: 15 and 16; nucleotide and amino acid sequences of V L : SEQ ID NOS: 17 and 18) as disclosed in WO
  • NYOl 924757 vl 96/40210 and IMCl 1F8 (nucleotide and amino acids sequences of V H : SEQ ID NOS: 19 and 20; nucleotide and amino acid sequences of V L : SEQ ID NOS:21 and 22).
  • An example of a binding domain specific for IGFR is IMC-A12 (nucleotide and amino acids sequences of V H : SEQ ID NOS: 23 and 24; nucleotide and amino acid sequences of V L : SEQ ID NOS: 25 and 26).
  • Antibodies that bind to FGF receptors include, for example, FR1-H7, FRl-Al, and FRl -4H (WO 2005/037235).
  • Antibodies that bind to PDGFR ⁇ include, for example, 3G3 and 7Gl 1 (Loizos et al., 2005, MoI. Cancer Ther. 4:369).
  • MSP-R include IMC-41 AlO and IMC-41 A12 (Pereira, International application , filed 13- May-2005).
  • binding domains such as the CDR regions
  • portions of the above listed binding domains may be incorporated into binding domains used to make the binding proteins described herein.
  • Certain preferred diabodies bind to two of the above listed receptors.
  • such a bispecific antigen-binding protein binds to KDR and FLT-I.
  • An example of such an antigen binding protein has two polypeptide chains as provided in the examples (nucleotide and amino acid sequences: first polypeptide - SEQ ID NOS:51 and 52; second polypeptide - SEQ ID NOS :53 and 54).
  • the linkers between the heavy and light chain variable domains are Gly-Gly-Gly-Gly-Ser, but a linker of 0 to 10 amino acids having any amino acid sequence can be used.
  • the antigen-binding protein incorporates a C H 3 domain but not a C H 2 domain.
  • a linker (Gly-Gly-Gly-Gly-Ser) 2 is incorporated between the hinge region and the IgGl C JJ 3 domain, but a linker of 0 to 30 amino acids having any amino acid sequence can be used.
  • an antigen-binding protein of the invention binds to HER2 and EGFR.
  • a di-diabody of the invention binds to EGFR and IGFR.
  • An example of an antigen binding protein that binds to EGFR and IGFR is provided in the examples (nucleotide and amino acid sequences: first polypeptide - SEQ ID NOS:55 and 56; second polypeptide - SEQ ID NOS:57 and 58).
  • the linkers between the heavy and light chain variable domains are Arg-Thr-Val- AIa- Ala.
  • the antigen-binding protein incorporates C H 2 and CH3 constant domains, and no linker was used between the hinge region and the constant domains.
  • an antigen-binding protein of the invention binds to EGFR and a VEGFR.
  • the VEGFR is VEGFR2.
  • Such an antibody is useful for blocking stimulation of vascular epithelial cells, by blocking signal transduction through both EGFR and VEGFR. This is particularly useful where angiogenesis occurs in response to EGFR ligands, particularly TGR ⁇ , secreted by tumor cells.
  • Di-diabodies of the invention can be used to cross-link antigens on target cells with antigens on immune system effector cells. This can be useful, for example, for promoting immune responses directed against cells which have a particular antigens of interest on the cell surface.
  • immune system effector cells include antigen specific cells such as T cells which activate cellular immune responses and nonspecific cells such as macrophages, neutrophils and natural killer (NK) cells which mediate cellular immune responses.
  • Di-diabodies of the invention can have a binding site for any cell surface antigen of an immune system effector cell.
  • cell surface antigens include, for example, cytokine and lymphokine receptors, Fc receptors, CD3, CD 16, CD28, CD32 and CD64.
  • antigen binding sites are provided by scFvs which are derived from antibodies to the aforementioned antigens and which are well known in the art.
  • Antigen-binding sites of the invention which are specific for cytokine and lymphokine receptors can also be sequences of amino acids which correspond to all or part of the natural ligand for the receptor.
  • an antigen-binding protein of the invention can have an antigen-binding site which comprises a sequence of amino acids corresponding or IL-2.
  • Other cytokines and lymphokines include, for example, interleukins such as interleukin-4 (IL-4) and interleukin-5 (IL-5), and colony-stimulating factors (CSFs) such as granulocyte-macrophage CSF (GM-CSF), and granulocyte CSF (G-CSF).
  • Di-diabodies of the invention are made by expressing two diabody chains, one of which is linked to at least one heavy chain constant domain that is capable of dimerization (e.g., CH2 and/or CH3).
  • Di-diabodies are conveniently produced in E. coli using DNA constructs which comprise bacterial secretion signal sequences at the start of each polypeptide chain.
  • a variety of bacterial signal sequences are known in the art.
  • a perferred signal sequence is from the pelB gene of Erwinia carotovora.
  • the DNA fragments coding for the diabodies can be cloned, e.g., into vectors employing human cytomegalovirus
  • NYOl 924757 vl (HCMV) promoters and enhancers for high level expression in mammalian cells such as, for example, CHO, NSO, COS-7, and PER.C6 cells, and cell lines of lymphoid origin such as lymphoma, myeloma, or hybridoma cells.
  • mammalian cells such as, for example, CHO, NSO, COS-7, and PER.C6 cells
  • cell lines of lymphoid origin such as lymphoma, myeloma, or hybridoma cells.
  • a selectable marker is a gene which encodes a protein necessary for the survival or growth of transformed host cells grown in a selective culture medium.
  • Typical selectable markers encode proteins that (a) confer resistance to antibiotics or other toxins, e.g. ampicillin, neomycin, methotrexate, or tetracycline, (b) complement auxotrophic deficiencies, or (c) supply critical nutrients not available from complex media, e.g. the gene encoding D-alanine racemase for Bacilli.
  • a particularly useful selectable marker confers resistance to methotrexate.
  • cells transformed with the DHFR selection gene are first identified by culturing all of the transformants in a culture medium that contains methotrexate (Mtx), a competitive antagonist of DHFR.
  • Mtx methotrexate
  • An appropriate host cell when wild-type DHFR is employed is the Chinese hamster ovary (CHO) cell line deficient in DHFR activity, prepared and propagated as described by Urlaub and Chasin (1980) Proc. Natl. Acad. ScL USA 11, 4216.
  • the transformed cells are then exposed to increased levels of methotrexate. This leads to the synthesis of multiple copies of the DHFR gene, and, concomitantly, multiple copies of other DNA comprising the expression vectors, such as the DNA encoding the antibody or antibody fragment.
  • an example of a suitable selection gene for use in yeast is the trpl gene present in the yeast plasmid YRp7. Stinchcomb et al. (1979) Nature, 282, 39; Kingsman et al. (1979) Gene 1, 141.
  • the trpl gene provides a selection marker for a mutant strain of yeast lacking the ability to grow in tryptophan, for example, ATCC No. 44076 or PEP4-1. Jones (1977) Genetics 85, 12.
  • the presence of the trpl lesion in the yeast host cell genome then provides an effective environment for detecting transformation by growth in the absence of tryptophan.
  • Leu2-deficient yeast strains (ATCC 20,622 or 38,626) are complemented by known plasmids bearing the Leu2 gene.
  • Transformed host cells are cultured by methods known in the art in a liquid medium containing assimilable sources of carbon, e.g. carbohydrates such as glucose or lactose, nitrogen, e.g. amino acids, peptides, proteins or their degradation products such as
  • the medium furthermore contains, for example, growth-promoting substances, such as trace elements, for example iron, zinc, manganese and the like.
  • Di-diabodies that bind to growth factor receptors are preferably capable of blocking activation of receptor tyrosine kinase (RTK) activity.
  • Tyrosine kinase inhibition can be determined using well-known methods, for example, by measuring the autophosphorylation level of recombinant kinase receptor, and/or phosphorylation of natural or synthetic substrates.
  • phosphorylation assays are useful in determining RTK antagonists of the present invention. Phosphorylation can be detected, for example, using an antibody specific for phosphotyrosine in an ELISA assay or on a western blot.
  • methods for detection of protein expression can be utilized to determine RTK antagonists, wherein expression of the proteins being measured is mediated by the RTK.
  • RTK immunohistochemistry
  • FISH fluorescence in situ hybridization
  • competitive radioligand binding assays solid matrix blotting techniques, such as Northern and Southern blots, reverse transcriptase polymerase chain reaction (RT-PCR) and ELISA.
  • RT-PCR reverse transcriptase polymerase chain reaction
  • ELISA e.g., Grandis et al., Cancer, (1996) 78:1284-92; Shimizu et al., Japan J. Cancer Res., (1994) 85:567-71; Sauter et al. Am. J.
  • the ability of a di-diabody to block ligand binding can be measured, for example, by an in vitro competitive assay.
  • a ligand or the RTK e.g., EGF for EGFR
  • a binding assay is carried out to determine the effectiveness of the di-diabody to competitively inhibit binding of the RTK to the immobilized ligand.
  • In vivo assays can also be utilized to determine RTK antagonists.
  • receptor tyrosine kinase inhibition can be observed by mitogenic assays using cell lines stimulated with receptor ligand in the presence and absence of inhibitor.
  • NYOl 924757 vl A431 cells (American Type Culture Collection (ATCC), Rockville, MD) stimulated with EGF can be used to assay EGFR inhibition.
  • Another method involves testing for inhibition of growth of EGFR-expressing tumor cells, using for example, human tumor cells injected into a mouse. See U.S. Patent No. 6,365,157 (Rockwell et al.).
  • Preferred di-diabodies of the instant invention have dual specificity and are capable of binding to two different antigens simultaneously.
  • the different antigens can be located on different cells or on the same cell.
  • Cross linking of antigen can be shown in vitro, for example by providing a solid surface to which a first antigen has been bound, adding a bispecific antibodies specific for the first antigen and a second antigen for which the binding protein is also specific and detecting the presence of bound second antigen.
  • Preferred bispecific di-diabodies of the invention are capable of blocking the interaction between two receptors and their respective ligands.
  • a di-diabodies specific for KDR and FIt-I inhibits VEGF induced cell migration as well as PlGF induced cell migration.
  • Combination of two receptor binding specificities in a bispecific di-diabodies can be more efficacious in inhibiting cell migration than the individual parent antibodies (see, e.g., WO 2004/003211; Zhu).
  • bispecific di-diabodies can be more potent inhibitors of cellular function.
  • VEGF-stimulated cellular functions such as, for example, proliferation of endothelial cells and VEGF- and PlGF-induced migration of human leukemia cells can be more efficiently inhibited by bispecific di- diabodies, even where affinity for one or both of the two target antigens is reduced.
  • a diabody specific (monovalent) for both KDR and FIt-I is more effective to inhibit VEGF or PlGF induced cell migration than a monospecific scFv directed at either of the target antigens (WO 2004/003211).
  • a di-diabody having dual specificity for both EGFR and IGFR that is capable of binding to both receptors and blocking interaction with their specific ligands is use to neutralizing both EFG and IGF-stimulated receptor activation and downstream signal transduction.
  • Stimulation of either EGFR or IGFR results in phosphorylation of common downstream signal transduction molecules, including Akt and p44/42, although to different extents, hi certain tumor cells, inhibition of EGFR function can be compensated by upregulation of other growth factor receptor signaling pathways, and
  • NYOl 924757 vl particularly by IGFR stimulation In contrast to treatment with an antibody binds to one receptor, and does not completely block phosphorylation of either Akt or p44/42, incubation of tumor cells with an antibody that binds to both EGFR and IGFR blocks phosphorylation of both Akt and p44/42. Accordingly, inhibition of IGFR signaling results in inhibition of tumor growth and increased sensitivity of tumor cells to certain therapeutic agents.
  • the antigen-binding proteins are generally useful for treating neoplastic diseases characterized by cell growth or transformation resulting from activation of multiple signal transduction pathways.
  • the antigen-binding proteins of the invention are useful for treatment of a variety of proliferative disorders.
  • the present invention provides for treatment of tumors that express and are stimulated through more than one receptor tyrosine kinase. Stimulation through more that one receptor can result in uncontrolled growth that is insensitive to blockage of each receptor alone.
  • stimulation of a second receptor can add to the activation observed in response to stimulation through a first receptor.
  • the contributions from the individual receptors can be multiplicative. In each of the above instances, significantly improved inhibition of tumor growth is observed in the presence of an antigen-binding protein that blocks both of the receptors.
  • the antigen-binding proteins of the invention are useful for treating diseases in which receptor stimulation is through an EGFR paracrine and/or autocrine loop.
  • EGFR expressing tumors are characteristically sensitive to EGF present in their environment, and can further be stimulated by tumor produced EGF or TGF- ⁇ .
  • the diseases and conditions that may be treated or prevented by the present methods include, for example, those in which tumor growth is stimulated. The method is therefore effective for treating a solid tumor that is not vascularized, or is not yet substantially vascularized.
  • antigen-binding proteins of the invention are useful for inhibiting angiogenesis associated with a hyperproliferative disease. For example, by blocking tumor associated angiogenesis, tumor growth may be inhibited.
  • the antigen- binding protein binds to a tumor associated RTK and inhibits production of angiogenic
  • NYOl 924757 vl ligands i.e., VEGF
  • VEGF vl ligands
  • the antigen- binding protein binds to multiple VEGF receptors, such that VEGF or other ligand of VEGFR (e.g., PlGF) ligand is blocked from binding to more than one type of VEGF receptor.
  • VEGFR e.g., PlGF
  • Tumors that may be treated include primary tumors and metastatic tumors, as well as refractory tumors.
  • Refractory tumors include tumors that fail to respond or are resistant to treatment with chemotherapeutic agents alone, antibodies alone, radiation alone or combinations thereof.
  • Refractory tumors also encompass tumors that appear to be inhibited by treatment with such agents, but recur up to five years, sometimes up to ten years or longer after treatment is discontinued.
  • the tumors may express EGFR or other RTK at normal levels or they may overexpress the RTK at levels, for example, that are at least 10, 100, or 1000 times normal levels.
  • tumors that express EGFR and are stimulated by a ligand of EGFR include carcinomas, gliomas, sarcomas, adenocarcinomas, adenosarcomas, and adenomas.
  • Such tumors can occur in virtually all parts of the body, including, for example, breast, heart, lung, small intestine, colon, spleen, kidney, bladder, head and neck, ovary, prostate, brain, pancreas, skin, bone, bone marrow, blood, thymus, uterus, testicles, cervix or liver.
  • tumors observed to overexpress EGFR include, but are not limited to, colorectal and head and neck tumors, especially squamous cell carcinoma of the head and neck, brain tumors such as glioblastomas, and tumors of the lung, breast, pancreas, esophagus, bladder, kidney, ovary, cervix, and prostate.
  • tumors observed to have constitutively active (i.e., unregulated) receptor tyrosine kinase activity include gliomas, non-small-cell lung carcinomas, ovarian carcinomas and prostate carcinomas.
  • tumors include Kaposi's sarcoma, CNS neoplasms, neuroblastomas, capillary hemangioblastomas, meningiomas and cerebral metastases, melanoma, gastrointestinal and renal carcinomas and sarcomas, rhabdomyosarcoma, glioblastoma, preferably glioblastoma multiforme, and leiomyosarcoma.
  • RTKs Overexpression of other RTKs can produce similar growth defects. For example, most metastatic bone cancers arise from primary tumors of prostate, breast, or lung. Prostate tumors initially may be hormone dependent, but loss of such dependence coincides with IGFR mediated stimulation of cells that migrate to bone.
  • hyperproliferative disease is defined as a condition caused by excessive growth of non-cancer cells that express a member of the EGFR family or other tyrosine kinase receptors. The excess cells generated by a hyperproliferative disease express the RTK at normal levels or they may overexpress the RTK.
  • hyperproliferative disease examples include psoriasis, actinic keratoses, and seborrheic keratoses, warts, keloid scars, and eczema. Also included are hyperproliferative diseases caused by virus infections, such as papilloma virus infection. For example, psoriasis comes in many different variations and degrees of severity.
  • psoriasis display characteristics such as pus-like blisters (pustular psoriasis), severe sloughing of the skin (erythrodeimic psoriasis), drop-like dots (guttae psoriasis) and smooth inflamed lesions (inverse psoriasis).
  • the treatment of all types of psoriasis e. g., psoriasis vulgaris, psoriasis pustulosa, psoriasis erythrodermica, psoriasis arthropathica, parapsoriasis, palmoplanar pustulosis
  • psoriasis vulgaris e.g., psoriasis vulgaris, psoriasis pustulosa, psoriasis erythrodermica, psoriasis arthropathica, parapsoriasis, palmoplanar
  • di-diabodies can be chemically or biosynthetically conjugated to other agents such as antineoplastic or anti-angiogenic agents for treatment of disease.
  • Anti-tumor agents linked to an antibody include any agents which destroy or damage a tumor to which the antibody has bound or in the environment of the cell to which the antibody has bound.
  • an anti-tumor agent is a toxic agent such as a chemotherapeutic agent or a radioisotope.
  • the chemotherapeutic agents are conjugated to the di-diabody using conventional methods ⁇ See, e.g., Hermentin and Seiler (1988) Behring Inst. Mitt. 82, 197-215), including by peptide and non-peptide linkers.
  • Di-diabodies can also be linked to detectable signal-producing agents useful in vivo and in vitro for diagnostic purposes.
  • the signal producing agent produces a measurable signal which is detectible by external means, usually the measurement of electromagnetic radiation.
  • the signal producing agent is an enzyme or chromophore, or emits light by fluorescence, phosphorescence or chemiluminescence.
  • Chromophores include dyes which absorb light in the ultraviolet or visible region, and can be substrates or degradation products of enzyme catalyzed reactions.
  • the invention further contemplates the use of di-diabodies with treatment or diagnostic agents incorporated into secondary reagents.
  • one member of a binding pair is linked to the di-diabody of the invention.
  • Anti-neoplastic agents are conjugated to second members of such pairs and are thereby directed to the site where the di-diabody is bound, hi a preferred embodiment, biotin is conjugated to a di-diabody, and thereby provides a target for an anti-neoplastic agent or other moiety which is conjugated to avidin or streptavidin.
  • biotin or another such moiety is linked to a di-diabody of the invention and used as a reporter, for example in a diagnostic system where a detectable signal-producing agent is conjugated to avidin or streptavidin.
  • Di-diabodies can be administered in combination with one or more suitable adjuvants, such as, for example, cytokines (IL-10 and IL- 13, for example) or other immune stimulators, such as, but not limited to, chemokine, tumor-associated antigens, and peptides. It should be appreciated, however, that administration of only a diabody is sufficient to prevent, inhibit, or reduce the progression of the tumor in a therapeutically effective manner.
  • suitable adjuvants such as, for example, cytokines (IL-10 and IL- 13, for example) or other immune stimulators, such as, but not limited to, chemokine, tumor-associated antigens, and peptides.
  • an antigen-binding protein of the invention that binds to an RTK and blocks ligand binding in combination with another antigen-binding protein that binds to ligand.
  • Ligand binding antibodies are well known in the art, and include, e.g., anti-VEGF (Avastin ® ; bevacizumab).
  • the di-diabodies of the invention are also to be used in combined treatment methods by administration with an anti-neoplastic agent such as a chemotherapeutic agent or a radioisotope.
  • chemotherapeutic agents include irinotecan (CPT-Il), anthracyclines (e.g. daunomycin and doxorubicin), methotrexate, vindesine, neocarzinostatin, cisplatin, chlorambucil, cytosine arabinoside, 5-fluorouridine, melphalan, ricin and calicheamicin.
  • a di-diabody and an anti-angiogenic or anti-neoplastic agent are admininstered to a patient in amounts effective to inhibit angiogenesis and/or reduce tumor growth.
  • the di-diabodies are also to be administered in combination with other treatment regimes, for examplej with treatments such as radiation therapy.
  • combination therapies see, e.g., U.S. Patent No. 6,217,866 (Schlessinger et al.) (Anti-EGFR antibodies in combination with anti-neoplastic agents); WO 99/60023 (Waksal et al.) (Anti-EGFR antibodies in combination with radiation).
  • any suitable anti-neoplastic agent can be used, such as a chemotherapeutic agent, radiation or combinations thereof.
  • the anti-neoplastic agents known in the art or being evaluated can be grouped in to classes based on their target or mode of action.
  • alkylating agents include, but are not limited to, cisplatin, cyclophosphamide, melphalan, and dacarbazine.
  • anti-metabolites include, but not limited to, doxorubicin, daunorubicin, and paclitaxel, gemcitabine, and topoisomerase inhibitors irinotecan (CPT-Il), aminocamptothecin, camptothecin, DX-8951f, and topotecan (topoisomerase I) and etoposide (VP-16) and teniposide (VM-26) (topoisomerase II).
  • the source can be either external (external beam radiation therapy — EBRT) or internal (brachytherapy - BT) to the patient being treated.
  • EBRT internal beam radiation therapy
  • brachytherapy - BT teniposide
  • Such classifications can be useful for choosing an antineoplastic agent to use. For example, it has been observed that antibodies that bind IGFR may be particularly effective when administered with a topoisomerase inhibitor.
  • the dose of anti-neoplastic agent administered depends on numerous factors, including, for example, the type of agent, the type and severity tumor being treated and the route of administration of the agent. It should be emphasized, however, that the present invention is not limited to any particular dose.
  • the di-diabody is administered before, during, or after commencing therapy with another agent, as well as any combination thereof, i.e., before and during, before and after, during and after, or before, during and after commencing the anti-neoplastic agent therapy.
  • the diabody can be administered between 1 and 30 days, preferably 3 and 20 days, more preferably between 5 and 12 days before commencing radiation therapy.
  • chemotherapy is administered concurrently with, prior to, or subsequent to antibody therapy.
  • any suitable method or route can be used to administer di-diabodies of the invention, and optionally, to co-administer anti-neoplastic agents, receptor antagonists, or other pharmaceutical composition.
  • anti ⁇ neoplastic agent regimens utilized according to the invention include any regimen believed to be optimally suitable for the treatment of a patient's neoplastic condition. Different malignancies can require use of specific anti-tumor diabodies and specific anti-neoplastic agents, which will be determined on a patient to patient basis. Routes of administration
  • NYOl 9247 5 7 vl include, for example, oral, intravenous, intraperitoneal, subcutaneous, or intramuscular administration.
  • the dose of anti-neoplastic agent administered depends on numerous factors, including, for example, the type of neoplastic agent, the type and severity tumor being treated and the route of administration of the antineoplastic agent. It should be emphasized, however, that the present invention is not limited to any particular method or route of administration.
  • di-diabodies of the invention where used in a mammal for the purpose of prophylaxis or treatment, will be administered in the form of a composition additionally comprising a pharmaceutically acceptable carrier.
  • suitable pharmaceutically acceptable carriers include, for example, one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof.
  • Pharmaceutically acceptable carriers can further comprise minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the binding proteins.
  • the compositions of the injection can, as is well known in the art, be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the mammal.
  • kits for inhibiting tumor growth and/or angiogenesis, or treating other disesase comprising a therapeutically effective amount of a human di-diabody.
  • the kits can further contain any suitable antagonist of, for example, another growth factor receptor involved in tumorigenesis or angiogenesis (e.g., EGFR, VEGFR-l/Flt-1, VEGFR-2/Flk-l/KDR, IGFR, PDGFR, NGFR, FGFR, etc, as described above).
  • the kits of the present invention can further comprise an anti-neoplastic agent. Examples of suitable anti-neoplastic agents in the context of the present invention have been described herein.
  • the kits of the present invention can further comprise an adjuvant; examples have also been described above.
  • kits which contain di-diabodies of the present invention.
  • the present receptor binding di-diabodies thus can be used in vivo and in vitro for investigative, diagnostic, prophylactic, or treatment methods, which are
  • soluble receptor fusion proteins the extracellular domain of kinase inserting domain-containing receptor (KDR)-alkaline phosphatase (AP), and the extracellular domain of fins-like tyrosine kinase receptor (FIt-I)-AP, were expressed in stably transfected NIH 3T3 cells and purified from cell culture supernatant by affinity chromatography using immobilized monoclonal antibody to AP as described (Lu et al. (2000) J. Biol. Chem. 275:14321-30).
  • VEGF165 Vascular endothelial growth factor
  • the single chain antibody directed against KDR, scFv pi Cl 1 was isolated from a phage display library constructed from the splenocytes of a mouse immunized with KDR-AP fusion protein (Zhu et al. (1998) Cancer Res. 58:3209-14).
  • Hybridoma cell line producing the anti-Flt-1 antibody, FBK612 (IgGl, K) was established at ImClone Systems Incorporated (New York, NY) from mice immunized with FIt-I -AP fusion protein.
  • the single chain version of FBK612, scFv The single chain version of FBK612, scFv
  • NYOl 924757 vl 612 was constructed from the antibody variable genes isolated from the hybridoma cells as previously reported (Lu et al. (2001)).
  • the diabodies were secreted from E. coli strain HB2151 containing the expression plasmid grown at 30°C in a shaker flask following the procedure previously described (Lu et al., 2000; Zhu et al., 1998).
  • a periplasmic extract of the cells was prepared by resuspending the cell pellet in 25 mM Tris (pH 7.5) containing 20% (w/v) sucrose, 200 mM NaCl, 1 mM EDTA and 0.1 mM PMSF 5 followed by incubation at 4°C with gentle shaking for 1 h.
  • the diabodies were purified from the supernatant by an anti-E tag affinity chromatography using the RPAS Purification Module (Amersham Pharmacia Biotech). To examine the purity of the diabody preparations, the purified proteins were electrophoresed in a 4-20% gradient polyacrylamide gel (Novex, San Diego, CA) and visualized by staining with Colloidal Blue Stain kit (No vex).
  • the di-diabody was also expressed in mammalian COS cells. Nucleic acids encoding both of the polypeptides, each fused with an identical immunoglobulin leader
  • NYOl 924757 vl peptide sequence on its N-terminus were cloned into a single expression vector (the glutamine synthetase expression system from Lonza Biologies Inc.).
  • Sub- confluent COS cells were transfected with the expression vector using Lipofectamine following the instructions of the manufacturer (Invitrogen). The cells were cultured in serum-free medium, and supernatant was collected at 48 to 72 h after the transfection. After centrifugation to remove the cell debris, the di-diabody was purified from the supernatant using the RPAS Purification Module as described above.
  • the yield of the purified di-diabody ranged from 150 to 250 ⁇ g per liter of overnight bacteria culture in shaker flask, and approximately 800 to 1250 ⁇ g per liter of supernatant 48 to 72 h after transfection of the mammalian culture.
  • the composition and purity of the purified di-diabody preparations, both from E. coli expression [di-diabody (B)] and from mammalian expression [di-diabody (M)] was analyzed using SDS-PAGE (Fig. 2).
  • the components of the "cross-over" scFv in a bispecific diabody were also resolved by electrophoresis.
  • the divalent bispecific diabody, DAB pi Cl 1/612 yielded two bands (Fig. 2 A, lane 3), representing each one of the two "cross ⁇ over" scFv polypeptide chains (theoretical m.w., 26693.8 and 25179.6 daltons for the upper and the lower band, respectively).
  • the di-diabody preparation also gave two major bands (Fig.
  • the lower band representing the "cross-over” scFv chain (the same band as in DAB plCl 1/612), whereas the upper band represents the "cross-over” scFv-hinge-C H 3 fusion in dimer form (theoretical m.w., 79175.3 daltons).
  • the di-diabody yielded two major bands: the lower band represents the original "cross ⁇ over” scFv chain, and the top band corresponds to the reduced scFv-hinge-CH3 fusion in monomer form.
  • DAB 612 yielded one major band under both reducing and non-reducing conditions (lane 2, theoretical m.w. 26916.9 daltons).
  • DAB plCl 1 gave two bands (lane 1): the upper band corresponds to the single polypeptide, VL-G4S-VH, with intact E-tag (theoretical m.w. 26542.4 daltons); the lower band represents the same polypeptide with E-tag degradation.
  • the di-diabody yielded three distinct peaks on SEC.
  • ASTRA-computed Zimm analysis indicated a monodispersed peak (75% of the population) with a M w value of 132 kD, a monodispersed peak (20% of the population) with a M w value of 270 kD and a polydispersed peak (5% of the population) with a M w value of 470 kD.
  • the 132 kD peak represents the correctly folded monomelic di-diabody
  • the 270 kD peak represents the dimeric form of the di-diabody
  • the polydispersed 470 kD peak is a heterogeneous population with respect to oligomeric state.
  • an intact IgG antibody with a theoretical m.w. of 150 kD yielded a single peak with a estimated M w value of 150 kD
  • a diabody with a theoretical m.w. of 60 kD yielded a single peak with a estimated M w value of 62 kD, under identical conditions.
  • Two ELISA assays were carried out to determine the dual antigen binding capability of the bispecific di-diabody.
  • a cross-linking assay was used to investigate whether the di-diabody is capable of binding both of its target antigens simultaneously.
  • the monospecific diabodies, the bispecific diabody and di-diabody were first incubated in the presence of 1 mM of dithiothreitol (DTT) in a 96-well Maxi-sorp microtiter plate (Nunc, Roskilde, Denmark) precoated with FIt-I-Fc fusion protein (1 ⁇ g/ml x 100 ⁇ l per well overnight at 4°C) at RT for 1 h.
  • DTT dithiothreitol
  • the plate was washed three times with PBS containing 0.1 % Tween (PBST), followed by incubation with KDR-AP fusion protein (100 ng/well) at RT for additional 1 h.
  • KDR-AP fusion protein 100 ng/well
  • the plate-bound KDR-AP was then quantified by the addition of AP
  • the di-diabody was reduced into two half molecules, each consisting of a single diabody-C ⁇ 3 fusion (Fig. 1). Both the reduced divalent bispecific diabody and di-diabody, but not the monospecific diabody (DAB pi Cl 1 and DAB 612), were able to cross-link the two target antigens (Fig. 3, top panel). This observation indicates that the di-diabody, a dimer of two diabody-C ⁇ 3 fusions, possesses four antigen-binding sites (two on each C H 3 arm).
  • the direct binding assay various amounts of the diabodies were added to KDR or FIt-I -coated 96-well plates and incubated at RT for 1 h, after which the plates were washed 3 times with PBS. The plates were then incubated at RT for 1 h with 100 ⁇ l of an anti-E tag antibody-HRP conjugate (Amersham Pharmacia Biotech). The plates were washed, peroxidase substrate added, and the absorbance at 450nm read following the procedure described previously (Lu et al., 1999; 2000).
  • coli-de ⁇ ved di- diabody (B) demonstrated kinetic constants that are very similar to those of the scFv 612 and DAB pi Cl 1/612, whereas the mammalian cell-derived material showed a much slower association rate ikon), along with a significantly improved dissociation rate (koff) that is approaching that of the bivalent DAB 612.
  • *A11 numbers are determined by BIAcore analysis and represent the mean ⁇ SE of at least four separate determinations. Kd values are calculated as the ratios of & off / Ar 0n . **NDB, no detectable binding.
  • the assay was carried out following a previously described protocol (Lu et al., 1999; 2000). Various amounts of the diabodies were mixed with a fixed amount of KDR-AP (100 ng) or FIt-I -AP fusion protein (100 ng) and incubated at RT for 1 h. The mixture were then transferred to 96-well microtiter plates precoated with VEGFl 65 (200 ng/well) and incubated at RT for an additional 2 h, after which the plates were washed 5 times with PBS. The substrate for AP was added, followed by reading of the absorbance at 405nm to quantify the plate-bound KDR-AP or FIt-I -AP molecules. The IC50, i.e., the antibody concentration required for 50% inhibition of KDR or FIt-I binding to VEGF, was then calculated.
  • Fig. 4 shows that the diabodies block soluble KDR and FIt-I from binding to immobilized VEGF, in a dose-dependent manner.
  • the tetravalent di-diabody preparations were more potent blockers to both KDR/VEGF and Flt-1/VEGF interaction than the divalent diabody, DAB plCl 1/612.
  • the IC 50 values for KDR blocking were approximately 0.4 nM for both di-diabody preparations, compared to that of 1.2 nM for DAB plCl 1/612 and 0.8 nM for the monospecific bivalent DAB plCl 1.
  • the IC 50 for FIt-I blocking was approximately 1 nM for both di-diabody preparations, 8 nM for DAB plCl 1/612 and 0.2 nM for the monospecific bivalent DAB 612.
  • DAB plCl 1 had no effects on Flt-1/VEGF interaction
  • DAB 612 had no effects on KDR/VEGF interaction.
  • HEL human leukemia cell lines
  • U937 that only expresses FIt-I
  • Leukemia cells, HEL and U937 were washed three times with serum-free plain RPMI 1640 medium and suspended in the medium at 1 x 10 6 /ml. Aliquots of 100 ⁇ l cell suspension were added to 8- ⁇ m-pore transwell inserts (Costar®, Corning Incorporated, Corning, NY) and incubated with the antibodies at various concentrations, 100 nM, 25 nM and 6.25 nM, for 30 min at 37°C.
  • the inserts were then placed into the wells of 24-well plates containing 0.5 ml of serum-free RPMI 1640 with or without VEGF165.
  • the migration was carried out at 37°C, 5% CO2 for 4 h.
  • Migrated cells were collected from the lower compartments and counted under a light microscope.
  • the stability of the di-diabodies was tested by examination of their binding activity to both KDR and FIt-I.
  • the divalent bispecific diabody, DAB plCl 1/612, and the monospecific diabodies, DAB plCl 1 and DAB 612 retained full antigen binding activity to their respective targets after incubation at 37 0 C in both PBS and mouse serum.
  • the mammalian cell-derived di-diabody (M) preparation retained better antigen-binding activity compared to the E.c ⁇ /z-derived preparation.
  • di-diabody (M) showed 94% of the original KDR-binding activity, compared to that of 83% for di-diabody (B).
  • di-diabody (M) retained 77% and 92% binding activities to KDR and FIt-I, respectively, compared to those of 63% and 86% for di-diabody (B).
  • variable regions of a fully human anti- ⁇ GFR antibody (IMC-11F8) and a fully human anti-IGF-IR antibody (IMC-Al 2) were used to construct a bispecific di-diabody.
  • Bispecific diabodies were constructed as above, followed by fusion of one of the diabody cross-over scFv chains to the Fc domain of an IgG (see Fig. 7A and 7B for details).
  • Co-expression in mammalian cells the Fc fusion along with the other cross-over scFv resulted in an IgG-like tetravalent molecule with two binding specificities (Fig. 7B).
  • the di-diabody was produced by stably transfected NSO cells in serum-free conditions and purified from the cell culture supernatant via a Protein A affinity column. Electrophoresis analysis of the purified di-diabody under non-reducing conditions yielded two major protein bands with
  • the di-diabodv binds to both EGFR and IGFR
  • a direct binding assay was used to compare antigen-binding efficiency of the di-diabody with its monospecific counterparts.
  • IMC-Al 2 and BVIC-11F8 bound only to their respective targets, whereas the di-diabody reacted to both immobilized EGFR and IGFR, with moderately lower efficiencies as compared to its monospecific counterparts (Fig. 2B and 2C).
  • the ED50 values ⁇ i.e., the antibody concentrations that yield 50% of maximum binding) to EGFR were 0.05 nM for BVIC-11F8 and 0.1 - 0.2 nM for the Di-diabody, and to IGFR were 0.1 nM for BVIC-A12 and 0.25 - 0.5 nM for the Di-diabody.
  • the efficacy of the di-diabody in inhibiting tumor cell proliferation in vitro was established using a well-characterized tumor cell line, DiFi, which is known to depend on EGFR for survival and growth.
  • DiFi a well-characterized tumor cell line
  • the anti-EGFR antibody (BvIC-11F8) significantly inhibited the proliferation of DiFi cells, whereas the anti-IGFR antibody (DVIC-A12), and the control antibody (EvIC-1121; anti-KDR) had little effect (Fig. 9).
  • Proliferation of the DiFi cells was also inhibited by the di-diabody, although at higher concentrations than BVIC-11F8.
  • the IC50 values were approximately 1 nM for IMC-11F8 (alone), InM for the coadministered combination of IMC-11F8 and IMC-Al 2, and 25 nM for the di-diabody.
  • the di-diabody was assayed for efficacy in blocking EGF and IGF-stimulated receptor phosphorylation and downstream signal transduction.
  • Incubation of MCF-7 cells with EGF or IGF results in significant phosphorylation of the respective receptor; incubation with a combination of EGF and IGF yields activation of both EGFR and IGFR (Fig. 1OA, lanes 2, 3, and 4).
  • Fig. 1OA EGFR and IGFR
  • the di-diabody like the mixture of both EVIC-11F8 and EV ⁇ C-12, significantly blocked activation of both receptors (Fig. 1OA, lanes 5 and 8).
  • the control antibody, IMC-1121 showed no effect on the phosphorylation of EGFR and IGFR (Fig. 1OA, lane 9).
  • IMC-11F8 significantly inhibited the activation of MAPK but only moderately reduced the activation of Akt (Fig. 1OB, lane 7), whereas EVIC-A12 strongly reduced Akt phosphorylation but was less effective in p44/p42 MAPK activation (Fig. 1OB, lane 6).
  • the di-diabody effectively blocked phosphorylation of both Akt and p44/p42 MAPK induced by EGF and IGF (Fig. 1OB, lane 5) as did the combination of BVIC-A12 and IMC-11F8 (Fig. 10, lane 8).
  • IMC-A12 has been shown to induce rapid and efficient internalization and degradation of cell surface expressed IGFR (Burtrum et al., 2003, Cancer Res. 63:8912).
  • the di-diabody retained the receptor modulation activity of EVIC-A12 on tumor cells, triggering significant IGFR internalization and degradation in MCF-7 cells after incubation at 37°C for
  • antitumor IgG antibodies can also cause direct tumor cell killing via mediating effective ADCC.
  • the ability of the di-diabody to mediate tumor-cell killing in the presence of human efffector cells was examined. As shown in Fig. 12, BVIC-11F8 demonstrated lytic activity against both A431 and BxPC3 cells but was ineffective against MCF-7 cells. EVIC-Al 2 was effective only towards MCF-7 cells but failed to kill A431 and BxPC3 cells. The di-diabody, mediated killing of all the three tumor lines.
  • HT29 xenografts were less responsive to treatment with individual anti-EGFR or anti-IGFR antibodies than to treatment with the di- diabody (Fig. 13B).
  • NYOl 924757 vl that is functionally equivalent to IMC-11F8 regarding both in vitro and in vivo antitumor activity (Marie, P. et al., 2004, Proc. Amer. Assoc. Cancer Res. 45, Abstract #5353) resulted in tumor inhibition of 47% and 35% at day 40 post treatment, respectively (p ⁇ 0.03 compared to the saline and the human IgG groups).
  • the di-diabody yielded a tumor growth inhibition rate of 58%, which is comparable to that achieved by the combination of both IMC-C225 and IMC-A12 (63% tumor growth inhibition). There is no statistically significant difference in overall tumor inhibition, however, between groups treated with the di-diabody, the antibody combination, or the individual antibody at the end of the study.

Abstract

The invention is directed to novel tetravalent antibodies, which are preferably bi-specific. The tetravalent bispecific antibodies can be efficiently expressed in prokaryotic and eukaryotic cells, and are useful in therapeutic and diagnostic methods. The invention further describes administration of the antibodies, either alone or in combination with anti­angiogenic or anti-neoplastic drugs to inhibit tumor growth and/or angiogenesis.

Description

NOVEL TETRAVALENT BISPECIFIC ANTIBODY
FILED OF THE INVENTION
[0001] The present invention is directed to novel tetravalent antibodies, which are preferably bi-specific. The tetravalent bispecific antibodies can be efficiently expressed in prokaryotic and eukaryotic cells, and are useful in therapeutic and diagnostic methods. The invention further relates to combination therapies using the antibodies with anti-angiogenic or anti-neoplastic drugs.
BACKGROUND
[0002] Bispecific antibodies (BsAb) are immunoglobulin (Ig)-based molecules that bind to two different epitopes on either the same or distinct antigens. Both laboratory and early clinical studies have demonstrated that BsAb may have significant applications in cancer therapy by either targeting tumor cells with cytotoxic agents such as effector cells, radionuclides, drugs and toxins (Weiner et al. (1997) Cancer Immunol. Immunother. 45:190- 2.; van Spriel et al. (2000) Immuol. Today 21:391-7; Segal et al. (2000) J. Immunol. Methods 248:1-6.), or by simultaneously targeting two different tumor targets (or epitopes) in order to enhance the biological activities of individual antibody therapeutics (Lu et al. (1999) J. Immunol. Methods 230:159-71; Lu et al. (2001) Cancer Res. 61:7002-8.; Lu et al. (2002) J. Immunol. Methods 267:213-26). A major obstacle in the development of BsAb-based therapeutics has been the difficulty in producing the materials in sufficient quantity and quality for clinical studies via traditional methods, including the hybrid hybridoma and chemical conjugation (Carter et al. (1995) J. Hematotherapy 4:463-70). Coexpression of two different sets of IgG light and heavy chains in hybrid hybridoma may produce up to 10 light- and heavy-chain pairs, with only one of these pairs the functional bispecific heterodimer (Suresh et al. (1986) Methods Enzymol. 121:210-28). On the other hand, chemical crosslinking of two IgGs or their fragments is often inefficient and can lead to the loss of antibody activity (Zhu et al. (1994) Cancer Lett. 86:127-34). In both methods, purification of the BsAb from the non-functional species, such as homodimers and mispaired heterodimers of non-cognate Ig light and heavy chains produced by the hybrid hybridoma, and multimeric aggregates resulting from chemical conjugation, is often difficult and the yield is usually low (Cao et al. (1998) Bioconj. Chem. 9:635-44).
[0003] To improve efficiency, a variety of recombinant methods have been developed for efficient production of BsAb, both as antibody fragments (Carter et al. (1995); Pluckthun et al (1997) rmmunotechology 3:83-105; Todorovska et al. (2001) J. Immunol. Methods 248:47-66) and full length IgG formats (Carter (2001) J. Immunol. Methods 248:7-15). For example, production of homogeneous full-length IgG-like BsAb has been achieved by the so- called "knobs-into-holes" engineering for efficient Ig CH3 domain heterodimerization (Ridgway et al. (1996) Protein Eng. 9:617-21; Merchant et al. (1998) Nat. Biotech. 16:677- 81) and by fusing two single chain Fv (scFv) of different specificities onto either the N- or the C-terminus of a full-length IgG molecule (Zhuang et al. (2000) Protein Eng. 13:361-7; Coloma and Morrison (1997) Nat. Biotechnol. 15:159-63). BsAbs have also been constructed by genetically fusing two single chain Fv (scFv) or Fab fragments with or without the use of flexible linkers (Mallender et al. (1994) J. Biol. Chem. 269:199-206; Mack et al. (1995) Proc. Natl. Acad. Sci. USA. 92:7021-5; Zapata et al. (1995) Protein Eng. 8:1057-62), via a dimerization device such as leucine zipper (Kostelny et al. (1992) J. Immunol. 148:1547-53; de Kruif et al. (1996) J. Biol. Chem. 271:7630-4), and Ig CL/CHI domains (Muller et al. (1998) FEBS Lett. 422:259-64); by diabody (Holliger et al. (1993) Proc. Nat. Acad. Sci. USA. 90:6444-8; Zhu et al. (1996) Bio/Technology (NY) 14:192-6); Fab-scFv fusion (Lu et al. (2002); Schoonjans et al.(2000) J. Immunol. 165:7050-7); and miniantibody formats (Pack et al. (1992) Biochemistry 31:1579-84; Pack et al. (1993) Bio/Technology 11:1271-7). In majority of the cases, these recombinant approaches result in the production of divalent bispecific antibody molecules that are monovalent to each of their target antigens.
[0004] A diabody is a form of scFv dimer which is constructed with a short linker (5- 10 amino acid residues) between the variable heavy (VH) and the variable light (VL) domains. The short linker prevents intrachain pairing of VH and the VL domains, but allows interchain pairing of VH and the VL domains, such that a bivalent antibody fragment with a molecular size of approximately 55 to 60 kDa is formed. (Holliger et al. (1993); Whitlow et al. (1994) Protein Eng. 7: 1017-26; Zhu et al. (1996)). A bispecific diabody can be produced by coexpression of two "cross-over" scFv fragments in which the VH and the VL domains for
- 2 -
NYOl 924757 vl each of the two binding sites are present on different polypeptide chains (Holliger et al. (1993); Zhu et al. (1996)). Interchain pairing of the two "cross-over" polypeptide chains results in the formation of a scFv dimer, or bispecific diabody, which is a divalent antibody molecule that is monovalent for each of its target antigens.
SUMMARY OF THE INVENTION
[0005] The invention provides a novel tetravalent bispecific antibody (BsAb) molecule. The BsAb comprises two identical diabodies (DAB), each having two antibody binding sites, preferably non-identical. One of the diabody chains includes an immunoglobulin hinge region and at least one heavy chain constant domain that is capable of self-association to form a dimer (e.g., a CH2 and/or a CR3 domain).
[0006] These novel proteins, termed "di-diabodies," offer several advantages as compared to previously described (BsAbs). Di-diabodies can be produced that are bispecific and bivalent. Di-diabodies have no constraints regarding selection of antigen-binding sites, and Fc constant domains and associated functions can be retained. The proteins are substantially homogeneous and can be produced bacterial, mammalian, or other cells without further processing. Notably, because the binding characteristics of bispecific di-diabodies for each of two different antigens are comparable to those of natural antibodies (i.e., full, monospecific divalent IgG molecules naturally produced by a host) for each antigen, a single pharmaceutical di-diabody preparation can be used in place of a combination of two monospecific antibodies. The di-diabodies can also be monospecific (i.e., four identical antibody binding sites). Further, the di-diabodies of the invention are efficiently produced in mammalian cells, and can be purified by conventional protein A chromatography in a single step.
[0007] Di-diabodies of the invention comprise at least one IgG constant domain that is capable of self association, such as, for example, CH2 or CH3 of IgG, IgA, or IgD, or CH2, CH3, or CR4 of IgE or IgM. Accordingly, di-diabodies possess the immunological functions associated with such domains that are present, such as Fc receptor binding, complement mediated cytotoxicity (CMC), or antibody dependent cell-mediated cytoxicity (ADCC).
[0008] hi certain embodiments, di-diabodies of the invention bind to and block activation of a growth factor receptor such as, but not limited to, VEGFR-I, VEGFR-2,
- 3 -
NYOl 924757 vl EGFR or IGFR. In certain embodiments, the di-diabodies bind to and block activation of two such receptors, hi other embodiments, di-diabodies are capable of simultaneous binding to a growth factor receptor on a target cell, and to a cell surface antigen of an immune system effector cell.
[0009] Di-diabodies of the invention are used to neutralize growth factor receptors, and to reduce tumor growth and/or inhibit angiogenesis associated with activation of such receptors. Further, the di-diabodies can be administered with anti-neoplastic agents such as chemotherapeutic agents or radiation.
BRIEF DESCRIPTION OF THE FIGURES
[0010] Figure 1 is a schematic diagram depicting expression constructs for various antibody fragments, hi the di-diabody, the first of two scFv polypeptide chains was fused to an IgG hinge region and CH3 domain. A diabody results from association of the first and second scFv chains. A tetrameric structure is formed by two diabodies associated by the hinge and CH3 domains. G4S: a 5 amino-acid linker "GGGGS". E: the 13 amino-acid E-tag.
[0011] Figure 2 shows SDS-PAGE analysis of the purified antibody fragments. Panel A: non-reducing conditions; Panel B: reducing conditions; lane 1: diabody (DAB) pi CIl; lane 2: DAB 612; lane 3: DAB plCl 1/612; lane 4: Di-diabody (B); lane 5, di-diabody (M). Also shown are molecular weight standards run under non-reducing conditions (from top to bottom: 176.5, 113.7, 80.9, 63.8, 49.5, 37.4, 28.0, and 19.6 kDa, respectively).
[0012] Figure 3 shows bispecific and dose-dependent binding of a di-diabody specific for KDR and FIt-I . Top panel: cross-linking assay using FIt-I -coated support and soluble KDR-AP. Middle and bottom panels: various concentrations of antibody fragments were bound to a KDR or FIt-I coated support, then incubated with a secondary HRP conjugated reagent. Following incubation with a peroxidase, OD450nm was read. Data shown represent the mean ± SD of triplicate samples.
[0013] Figure 4 shows inhibition of binding of KDR and FIt-I to immobilized VEGF by the bispecific antibody fragments. Various concentrations of antibody fragments were incubated with a fixed amount of KDR-AP or FIt-I -AP fusion proteins. Bound KDR-AP or Flt-l-AP was quantified by incubation with a peroxidase substrate and OD405nm measurement. Data shown represent the mean ± SD of triplicate samples.
_ 4 _
NYOl 924757 vl [0014] Figure 5 shows inhibition of VEGF-induced migration of human leukemia cells by the bispecific antibody fragments. HEL or U937 cells on supports were incubated with various concentrations of antibodies, then placed into wells containing VEGF. Migrated cells were collected and counted. Data shown are the mean ± SD of triplicate determinations.
[0015] Figure 6 shows the stability of di-diabody preparations incubated in PBS or mouse serum. The di-diabodies were incubated with PBS or serum at 370C for up to 72 hours. Data are expressed as percentage of antigen binding activity of each antibody preparation retained after 37°C incubation in comparison to that of samples incubated at 40C in PBS. Data shown represent the mean ± SD of triplicate samples.
[0016] Figure 7 depicts construction and production of a di-diabody having binding sites specific for EGFR and IGFR. Panels (A) and (B) are schematic diagrams showing the expression constructs. L: Linker (Arg-Thr-Val-Ala-Ala); H: IgGl hinge region. Purified di- diabody preparations were analyzed by SDS-PAGE under non-reducing (C) or reducing (D) conditions. Lane 1 : IMC-11F8 IgG; lane 2: the di-diabody. Also shown on the left side are molecular weight standards.
[0017] Figure 8 shows bispecific and dose-dependent binding of the EGFR x IGFR di-diabody. (A) Receptor cross-linking assay. Antibody preparations were first incubated with a biotin-labeled IGFR in solution and then transferred to a microtiter plate coated with EGFR, followed by incubation with streptoavidin-HRP to measure the plate-bound biotin activity. (B) and (C) Dose-dependent binding of the di-diabody to immobilized EGFR and IGFR. Various amounts of antibodies were incubated in 96-well plates coated with human EGFR (B) or IGFR extracellular domain (C)washed, and incubated with rabbit anti-human IgG Fc-HRP conjugate. Peroxidase substrate was added, and OD450nm was measured. Data shown represent the mean ± SD of triplicate samples.
[0018] Figure 9 shows antiproliferative activity of the EGFR x IGFR di-diabody. Various amounts of the antibodies were incubated with DiFi cells in culture for 4 days followed by incubation with MTT for 4 h. The plates were washed and incubated with HCl/isopropanol at RT for 10 min, followed by optical density reading at 570nm. Data shown represent the mean ± SD of triplicate samples.
[0019] Figure 10 shows inhibition of EGF and IGF-stimulated activation of receptors and downstream signaling molecules, Akt and MAPK p44/p42. MCF-7 cells were incubated
- 5 -
NYOl 924757 vl with various antibodies at 37°C for 30 min, followed by stimulation with EGF, IGF, or both at 37°C for 20 min. Phosphorylation of EGFR, IGFR, as well as Akt and MAPK p44/p42 was analyzed. Lane 1, no treatment; lane 2, IGF alone; lane 3, EGF alone; lane 4, IGF plus EGF; lane 5 to 9, IGF plus EGF in the presence of the di-diabody (lane 5), IMC-Al 2 (lane 6), IMC-11F8 (lane 7), IMC-A12 plus IMC-11F8 (lane 8), or negative control (lane 9, IMC- 1121, anti-VEGFR2). Results shown are the representative of three separate experiments. Similar results were also observed when BxPC3 and HT29 cells were used as the target cells.
[0020] Figure 11 shows IGFR internalization and degradation in tumor cells. Tumor cells were plated and incubated overnight in serum-free medium. IGF-I, EGF (50 nM), IGF plus EGF (I + E) or various antibodies was then added and incubated at 37°C for up to 4 h. Cells were washed in ice-cold PBS, lysed, and electrophoresed using 4-12% Tris-Glycine gels. Proteins were transferred to nitrocellulose membranes and were detected by Western blotting using antibody C-20 (rabbit anti-IGFR) and IMC-11F8 (human anti-EGFR), followed by an anti-rabbit or anti-human antibody-HRP conjugate. The signals were visualized with the ECL reagent. (A) The di-diabody induced IGFR degradation in a dose- dependent manner in MCF-7 cells. (B) The di-diabody (100 nM) induced IGFR degradation as efficiently as EVIC-A12 and the combination of IMC-A12 and IMC-11F8 after 4 h incubation at 37°C. (C) The Di-diabody (100 nM) induced IGFR degradation in BxPC3 cells in a time-dependent manner when incubated at 370C. DAb, the Di-diabody; A + F, IMC-A12 plus IMC-11F8; 1121, the control antibody, MC-1121.
[0021] Figure 12 shows di-diabody mediated ADCC in the presence of human PBMC. Tumor cells were incubated in the wells of 96-well plates with IMC-11F8, IMC- A12, the Di-diabody, or a normal human IgG at 37°C for 30 min, followed by addition of PBMC at an effector/tumor cell ratios of 100/1 and incubation at 37°C for an additional 4 h. After centrifugation at 1500 rpm for 10 min, 100 μl of supernatant was transferred to 96 well flat bottom plates, followed by the addition of 100 μl/well LDH assay reagent and reading of the absorbance at 490nm. The percentages of specific cell lysis were calculated as described in the Materials and Methods. Data represent the means of triplicate determinations.
[0022] Figure 13 shows inhibition of growth of human tumor xenografts in nude mice by the di-diabody. Female athymic nu/nu mice were injected subcutaneously on the lateral dorsal surface with BxPC3 (2 x 106/mouse) or HT-29 (5 x 106/mouse) tumor cells. When
- 6 -
NYOl 924757 vl tumors reached approximately 200-300 mm3 mice were randomized by tumor size and divided into treatment groups. The mice were treated by IMC-11F8 (or the equivalent IMC- C225), IMC-A12, IMC-11F8 (or IMC-C225) plus IMC-A12, the di-diabody, or the control articles (saline or normal human IgG). Each antibody was administered by intraperitoneal injections at 40 mg/kg twice a week. Tumor volume and body weight of each animal was measured twice a week. Data represent the mean ± SE of tumor sizes from 10 to 12 animals in each group.
DETAILED DESCRIPTION OF THE INVENTION
[0023] The invention provides an antigen-binding protein which is a tetravalent BsAb molecule, comprising two identical diabodies. Each diabody is a heterodimer of two polypeptide chains and is preferably bispecific. According to the invention, one of the polypeptides of the diabody is fused, via a hinge region, to the N-terminus of an immunoglobulin constant domain that is capable of stable self association (e.g., CH2 and/or CR3 if IgGl). Coexpression of the two polypeptide chains in a single host results in the formation of bispecific diabody-constant domain fusion, which in turn dimerizes via both the hinge and the constant domain to form a bispecific tetravalent protein (a "di-diabody"). The di-diabody can be efficiently expressed in E.coli and mammalian cells, and is more efficient in binding to its targets and in blocking target-associated biological activities than the corresponding diabody.
[0024] Antibody specificity refers to selective recognition of the antibody for a particular epitope of an antigen. Natural antibodies, for example, are monospecific. Bispecific antibodies (BsAbs) are antibodies which have two different antigen-binding specificities or sites. Where an antigen-binding protein has more than one specificity, the recognized epitopes may be associated with a single antigen or with more than one antigen.
[0025] A natural antibody molecule is composed of two identical heavy chains and two identical light chains. Each light chain is covalently linked to a heavy chain by an interchain disulfide bond. The two heavy chains are further linked to one another by multiple disulfide bonds at the hinge region. The individual chains fold into domains having similar sizes (about 110-125 amino acids) and structures, but different functions. The light chain comprises one variable domain (VL) and one constant domain (CL). The heavy chain
- 7 -
NYOl 924757 vl comprises one variable domain (VH) and, depending on the class or isotype of antibody, three or four constant domains (CHI, CH 2, CH3 and CH4). In mice and humans, the isotypes are IgA, IgD, IgE, IgG, and IgM, with IgA and IgG further subdivided into subclasses or subtypes. The portion of an antibody consisting of VL and VH domains is designated "Fv" and constitutes the antigen-binding site. A single chain Fv (scFv) is an engineered protein containing a VL domain and a VH domain on one polypeptide chain, wherein the N terminus of one domain and the C terminus of the other domain are joined by a flexible linker. "Fab" refers to the portion of the antibody consisting of VL, VH, CL and CHI domains.
[0026] The variable domains show considerable amino acid sequence variablity from one antibody to the next, particularly at the location of the antigen binding site. Three regions, called "hypervariable" or "complementarity-determining regions" (CDRs) are found in each of VL and VH-
[0027] "Fc" is the designation for the portion of an antibody which comprises paired heavy chain constant domains, hi an IgG antibody, for example, the Fc comprises CR2 and CH3 domains. The Fc of an IgA or an IgM antibody further comprises a CH4 domain. The Fc is associated with Fc receptor binding, activation of complement-mediated cytotoxicity and antibody-dependent cellular-cytotoxicity. For natural antibodies such as IgA and IgM, which are complexes of multiple IgG like proteins, complex formation requires Fc constant domains.
[0028] Finally, the "hinge" region separates the Fab and Fc portions of the antibody, providing for mobility of Fabs relative to each other and relative to Fc, as well as including multiple disulfide bonds for covalent linkage of the two heavy chains.
[0029] Diabodies contain two polypeptide chains which when associated, form two antibody binding sites. Each polypeptide chain contains an immunoglobulin heavy chain variable (VH) domain and an immunoglobulin light chain variable (VL) domain, joined by short peptide linker (5-10 amino acids). Notably, the order of the VH and VL domains of one polypeptide chain is reversed with respect to the other (i.e., Vπ-linker-VL and VL-linker-Vπ) Formation of heterodimers of the two non-identical chains is promoted by the combination of short peptide linkers, which inhibit intrachain VH-VL association, and the order of the variable domains of the two polypeptides. Accordingly, in the heterodimer, VH of the first polypeptide is paired with VL of the second polypeptide, and VH of the second polypeptide is
- 8 -
NYQl 924757 vl paired with VL of the first polypeptide. Where the VH domains are identical and the VL domains are identical, two heterodimer contains two identical binding sites. Alternatively, two different VH domains and two different corresponding VL domains may be employed such that the heterodimer contains two different binding sites and is bispecific (i.e., VB1- linker-Vu and VL1 -linker- VH2 ; see Fig. 1). The bispecific di-diabody that results from linking two such bispecific diabodies, is thus bivalent for each of the two different binding sites.
[0030] Di-diabodies of the invention which are bispecific have a combination of desirable features. First, they are essentially homogeneous. By design, mispairing of antibody heavy and light chains is greatly reduced or eliminated. For example, a typical bispecific antibody requires the use of two different heavy chains to provide two specificities. Four combinations are possible when the heavy chains are arranged into an IgG type molecule. Two of those consist of mispaired heavy chains such that the product is monospecific. Contrarywise, in proteins of the invention, mispairing is substantially eliminated. Pairing of two diabody polypeptides by heterodimerization is favored, and dimerization of the bispecific diabodies is by a different interaction. Accordingly, the products are bispecific.
[0031] A second advantage of the di-diabodies is that they are bivalent for each binding specificity. A feature of a natural antibody which is missing from a dimeric BsAb is that the natural antibody is bivalent for the antibody binding site that it comprises. A dimeric BsAb is monovalent for each of the two binding sites that it comprises. This is significant for antibody function because bivalency allows for cooperativity of binding and a significant increase in binding avidity with resepect to a molecule comprising a single antigen-binding site.
[0032] A third advantage of the di-diabodies is that heavy chain constant domains which constitute the Fc region (e.g., CH2 and/or CH3 for an IgGl molecule) of a natural antibody and which provide other antibody functions can be present. Furthermore, the multiple binding domains are separated from the constant domains such that functions provided by the constant domains are not impaired. Constant domain functions include binding to certain accessory molecules (e.g., binding to cell surface and soluble Fc receptors, J chain association for IgA and IgM, S protein for IgA), activation of the complement
- 9 -
NYOl 924757 vl pathway (complement mediated cytoxicity, CMC), recognition of antibody bound to target cells by several different leukocyte populations (antibody-dependent cell-mediated cytoxicity, ADCC) and opsonization (enhancement of phagocytosis). Also, the Fc heavy chain constant domain(s) can confer increased serum half-life.
[0033] A fourth advantage of proteins of the invention is that there is no requirement for processing in vitro to obtain the complete product. Though rearranged in an artificial manner, each of the domains has a natural character which allows expression in a biological system. For example, di-diabodies can be expressed in prokaryotic and eukaryotic expression systems. The proteins that are produced are substantially bispecific. Although multimeric forms of the bispecific di-diabody may be expressed to some degree (see Examples), these are easily removed from preparations by chromatography if necessary.
[0034] The present invention is also applicable to production of monospecific tetravalent antigen-binding proteins. In such proteins, all four binding sites of the di-diabody have the same specificity.
[0035] An antigen binding site for use in a di-diabody can be obtained by a variety of methods. The amino acid sequences of the VH and VL portions of a selected binding domain correspond to a naturally-occurring antibody or are chosen or modified to obtained desired immunogenic or binding characteristics. For example, VH and VL domains can be obtained directly from a monoclonal antibody which has the desired binding characteristics. Alternatively, VH and VL domains can be from libraries of V gene sequences from a mammal of choice. Elements of such libraries express random combinations of VH and VL domains and are screened with any desired antigen to identify those elements which have desired binding characteristics. Particularly preferred is a human V gene library. Methods for such screening are known in the art. VH and VL domains from a selected non-human source may be incorporated into chimeric di-diabodies. For example, for administration to a human, it may be desired to use a di-diabody with one or more functional human constant domains wherein the VH and VL domains have been selected from a non-human source. To maximize constant domain associated function or to reduce immunogenicity of the antibody, human constant domains are preferred.
[0036] Alternatively, a di-diabody can be made that is "humanized." Humanized variable domains are constructed in which amino acid sequences which comprise one or more
- 10 -
NYOl 9247S7 vl complementarity determining regions (CDRs) of non-human origin are grafted to human framework regions (FRs). For examples, see: Jones, P. T. et al., (1996) Nature 321, 522-525; Riechman, L. et al., (1988) Nature 332, 323-327; U.S. Patent No. 5,530,101 to Queen et al. A humanized construct is particularly valuable for elimination of adverse immunogenic characteristics, for example, where an antigen binding domain from a non-human source is desired to be used for treatment in a human. Variable domains have a high degree of structural homology, allowing easy identification of amino acid residues within variable domains which corresponding to CDRs and FRs. See, e.g., Kabat, E.A., et al. (1991) Sequences of Proteins of Immunological Interest. 5th ed. National Center for Biotechnology Information, National Institutes of Health, Bethesda, MD. Thus, amino acids which participate in antigen binding are easily identified, hi addition, methods have been developed to preserve or to enhance affinity for antigen of humanized binding domains comprising grafted CDRs. One way is to include in the recipient variable domain the foreign framework residues which influence the conformation of the CDR regions. A second way is to graft the foreign CDRs onto human variable domains with the closest homology to the foreign variable region. Queen, C. et al., (1989) Proc. Natl. Acad. Sci. USA 86, 10029-10033. CDRs are most easily grafted onto different FRs by first amplifying individual FR sequences using overlapping primers which include desired CDR sequences, and joining the resulting gene segments in subsequent amplification reactions. Grafting of a CDR onto a different variable domain can further involve the substitution of amino acid residues which are adjacent to the CDR in the amino acid sequence or packed against the CDR in the folded variable domain structure which affect the conformation of the CDR. Humanized variable domains of the invention therefore include human domains which comprise one or more non-human CDRs as well as such domains in which additional substitutions or replacements have been made to preserve or enhance binding characteristics.
[0037] Di-diabodies of the invention may also employ variable domains which have been made less immunogenic by replacing surface-exposed residues to make the di-diabody appear as self to the immune system (Padlan, E.A. (1991) MoI. Immunol. 28, 489-498). Antibodies have been modified by this process with no loss of affinity (Roguska et al. (1994) Proc. Natl. Acad. Sci. USA 91, 969-973). Because the internal packing of amino acid residues in the vicinity of the antigen binding site remains unchanged, affinity is preserved.
- 11 -
NYOl 924757 vl Substitution of surface-exposed residues according to the invention for the purpose of reduced immunogenicity does not mean substitution of CDR residues or adjacent residues which influence binding characteristics.
[0038] The invention contemplates binding domains which are essentially human. Human binding domains are obtained from phage display libraries wherein combinations of human heavy and light chain variable domains are displayed on the surface of filamentous phage (See, e.g., McCafferty et al. (1990) Nature 348, 552-554; Aujame et al. (1997) Human Antibodies 8, 155-168). Combinations of variable domains are typically displayed on filamentous phage in the form of Fabs or scFvs. The library is screened for phage bearing combinations of variable domains having desired antigen binding characteristics. Preferred variable domain combinations display high affinity for a selected antigen and little cross- reactivity to other related antigens. By screening very large repertoires of antibody fragments, (see e.g., Griffiths et al. (1994) EMBO J. 13, 3245-3260) a good diversity of high affinity Mabs are isolated, with many expected to have sub-nanomolar affinities for the desired antigen.
[0039] Alternatively, human binding domains can be obtained from transgenic animals into which unrearranged human Ig gene segments have been introduced and in which the endogenous mouse Ig genes have been inactivated (reviewed in Bruggemann and Taussig (1997) Curr. Opin. Biotechnol. 8, 455-458). Preferred transgenic animals contain very large contiguous Ig gene fragments that are over 1 Mb in size (Mendez et al. (1997) Nature Genet. 15, 146-156) but human Mabs of moderate affinity can be raised from transgenic animals containing smaller gene loci (See, e.g., Wagner et al. (1994) Eur. J. Immunol. 42, 2672-2681; Green et al. (1994) Nature Genet. 7, 13-21).
[0040] In a physiological immune response, mutation and selection of expressed antibody genes leads to the production of antibodies having high affinity for their target antigen. The VH and VL domains incorporated into di-diabodies of the invention can similarly be subject to in vitro mutation and screening procedures to obtain high affinity variants. Thus, binding domains of the invention include those for which binding characteristics have been improved by direct mutation or by methods of affinity maturation. Affinity and specificity may be modified or improved by mutating CDRs and screening for antigen binding sites having the desired characteristics (See, e.g., Yang et al. (1995) J. MoI.
- 12 -
NYOl 924757 vl Bio. 254, 392-403). CDRs are mutated in a variety of ways. One way is to randomize individual residues or combinations of residues so that in a population of otherwise identical antigen binding sites, all twenty amino acids, or a subset thereof, are found at particular positions. Alternatively, mutations are induced over a range of CDR residues by error prone PCR methods (See, e.g., Hawkins et al. (1992) J. MoI. Bio. 226, 889-896). Phage display vectors containing heavy and light chain variable region genes are propagated in mutator strains of E. coli (See, e.g., Low et al. (1996) J. MoI. Bio. 250, 359-368). These methods of mutagenesis are illustrative of the many methods known to one of skill in the art.
[0041] Each variable domain of the antibodies of the present invention may be a complete immunoglobulin heavy or light chain variable domain, or it may be a functional equivalent or a mutant or derivative of a naturally occurring domain, or a synthetic domain constructed, for example, in vitro using a technique such as one described in WO 93/11236 (Medical Research Council / Griffiths et al.). For instance, it is possible to incorporate domains corresponding to antibody variable domains which are missing one or more amino acids. The important characterizing feature is the ability of each variable domain to associate with a complementary variable domain to form an antigen binding site.
[0042] Antigen-binding proteins of the invention have binding sites for any epitope, antigenic site or protein. Of particular interest are di-diabodies that are useful for treatment of disease. Preferred di-diabodies neutralize receptor proteins, such as receptors which are involved in angiogenesis and/or oncogenesis. Neutralizing a receptor means inactivating the intrinsic kinase activity of the receptor to transduce a signal. A reliable assay for receptor neutralization is the inhibition of receptor phosphorylation. The present invention is not limited by any particular mechanism of receptor neutralization. Some possible mechanisms include preventing binding of the ligand to the extracellular binding domain of the receptor, and preventing dimerization or oligomerization of receptor. Other mechanisms cannot, however, be ruled out.
[0043] Neutralization of activation of a receptor in a sample of endothelial or non- endothelial cells, such as tumor cells, may be performed in vitro or in vivo. Neutralizing activation of a receptor in a sample of receptor expressing cells comprises contacting the cells with an antibody of the invention. In vitro, the cells are contacted with the antibody before, simultaneously with, or after, adding VEGF to the cell sample. In vivo, an antibody of the
- 13 -
NYOl 924757 vl invention is contacted with a receptor by administration to a mammal. Methods of administration to a mammal include, for example, oral, intravenous, intraperitoneal, subcutaneous, or intramuscular administration.
[0044] Examples of such receptors include, but are not limited to VEGF receptors (e.g., VEGFR-2/KDR/Flk-l, VEGFR2/F1M, VEGFR3/Flt-4), epidermal growth factor receptor (EGFR), insulin-like growth factor receptor (IGFR) and the like. Additional non- limiting examples of receptor tyrosine kinases include Flt-4, HER2/neu, Tek and Tie2.
[0045] Other factors implicated as possible regulators of angiogenesis and/or growth of tumors in vivo include fibroblast growth factor (FGF), platelet derived growth factor (PDGF), and nerve growth factor (NGF). The corresponding receptors are fibroblast growth factor (FGF-R), platelet derived growth factor receptor (PDGF-R), and nerve growth factor receptor (NGFR). Another receptor implicated in cell migration, morphology changes, and invasiveness is macrophage-stimulating protein receptor ("MSP-R" or "RON"). Receptors of interest include human proteins and homologues from other mammals.
[0046] Di-diabodies can incorporate immunoglobulin binding domains from any source. For example, antibodies are known for the above listed receptors and are sources of VH and VL domains for use in di-diabodies of the present invention. Examples of binding domains specific for KDR include IMC-ICl 1 (nucleotide and amino acids sequences of VH: SEQ ID NOS:1 and 2; nucleotide and amino acid sequences of VL: SEQ ID NOS:3 and 4) (see, WO 00/44777), EVIC-2C6 (nucleotide and amino acids sequences of VH: SEQ ID NOS: 5 and 6; nucleotide and amino acid sequences of VL: SEQ ID NOS:7 and 8) (see, WO 03/075840), and IMC-1121 (nucleotide and amino acids sequences of VH: SEQ ID NOS: 5 and 6; nucleotide and amino acid sequences of VL: SEQ ID NOS:9 and 10) (see, WO 03/075840). Examples of binding domains specific for FIt-I include 6.12 (nucleotide and amino acids sequences of VH: SEQ ID NOS: 11 and 12; nucleotide and amino acid sequences of VL: SEQ ID NOS:13 and 14) and BVIC-18F1 (nucleotide and amino acids sequences of VH: SEQ ID NOS :27 and 28; nucleotide and amino acid sequences of VL: SEQ ID NOS :29 and 30).
[0047] Binding domains specific for EGFR include, for example, ERBITUX® (Cetuximab; IMC-C225) (nucleotide and amino acids sequences of VH: SEQ ID NOS: 15 and 16; nucleotide and amino acid sequences of VL: SEQ ID NOS: 17 and 18) as disclosed in WO
- 14 -
NYOl 924757 vl 96/40210 and IMCl 1F8 (nucleotide and amino acids sequences of VH: SEQ ID NOS: 19 and 20; nucleotide and amino acid sequences of VL: SEQ ID NOS:21 and 22). An example of a binding domain specific for IGFR is IMC-A12 (nucleotide and amino acids sequences of VH: SEQ ID NOS: 23 and 24; nucleotide and amino acid sequences of VL: SEQ ID NOS: 25 and 26). Antibodies that bind to FGF receptors include, for example, FR1-H7, FRl-Al, and FRl -4H (WO 2005/037235). Antibodies that bind to PDGFRα include, for example, 3G3 and 7Gl 1 (Loizos et al., 2005, MoI. Cancer Ther. 4:369). Antibodies that bind to RON or
MSP-R include IMC-41 AlO and IMC-41 A12 (Pereira, International application , filed 13-May-2005).
[0048] Further, portions of the above listed binding domains, such as the CDR regions, may be incorporated into binding domains used to make the binding proteins described herein.
[0049] Certain preferred diabodies bind to two of the above listed receptors. In one preferred embodiment, such a bispecific antigen-binding protein binds to KDR and FLT-I. An example of such an antigen binding protein has two polypeptide chains as provided in the examples (nucleotide and amino acid sequences: first polypeptide - SEQ ID NOS:51 and 52; second polypeptide - SEQ ID NOS :53 and 54). For each polypeptide, the linkers between the heavy and light chain variable domains are Gly-Gly-Gly-Gly-Ser, but a linker of 0 to 10 amino acids having any amino acid sequence can be used. The antigen-binding protein incorporates a CH3 domain but not a CH2 domain. A linker (Gly-Gly-Gly-Gly-Ser)2 is incorporated between the hinge region and the IgGl CJJ3 domain, but a linker of 0 to 30 amino acids having any amino acid sequence can be used.
[0050] In a second preferred embodiment, an antigen-binding protein of the invention binds to HER2 and EGFR. hi yet another preferred embodiment, a di-diabody of the invention binds to EGFR and IGFR. An example of an antigen binding protein that binds to EGFR and IGFR is provided in the examples (nucleotide and amino acid sequences: first polypeptide - SEQ ID NOS:55 and 56; second polypeptide - SEQ ID NOS:57 and 58). In this example, the linkers between the heavy and light chain variable domains are Arg-Thr-Val- AIa- Ala. The antigen-binding protein incorporates CH2 and CH3 constant domains, and no linker was used between the hinge region and the constant domains.
- 15 -
NYOl 924757 vl [0051] In another embodiment, an antigen-binding protein of the invention binds to EGFR and a VEGFR. In a preferred embodiment, the VEGFR is VEGFR2. Such an antibody is useful for blocking stimulation of vascular epithelial cells, by blocking signal transduction through both EGFR and VEGFR. This is particularly useful where angiogenesis occurs in response to EGFR ligands, particularly TGRα, secreted by tumor cells.
[0052] Di-diabodies of the invention can be used to cross-link antigens on target cells with antigens on immune system effector cells. This can be useful, for example, for promoting immune responses directed against cells which have a particular antigens of interest on the cell surface. According to the invention, immune system effector cells include antigen specific cells such as T cells which activate cellular immune responses and nonspecific cells such as macrophages, neutrophils and natural killer (NK) cells which mediate cellular immune responses.
[0053] Di-diabodies of the invention can have a binding site for any cell surface antigen of an immune system effector cell. Such cell surface antigens include, for example, cytokine and lymphokine receptors, Fc receptors, CD3, CD 16, CD28, CD32 and CD64. In general, antigen binding sites are provided by scFvs which are derived from antibodies to the aforementioned antigens and which are well known in the art. Antigen-binding sites of the invention which are specific for cytokine and lymphokine receptors can also be sequences of amino acids which correspond to all or part of the natural ligand for the receptor. For example, where the cell-surface antigen is an IL-2 receptor, an antigen-binding protein of the invention can have an antigen-binding site which comprises a sequence of amino acids corresponding or IL-2. Other cytokines and lymphokines include, for example, interleukins such as interleukin-4 (IL-4) and interleukin-5 (IL-5), and colony-stimulating factors (CSFs) such as granulocyte-macrophage CSF (GM-CSF), and granulocyte CSF (G-CSF).
[0054] Di-diabodies of the invention are made by expressing two diabody chains, one of which is linked to at least one heavy chain constant domain that is capable of dimerization (e.g., CH2 and/or CH3). Di-diabodies are conveniently produced in E. coli using DNA constructs which comprise bacterial secretion signal sequences at the start of each polypeptide chain. A variety of bacterial signal sequences are known in the art. A perferred signal sequence is from the pelB gene of Erwinia carotovora. The DNA fragments coding for the diabodies can be cloned, e.g., into vectors employing human cytomegalovirus
- 16 -
NYOl 924757 vl (HCMV) promoters and enhancers for high level expression in mammalian cells, such as, for example, CHO, NSO, COS-7, and PER.C6 cells, and cell lines of lymphoid origin such as lymphoma, myeloma, or hybridoma cells. {See, e.g., Bendig, et al., U.S. Patent 5,840,299; Maeda, et al. (1991) Hum. Antϊbod. Hybridomas 2, 124-134).
[0055] A selectable marker is a gene which encodes a protein necessary for the survival or growth of transformed host cells grown in a selective culture medium. Typical selectable markers encode proteins that (a) confer resistance to antibiotics or other toxins, e.g. ampicillin, neomycin, methotrexate, or tetracycline, (b) complement auxotrophic deficiencies, or (c) supply critical nutrients not available from complex media, e.g. the gene encoding D-alanine racemase for Bacilli. A particularly useful selectable marker confers resistance to methotrexate. For example, cells transformed with the DHFR selection gene are first identified by culturing all of the transformants in a culture medium that contains methotrexate (Mtx), a competitive antagonist of DHFR. An appropriate host cell when wild-type DHFR is employed is the Chinese hamster ovary (CHO) cell line deficient in DHFR activity, prepared and propagated as described by Urlaub and Chasin (1980) Proc. Natl. Acad. ScL USA 11, 4216. The transformed cells are then exposed to increased levels of methotrexate. This leads to the synthesis of multiple copies of the DHFR gene, and, concomitantly, multiple copies of other DNA comprising the expression vectors, such as the DNA encoding the antibody or antibody fragment.
[0056] Where it is desired to express a gene construct in yeast, an example of a suitable selection gene for use in yeast is the trpl gene present in the yeast plasmid YRp7. Stinchcomb et al. (1979) Nature, 282, 39; Kingsman et al. (1979) Gene 1, 141. The trpl gene provides a selection marker for a mutant strain of yeast lacking the ability to grow in tryptophan, for example, ATCC No. 44076 or PEP4-1. Jones (1977) Genetics 85, 12. The presence of the trpl lesion in the yeast host cell genome then provides an effective environment for detecting transformation by growth in the absence of tryptophan. Similarly, Leu2-deficient yeast strains (ATCC 20,622 or 38,626) are complemented by known plasmids bearing the Leu2 gene.
[0057] Transformed host cells are cultured by methods known in the art in a liquid medium containing assimilable sources of carbon, e.g. carbohydrates such as glucose or lactose, nitrogen, e.g. amino acids, peptides, proteins or their degradation products such as
- 17 -
NYOl 924757 vl peptones, ammonium salts or the like, and inorganic salts, e.g. sulfates, phosphates and/or carbonates of sodium, potassium, magnesium and calcium. The medium furthermore contains, for example, growth-promoting substances, such as trace elements, for example iron, zinc, manganese and the like.
[0058] Di-diabodies that bind to growth factor receptors are preferably capable of blocking activation of receptor tyrosine kinase (RTK) activity. Tyrosine kinase inhibition can be determined using well-known methods, for example, by measuring the autophosphorylation level of recombinant kinase receptor, and/or phosphorylation of natural or synthetic substrates. Thus, phosphorylation assays are useful in determining RTK antagonists of the present invention. Phosphorylation can be detected, for example, using an antibody specific for phosphotyrosine in an ELISA assay or on a western blot. Some assays for tyrosine kinase activity are described in Panek et al., J. Pharmacol. Exp. Thera. (1997) 283:1433-44 and Batley et al., Life Sci. (1998) 62:143-50.
[0059] In addition, methods for detection of protein expression can be utilized to determine RTK antagonists, wherein expression of the proteins being measured is mediated by the RTK. These methods include immunohistochemistry (IHC) for detection of protein expression, fluorescence in situ hybridization (FISH) for detection of gene amplification, competitive radioligand binding assays, solid matrix blotting techniques, such as Northern and Southern blots, reverse transcriptase polymerase chain reaction (RT-PCR) and ELISA. See, e.g., Grandis et al., Cancer, (1996) 78:1284-92; Shimizu et al., Japan J. Cancer Res., (1994) 85:567-71; Sauter et al. Am. J. Path., (1996) 148:1047-53; Collins, Glia, (1995) 15:289-96; Radinsky et al, Clin. Cancer Res, (1995) 1:19-31; Petrides et al. Cancer Res, (1990) 50:3934-39; Hoffmann et al. Anticancer Res, (1997) 17:4419-26; Wikstrand et al. Cancer Res, (1995) 55:3140-48.
[0060] The ability of a di-diabody to block ligand binding can be measured, for example, by an in vitro competitive assay. In such an assay, a ligand or the RTK (e.g., EGF for EGFR) is immobilized, and a binding assay is carried out to determine the effectiveness of the di-diabody to competitively inhibit binding of the RTK to the immobilized ligand.
[0061] In vivo assays can also be utilized to determine RTK antagonists. For example, receptor tyrosine kinase inhibition can be observed by mitogenic assays using cell lines stimulated with receptor ligand in the presence and absence of inhibitor. For example,
- 18 -
NYOl 924757 vl A431 cells (American Type Culture Collection (ATCC), Rockville, MD) stimulated with EGF can be used to assay EGFR inhibition. Another method involves testing for inhibition of growth of EGFR-expressing tumor cells, using for example, human tumor cells injected into a mouse. See U.S. Patent No. 6,365,157 (Rockwell et al.).
[0062] Preferred di-diabodies of the instant invention have dual specificity and are capable of binding to two different antigens simultaneously. The different antigens can be located on different cells or on the same cell. Cross linking of antigen can be shown in vitro, for example by providing a solid surface to which a first antigen has been bound, adding a bispecific antibodies specific for the first antigen and a second antigen for which the binding protein is also specific and detecting the presence of bound second antigen.
[0063] Preferred bispecific di-diabodies of the invention are capable of blocking the interaction between two receptors and their respective ligands. For example, a di-diabodies specific for KDR and FIt-I inhibits VEGF induced cell migration as well as PlGF induced cell migration. Combination of two receptor binding specificities in a bispecific di-diabodies, can be more efficacious in inhibiting cell migration than the individual parent antibodies (see, e.g., WO 2004/003211; Zhu).
[0064] Compared to antibodies that are monospecific, bispecific di-diabodies can be more potent inhibitors of cellular function. For example, VEGF-stimulated cellular functions such as, for example, proliferation of endothelial cells and VEGF- and PlGF-induced migration of human leukemia cells can be more efficiently inhibited by bispecific di- diabodies, even where affinity for one or both of the two target antigens is reduced. For example, a diabody specific (monovalent) for both KDR and FIt-I is more effective to inhibit VEGF or PlGF induced cell migration than a monospecific scFv directed at either of the target antigens (WO 2004/003211).
[0065] hi another example, a di-diabody having dual specificity for both EGFR and IGFR that is capable of binding to both receptors and blocking interaction with their specific ligands is use to neutralizing both EFG and IGF-stimulated receptor activation and downstream signal transduction. Stimulation of either EGFR or IGFR results in phosphorylation of common downstream signal transduction molecules, including Akt and p44/42, although to different extents, hi certain tumor cells, inhibition of EGFR function can be compensated by upregulation of other growth factor receptor signaling pathways, and
- 19 -
NYOl 924757 vl particularly by IGFR stimulation. In contrast to treatment with an antibody binds to one receptor, and does not completely block phosphorylation of either Akt or p44/42, incubation of tumor cells with an antibody that binds to both EGFR and IGFR blocks phosphorylation of both Akt and p44/42. Accordingly, inhibition of IGFR signaling results in inhibition of tumor growth and increased sensitivity of tumor cells to certain therapeutic agents.
[0066] Inhibition of phosphorylation of such common signal transduction cascade components is also observed with antibodies that bind to other RTKs, such as , for example, RON. Accordingly, the antigen-binding proteins are generally useful for treating neoplastic diseases characterized by cell growth or transformation resulting from activation of multiple signal transduction pathways.
[0067] The antigen-binding proteins of the invention are useful for treatment of a variety of proliferative disorders. For example, the present invention provides for treatment of tumors that express and are stimulated through more than one receptor tyrosine kinase. Stimulation through more that one receptor can result in uncontrolled growth that is insensitive to blockage of each receptor alone. Alternatively, stimulation of a second receptor can add to the activation observed in response to stimulation through a first receptor. Alternatively, the contributions from the individual receptors can be multiplicative. In each of the above instances, significantly improved inhibition of tumor growth is observed in the presence of an antigen-binding protein that blocks both of the receptors.
[0068] The antigen-binding proteins of the invention are useful for treating diseases in which receptor stimulation is through an EGFR paracrine and/or autocrine loop. For example, EGFR expressing tumors are characteristically sensitive to EGF present in their environment, and can further be stimulated by tumor produced EGF or TGF-α. While not intending to be bound to any particular mechanism, the diseases and conditions that may be treated or prevented by the present methods include, for example, those in which tumor growth is stimulated. The method is therefore effective for treating a solid tumor that is not vascularized, or is not yet substantially vascularized.
[0069] Certain antigen-binding proteins of the invention are useful for inhibiting angiogenesis associated with a hyperproliferative disease. For example, by blocking tumor associated angiogenesis, tumor growth may be inhibited. In one embodiment, the antigen- binding protein binds to a tumor associated RTK and inhibits production of angiogenic
- 20 -
NYOl 924757 vl ligands (i.e., VEGF) by the tumor, and also binds to a VEGF receptor associated with cells of the vasculature to inhibit proliferation of such cells. In a different embodiment, the antigen- binding protein binds to multiple VEGF receptors, such that VEGF or other ligand of VEGFR (e.g., PlGF) ligand is blocked from binding to more than one type of VEGF receptor.
[0070] Tumors that may be treated include primary tumors and metastatic tumors, as well as refractory tumors. Refractory tumors include tumors that fail to respond or are resistant to treatment with chemotherapeutic agents alone, antibodies alone, radiation alone or combinations thereof. Refractory tumors also encompass tumors that appear to be inhibited by treatment with such agents, but recur up to five years, sometimes up to ten years or longer after treatment is discontinued. The tumors may express EGFR or other RTK at normal levels or they may overexpress the RTK at levels, for example, that are at least 10, 100, or 1000 times normal levels.
[0071] Examples of tumors that express EGFR and are stimulated by a ligand of EGFR include carcinomas, gliomas, sarcomas, adenocarcinomas, adenosarcomas, and adenomas. Such tumors can occur in virtually all parts of the body, including, for example, breast, heart, lung, small intestine, colon, spleen, kidney, bladder, head and neck, ovary, prostate, brain, pancreas, skin, bone, bone marrow, blood, thymus, uterus, testicles, cervix or liver. Some tumors observed to overexpress EGFR that may be treated according to the present invention include, but are not limited to, colorectal and head and neck tumors, especially squamous cell carcinoma of the head and neck, brain tumors such as glioblastomas, and tumors of the lung, breast, pancreas, esophagus, bladder, kidney, ovary, cervix, and prostate. Non-limiting examples of tumors observed to have constitutively active (i.e., unregulated) receptor tyrosine kinase activity include gliomas, non-small-cell lung carcinomas, ovarian carcinomas and prostate carcinomas. Other examples of tumors include Kaposi's sarcoma, CNS neoplasms, neuroblastomas, capillary hemangioblastomas, meningiomas and cerebral metastases, melanoma, gastrointestinal and renal carcinomas and sarcomas, rhabdomyosarcoma, glioblastoma, preferably glioblastoma multiforme, and leiomyosarcoma. Overexpression of other RTKs can produce similar growth defects. For example, most metastatic bone cancers arise from primary tumors of prostate, breast, or lung. Prostate tumors initially may be hormone dependent, but loss of such dependence coincides with IGFR mediated stimulation of cells that migrate to bone.
- 21 - .
NYOl 924757 vl [0072] The antigen-binding proteins are also useful for treating hyperproliferative disesases other than tumors comprising administering to the mammal an effective amount of the antibody of the present invention. As disclosed herein, "hyperproliferative disease" is defined as a condition caused by excessive growth of non-cancer cells that express a member of the EGFR family or other tyrosine kinase receptors. The excess cells generated by a hyperproliferative disease express the RTK at normal levels or they may overexpress the RTK.
[0073] Examples of hyperproliferative disease include psoriasis, actinic keratoses, and seborrheic keratoses, warts, keloid scars, and eczema. Also included are hyperproliferative diseases caused by virus infections, such as papilloma virus infection. For example, psoriasis comes in many different variations and degrees of severity. Different types of psoriasis display characteristics such as pus-like blisters (pustular psoriasis), severe sloughing of the skin (erythrodeimic psoriasis), drop-like dots (guttae psoriasis) and smooth inflamed lesions (inverse psoriasis). The treatment of all types of psoriasis (e. g., psoriasis vulgaris, psoriasis pustulosa, psoriasis erythrodermica, psoriasis arthropathica, parapsoriasis, palmoplanar pustulosis) is contemplated by the invention.
[0074] According to the invention, di-diabodies can be chemically or biosynthetically conjugated to other agents such as antineoplastic or anti-angiogenic agents for treatment of disease. Anti-tumor agents linked to an antibody include any agents which destroy or damage a tumor to which the antibody has bound or in the environment of the cell to which the antibody has bound. For example, an anti-tumor agent is a toxic agent such as a chemotherapeutic agent or a radioisotope. The chemotherapeutic agents are conjugated to the di-diabody using conventional methods {See, e.g., Hermentin and Seiler (1988) Behring Inst. Mitt. 82, 197-215), including by peptide and non-peptide linkers.
[0075] Di-diabodies can also be linked to detectable signal-producing agents useful in vivo and in vitro for diagnostic purposes. The signal producing agent produces a measurable signal which is detectible by external means, usually the measurement of electromagnetic radiation. For the most part, the signal producing agent is an enzyme or chromophore, or emits light by fluorescence, phosphorescence or chemiluminescence. Chromophores include dyes which absorb light in the ultraviolet or visible region, and can be substrates or degradation products of enzyme catalyzed reactions.
- 22 -
NYOl 924757 vl [0076] The invention further contemplates the use of di-diabodies with treatment or diagnostic agents incorporated into secondary reagents. For example, one member of a binding pair is linked to the di-diabody of the invention. Anti-neoplastic agents, for example, are conjugated to second members of such pairs and are thereby directed to the site where the di-diabody is bound, hi a preferred embodiment, biotin is conjugated to a di-diabody, and thereby provides a target for an anti-neoplastic agent or other moiety which is conjugated to avidin or streptavidin. Alternatively, biotin or another such moiety is linked to a di-diabody of the invention and used as a reporter, for example in a diagnostic system where a detectable signal-producing agent is conjugated to avidin or streptavidin.
[0077] Di-diabodies can be administered in combination with one or more suitable adjuvants, such as, for example, cytokines (IL-10 and IL- 13, for example) or other immune stimulators, such as, but not limited to, chemokine, tumor-associated antigens, and peptides. It should be appreciated, however, that administration of only a diabody is sufficient to prevent, inhibit, or reduce the progression of the tumor in a therapeutically effective manner.
[0078] hi certain embodiments, it can be desirable to administer an antigen-binding protein of the invention that binds to an RTK and blocks ligand binding in combination with another antigen-binding protein that binds to ligand. Ligand binding antibodies are well known in the art, and include, e.g., anti-VEGF (Avastin®; bevacizumab).
[0079] The di-diabodies of the invention are also to be used in combined treatment methods by administration with an anti-neoplastic agent such as a chemotherapeutic agent or a radioisotope. Suitable chemotherapeutic agents are known to those skilled in the art and include irinotecan (CPT-Il), anthracyclines (e.g. daunomycin and doxorubicin), methotrexate, vindesine, neocarzinostatin, cisplatin, chlorambucil, cytosine arabinoside, 5-fluorouridine, melphalan, ricin and calicheamicin. A di-diabody and an anti-angiogenic or anti-neoplastic agent are admininstered to a patient in amounts effective to inhibit angiogenesis and/or reduce tumor growth. The di-diabodies are also to be administered in combination with other treatment regimes, for examplej with treatments such as radiation therapy. For examples of combination therapies, see, e.g., U.S. Patent No. 6,217,866 (Schlessinger et al.) (Anti-EGFR antibodies in combination with anti-neoplastic agents); WO 99/60023 (Waksal et al.) (Anti-EGFR antibodies in combination with radiation).
- 23 -
NYOl 924757 vl [0080] Any suitable anti-neoplastic agent can be used, such as a chemotherapeutic agent, radiation or combinations thereof. The anti-neoplastic agents known in the art or being evaluated can be grouped in to classes based on their target or mode of action. For example, alkylating agents include, but are not limited to, cisplatin, cyclophosphamide, melphalan, and dacarbazine. Examples of anti-metabolites include, but not limited to, doxorubicin, daunorubicin, and paclitaxel, gemcitabine, and topoisomerase inhibitors irinotecan (CPT-Il), aminocamptothecin, camptothecin, DX-8951f, and topotecan (topoisomerase I) and etoposide (VP-16) and teniposide (VM-26) (topoisomerase II). For radiation, the source can be either external (external beam radiation therapy — EBRT) or internal (brachytherapy - BT) to the patient being treated. Such classifications can be useful for choosing an antineoplastic agent to use. For example, it has been observed that antibodies that bind IGFR may be particularly effective when administered with a topoisomerase inhibitor.
[0081] The dose of anti-neoplastic agent administered depends on numerous factors, including, for example, the type of agent, the type and severity tumor being treated and the route of administration of the agent. It should be emphasized, however, that the present invention is not limited to any particular dose.
[0082] hi a combination therapy, the di-diabody is administered before, during, or after commencing therapy with another agent, as well as any combination thereof, i.e., before and during, before and after, during and after, or before, during and after commencing the anti-neoplastic agent therapy. For example, for treatment of a tumor or neoplastic disease, the diabody can be administered between 1 and 30 days, preferably 3 and 20 days, more preferably between 5 and 12 days before commencing radiation therapy. In a preferred embodiment of the invention, chemotherapy is administered concurrently with, prior to, or subsequent to antibody therapy.
[0083] hi the present invention, any suitable method or route can be used to administer di-diabodies of the invention, and optionally, to co-administer anti-neoplastic agents, receptor antagonists, or other pharmaceutical composition. For example, anti¬ neoplastic agent regimens utilized according to the invention include any regimen believed to be optimally suitable for the treatment of a patient's neoplastic condition. Different malignancies can require use of specific anti-tumor diabodies and specific anti-neoplastic agents, which will be determined on a patient to patient basis. Routes of administration
- 24 -
NYOl 924757 vl include, for example, oral, intravenous, intraperitoneal, subcutaneous, or intramuscular administration. The dose of anti-neoplastic agent administered depends on numerous factors, including, for example, the type of neoplastic agent, the type and severity tumor being treated and the route of administration of the antineoplastic agent. It should be emphasized, however, that the present invention is not limited to any particular method or route of administration.
[0084] It is understood that di-diabodies of the invention, where used in a mammal for the purpose of prophylaxis or treatment, will be administered in the form of a composition additionally comprising a pharmaceutically acceptable carrier. Suitable pharmaceutically acceptable carriers include, for example, one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof. Pharmaceutically acceptable carriers can further comprise minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the binding proteins. The compositions of the injection can, as is well known in the art, be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the mammal.
[0085] The present invention also includes kits for inhibiting tumor growth and/or angiogenesis, or treating other disesase, comprising a therapeutically effective amount of a human di-diabody. The kits can further contain any suitable antagonist of, for example, another growth factor receptor involved in tumorigenesis or angiogenesis (e.g., EGFR, VEGFR-l/Flt-1, VEGFR-2/Flk-l/KDR, IGFR, PDGFR, NGFR, FGFR, etc, as described above). Alternatively, or in addition, the kits of the present invention can further comprise an anti-neoplastic agent. Examples of suitable anti-neoplastic agents in the context of the present invention have been described herein. The kits of the present invention can further comprise an adjuvant; examples have also been described above.
[0086] Also included within the scope of the present invention is use of the present di-diabodies in vivo and in vitro for investigative or diagnostic methods, which are well known in the art. The diagnostic methods include kits which contain di-diabodies of the present invention.
[0087] Accordingly, the present receptor binding di-diabodies thus can be used in vivo and in vitro for investigative, diagnostic, prophylactic, or treatment methods, which are
- 25 -
NYOl 924757 vl well known in the art. Of course, it is to be understood and expected that variations in the principles of invention herein disclosed can be made by one skilled in the art and it is intended that such modifications are to be included within the scope of the present invention.
EXAMPLES
[0088] The following examples further illustrate the invention, but should not be construed to limit the scope of the invention in any way. Detailed descriptions of conventional methods, such as those employed in the construction of vectors and plasmids, the insertion of genes encoding polypeptides into such vectors and plasmids, the introduction of plasmids into host cells, and the expression and determination thereof of genes and gene products can be obtained from numerous publications, including Sambrook, J et al., (1989) Molecular Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor Laboratory Press; and Coligan, J. et al. (1994) Current Protocols in Immunology, Wiley & Sons, Incorporated All references mentioned herein are incorporated in their entirety.
Cell lines and proteins
[0089] Primary-cultured human umbilical vein endothelial cells (HUVEC) were maintained in EBM-2 medium (Clonetics, Walkersville, MD) at 37°C, 5% CO2. The human leukemia cell lines, HEL and U937, were maintained in RPMI containing 10% of fetal calf serum. The soluble receptor fusion proteins, the extracellular domain of kinase inserting domain-containing receptor (KDR)-alkaline phosphatase (AP), and the extracellular domain of fins-like tyrosine kinase receptor (FIt-I)-AP, were expressed in stably transfected NIH 3T3 cells and purified from cell culture supernatant by affinity chromatography using immobilized monoclonal antibody to AP as described (Lu et al. (2000) J. Biol. Chem. 275:14321-30). Vascular endothelial growth factor (VEGF165) protein was expressed in baculovirus and purified following the procedures described (Lu et al. (2000)). The single chain antibody directed against KDR, scFv pi Cl 1, was isolated from a phage display library constructed from the splenocytes of a mouse immunized with KDR-AP fusion protein (Zhu et al. (1998) Cancer Res. 58:3209-14). Hybridoma cell line producing the anti-Flt-1 antibody, FBK612 (IgGl, K), was established at ImClone Systems Incorporated (New York, NY) from mice immunized with FIt-I -AP fusion protein. The single chain version of FBK612, scFv
- 26 -
NYOl 924757 vl 612, was constructed from the antibody variable genes isolated from the hybridoma cells as previously reported (Lu et al. (2001)).
Construction and expression of a bispecific diabody
[0090] Two antibodies directed against the two different VEGF receptors, KDR and FIt-I, were used for the construction of a bispecific diabodies. PlCIl binds specifically to KDR and blocks KDR/VEGF interaction, whereas 612 binds to FIt-I and blocks FIt-I from binding to VEGF. The gene encoding a previously created divalent bispecific diabody, DAB plCl 1/612 (WO 2004/003211), was used as the template to construct the tetravalent di- diabody. The gene encoding one of the "cross-over" scFv chains in DAB plCl 1/612 (Lu et al. (2000)) was first fused to a gene encoding the CR3 domain of an IgG via a hinge region and peptide linker (EPKSCDKTHTCPPCGGGGSGGGGS), followed by subcloning of the resulted fusion polypeptide into the expression vector with its partner, the other "cross-over" scFv chain, for the expression of the soluble di-diabody (Fig. 1). A 13-amino-acid E tag, was added to the C-terminus of the smaller polypeptide chain for purification and assay purposes (Lu et al., 2000). All sequences encoding the polypeptides were verified by DNA sequencing.
Expression and purification of the di-diabody
[0091] The diabodies were secreted from E. coli strain HB2151 containing the expression plasmid grown at 30°C in a shaker flask following the procedure previously described (Lu et al., 2000; Zhu et al., 1998). A periplasmic extract of the cells was prepared by resuspending the cell pellet in 25 mM Tris (pH 7.5) containing 20% (w/v) sucrose, 200 mM NaCl, 1 mM EDTA and 0.1 mM PMSF5 followed by incubation at 4°C with gentle shaking for 1 h. After centrifugation at 12,000 rpm (17211 x g) for 20 min, the diabodies were purified from the supernatant by an anti-E tag affinity chromatography using the RPAS Purification Module (Amersham Pharmacia Biotech). To examine the purity of the diabody preparations, the purified proteins were electrophoresed in a 4-20% gradient polyacrylamide gel (Novex, San Diego, CA) and visualized by staining with Colloidal Blue Stain kit (No vex).
[0092] The di-diabody was also expressed in mammalian COS cells. Nucleic acids encoding both of the polypeptides, each fused with an identical immunoglobulin leader
- 27 -
NYOl 924757 vl peptide sequence on its N-terminus (Zhu, et al., 1998), were cloned into a single expression vector (the glutamine synthetase expression system from Lonza Biologies Inc.). Sub- confluent COS cells were transfected with the expression vector using Lipofectamine following the instructions of the manufacturer (Invitrogen). The cells were cultured in serum-free medium, and supernatant was collected at 48 to 72 h after the transfection. After centrifugation to remove the cell debris, the di-diabody was purified from the supernatant using the RPAS Purification Module as described above.
[0093] The yield of the purified di-diabody ranged from 150 to 250 μg per liter of overnight bacteria culture in shaker flask, and approximately 800 to 1250 μg per liter of supernatant 48 to 72 h after transfection of the mammalian culture. The composition and purity of the purified di-diabody preparations, both from E. coli expression [di-diabody (B)] and from mammalian expression [di-diabody (M)], was analyzed using SDS-PAGE (Fig. 2). The components of the "cross-over" scFv in a bispecific diabody were also resolved by electrophoresis. Under non-reducing conditions, the divalent bispecific diabody, DAB pi Cl 1/612, yielded two bands (Fig. 2 A, lane 3), representing each one of the two "cross¬ over" scFv polypeptide chains (theoretical m.w., 26693.8 and 25179.6 daltons for the upper and the lower band, respectively). The di-diabody preparation also gave two major bands (Fig. 2A, lane 4), the lower band representing the "cross-over" scFv chain (the same band as in DAB plCl 1/612), whereas the upper band represents the "cross-over" scFv-hinge-CH3 fusion in dimer form (theoretical m.w., 79175.3 daltons). Under the reduced conditions (Fig. 2B), the di-diabody yielded two major bands: the lower band represents the original "cross¬ over" scFv chain, and the top band corresponds to the reduced scFv-hinge-CH3 fusion in monomer form. As expected, monospecific bivalent diabodies DAB 612 yielded one major band under both reducing and non-reducing conditions (lane 2, theoretical m.w. 26916.9 daltons). DAB plCl 1 gave two bands (lane 1): the upper band corresponds to the single polypeptide, VL-G4S-VH, with intact E-tag (theoretical m.w. 26542.4 daltons); the lower band represents the same polypeptide with E-tag degradation.
Determination of antibody molecular mass in solution
[0094] The molecular mass of the purified di-diabody was determined by a size exclusion chromatography / multi-angle light scattering (SEC-MALS) analysis following a procedure previously described (Folta-Stogniew and Williams, 1999). Briefly, 25-50 μg of
- 28 -
NYOl 924757 vl each purified antibody preparation in 100 μl PBS (pH 7.0) was applied to a Bio-Sep 3000 column (Phenomenex, Torrance, CA) linked to an HPLC system with UV and refractive index detectors (Agilent 1100, Agilent, Palo Alto, CA), and followed by a Mini-Dawn LS (Wyatt Technology, Santa Barbara, CA). The column was equilibrated in PBS (pH 7.0) and run at a flow rate of 0.5 ml/min. The weight average molecular masses (Mw) were calculated throughout the entire eluting protein peak at 8.3 μl intervals using ASTRA software (Wyatt Technology). A Zimm fit was used for all samples and was performed as previously described (Folta-Stogniew et al. (1999) J. Biomol. Tech. 10:51-63).
[0095] The di-diabody yielded three distinct peaks on SEC. ASTRA-computed Zimm analysis indicated a monodispersed peak (75% of the population) with a Mw value of 132 kD, a monodispersed peak (20% of the population) with a Mw value of 270 kD and a polydispersed peak (5% of the population) with a Mw value of 470 kD. Based on the calculated theoretical molecular masses, the 132 kD peak represents the correctly folded monomelic di-diabody, and the 270 kD peak represents the dimeric form of the di-diabody, and the polydispersed 470 kD peak is a heterogeneous population with respect to oligomeric state. As controls, an intact IgG antibody with a theoretical m.w. of 150 kD yielded a single peak with a estimated Mw value of 150 kD, and a diabody with a theoretical m.w. of 60 kD yielded a single peak with a estimated Mw value of 62 kD, under identical conditions. These results suggest that the majority, i.e., 95% (75% monomeric plus 20% dimeric di-diabody population) of the purified di-diabody preparation is correctly folded tetravalent molecules.
Dual specificity and antigen-bindins kinetics of the bispecific diabodies
[0096] Two ELISA assays were carried out to determine the dual antigen binding capability of the bispecific di-diabody. First, a cross-linking assay was used to investigate whether the di-diabody is capable of binding both of its target antigens simultaneously. The monospecific diabodies, the bispecific diabody and di-diabody were first incubated in the presence of 1 mM of dithiothreitol (DTT) in a 96-well Maxi-sorp microtiter plate (Nunc, Roskilde, Denmark) precoated with FIt-I-Fc fusion protein (1 μg/ml x 100 μl per well overnight at 4°C) at RT for 1 h. The plate was washed three times with PBS containing 0.1 % Tween (PBST), followed by incubation with KDR-AP fusion protein (100 ng/well) at RT for additional 1 h. The plate-bound KDR-AP was then quantified by the addition of AP
- 29 -
NYOl 924757 vl substrate, p-nitrophenyl phosphate (Sigma, St. Louis, MO), followed by reading of the absorbance at 405nm (Zhu et al., 1998).
[0097] Under these conditions, the di-diabody was reduced into two half molecules, each consisting of a single diabody-Cπ3 fusion (Fig. 1). Both the reduced divalent bispecific diabody and di-diabody, but not the monospecific diabody (DAB pi Cl 1 and DAB 612), were able to cross-link the two target antigens (Fig. 3, top panel). This observation indicates that the di-diabody, a dimer of two diabody-Cκ3 fusions, possesses four antigen-binding sites (two on each CH3 arm).
[0098] In the second assay, the direct binding assay, various amounts of the diabodies were added to KDR or FIt-I -coated 96-well plates and incubated at RT for 1 h, after which the plates were washed 3 times with PBS. The plates were then incubated at RT for 1 h with 100 μl of an anti-E tag antibody-HRP conjugate (Amersham Pharmacia Biotech). The plates were washed, peroxidase substrate added, and the absorbance at 450nm read following the procedure described previously (Lu et al., 1999; 2000).
[0099] All the bispecific diabodies bind to both KDR and FIt-I in a dose-dependent manner (Fig. 3). In each case, the di-diabody expressed from mammalian cells is the most efficient binder, with the binding efficiency following the order of: di-diabody (M) (mammalian produced) > di-diabody (B) {E.coli produced) > DAB plCl 1 > DAB plCl 1/612 (for KDR binding), or di-diabody (M) > DAB 612 > di-diabody (B) > DAB plCl 1/612 (for FIt-I binding). As expected, the KDR-specifϊc DAB pi Cl 1 did not bind to FIt-I, nor did the FIt-I -specific DAB 612 bind to KDR.
[0100] The binding kinetics of the diabodies to KDR and FIt-I were measured using BIAcore biosensor (BIAcore 3000, Biacore, Inc., Uppsala, Sweden). KDR-AP or Flt-l-AP fusion protein was immobilized onto a sensor chip (CM5 Research Grade, Biacore, Inc.) and soluble antibodies were injected at concentrations ranging from 1.5 nM to 200 nM in PBS, at a flow rate of 10 μl/min. Sensorgrams were obtained at each concentration and were evaluated using a program, BIA Evaluation 2.0, to determine the rate constants kon and koff. The affinity constant (KJ was calculated from the ratio of rate constants kojf/kon.
[0101] The KDR binding characteristics (kinetics and affinity) of divalent DAB plCl 1/612 were similar to those of the scFv plCll. (Table 1) Both tetravalent di-diabodies demonstrated a modestly improved dissociation rate (koff) over the monovalent scFv pi Cl 1 -
- 30 -
NYOl 924757 vl with an off-rate approaching that of the bivalent (monospecific) DAB p 1 C 11. Converting scFv 612 into a bivalent diabody resulted in a dramatic improvement (~10-fold) in binding affinity to FIt-I. While both di-diabody preparations showed a similar overall binding affinity to the divalent DAB pi Cl 1/612 for FIt-I, significant differences were observed in the individual binding kinetic constants between the two preparations. The E. coli-deήved di- diabody (B) demonstrated kinetic constants that are very similar to those of the scFv 612 and DAB pi Cl 1/612, whereas the mammalian cell-derived material showed a much slower association rate ikon), along with a significantly improved dissociation rate (koff) that is approaching that of the bivalent DAB 612.
- 31 -
NYOl 924757 vl Table 1 Binding kinetics of the antibody fragments as determined by BIAcore analysis
KDRbinding FIt-I binding
Antibody Ar0n (IO 4M-1S "1) ^(10 "V1) Krf (nM) Ar0n (IO 4M-1S -1) Aro#(10 "4S -1) Krf (nM)
scFvplCll 7.4±0.88* 1.2±0.36 1.7±0.66 NDB** NDB NDB
DABplCll 6.2 ±0.95 0.64±0.24 1.0±0.26 NDB NDB NDB
DABpICl 1/612 6.1 ±0.36 1.1±0.24 1.7±0.32 13.0±3.9 22.4±5.2 18.0±7.8
Di-diabody (B) 6.7 ±2.2 0.78±0.15 1.3±0.52 18.3±7.0 16.3±1.1 10.0+2.9
Di-diabody (M) 4.7 ±0.51 0.82±0.26 1.8±0.68 2.4±0.99 2.7±0.84 16.0±5.0 scFv612 NDB NBD NDB 20.0±4.7 17.0±4.3 8.8±1.9
DAB612 NDB NDB NDB 42.1±10.7 3.3±0.79 0.8±0.17
*A11 numbers are determined by BIAcore analysis and represent the mean ± SE of at least four separate determinations. Kd values are calculated as the ratios of &off / Ar0n. **NDB, no detectable binding.
Dual blocking activity of the bispeciβc diabody
[0102] The assay was carried out following a previously described protocol (Lu et al., 1999; 2000). Various amounts of the diabodies were mixed with a fixed amount of KDR-AP (100 ng) or FIt-I -AP fusion protein (100 ng) and incubated at RT for 1 h. The mixture were then transferred to 96-well microtiter plates precoated with VEGFl 65 (200 ng/well) and incubated at RT for an additional 2 h, after which the plates were washed 5 times with PBS. The substrate for AP was added, followed by reading of the absorbance at 405nm to quantify the plate-bound KDR-AP or FIt-I -AP molecules. The IC50, i.e., the antibody concentration required for 50% inhibition of KDR or FIt-I binding to VEGF, was then calculated.
[0103] Fig. 4 shows that the diabodies block soluble KDR and FIt-I from binding to immobilized VEGF, in a dose-dependent manner. Following the same pattern seen in the binding assay: the tetravalent di-diabody preparations were more potent blockers to both KDR/VEGF and Flt-1/VEGF interaction than the divalent diabody, DAB plCl 1/612. The IC50 values for KDR blocking were approximately 0.4 nM for both di-diabody preparations, compared to that of 1.2 nM for DAB plCl 1/612 and 0.8 nM for the monospecific bivalent DAB plCl 1. The IC50 for FIt-I blocking was approximately 1 nM for both di-diabody preparations, 8 nM for DAB plCl 1/612 and 0.2 nM for the monospecific bivalent DAB 612. As expected, DAB plCl 1 had no effects on Flt-1/VEGF interaction, whereas DAB 612 had no effects on KDR/VEGF interaction.
Inhibition of VEGF-induced migration of leukemia cells
[0104] The di-diabodies were tested for their activity in inhibiting VEGF-induced cell migration. Two human leukemia cell lines, HEL that expresses both KDR and FIt-I, and U937 that only expresses FIt-I, were used. Leukemia cells, HEL and U937, were washed three times with serum-free plain RPMI 1640 medium and suspended in the medium at 1 x 106/ml. Aliquots of 100 μl cell suspension were added to 8-μm-pore transwell inserts (Costar®, Corning Incorporated, Corning, NY) and incubated with the antibodies at various concentrations, 100 nM, 25 nM and 6.25 nM, for 30 min at 37°C. The inserts were then placed into the wells of 24-well plates containing 0.5 ml of serum-free RPMI 1640 with or without VEGF165. The migration was carried out at 37°C, 5% CO2 for 4 h. Migrated cells were collected from the lower compartments and counted under a light microscope.
NYQl 924151 vl 33 [0105] Both DAB plCl 1 and DAB 612 effectively inhibited VEGF-induced migration of HEL cells, whereas migration of U937 cells was only inhibited by DAB 612, but not by DAB pi Cl 1 (Fig. 5). The bispecific diabodies, including both the bivalent diabody and the tetravalent di-diabodies, were equal potent inhibitors to VEGF-induced migration of both HEL and U937 cells at all three different antibody concentrations tested. As expected, a control antibody directed against insulin-like growth factor receptor, A12, did not show significant inhibition in VEGF-induced cell migration in these assays.
Stability of di-diabodies
[0106] Various diabody preparations were incubated in PBS or mouse serum at 37°C for up to 72 h. Aliquots of samples were removed at 24 and 72 h and assayed for efficiency for binding to both KDR and FIt-I using the ELISA assay described above.
[0107] The stability of the di-diabodies was tested by examination of their binding activity to both KDR and FIt-I. As shown in Fig. 6, the divalent bispecific diabody, DAB plCl 1/612, and the monospecific diabodies, DAB plCl 1 and DAB 612, retained full antigen binding activity to their respective targets after incubation at 370C in both PBS and mouse serum. At each time point tested the mammalian cell-derived di-diabody (M) preparation retained better antigen-binding activity compared to the E.cø/z-derived preparation. At 24 h, di-diabody (M) showed 94% of the original KDR-binding activity, compared to that of 83% for di-diabody (B). At 72 h, di-diabody (M) retained 77% and 92% binding activities to KDR and FIt-I, respectively, compared to those of 63% and 86% for di-diabody (B).
Construction and production of anti-EGFR/anti-IGR-IR di-diabody
[0108] The variable regions of a fully human anti-ΕGFR antibody (IMC-11F8) and a fully human anti-IGF-IR antibody (IMC-Al 2) were used to construct a bispecific di-diabody. Bispecific diabodies were constructed as above, followed by fusion of one of the diabody cross-over scFv chains to the Fc domain of an IgG (see Fig. 7A and 7B for details). Co- expression in mammalian cells the Fc fusion along with the other cross-over scFv resulted in an IgG-like tetravalent molecule with two binding specificities (Fig. 7B). The di-diabody was produced by stably transfected NSO cells in serum-free conditions and purified from the cell culture supernatant via a Protein A affinity column. Electrophoresis analysis of the purified di-diabody under non-reducing conditions yielded two major protein bands with
- 34 -
NYQl 924757 vl expected mobility, the Fc fusion (the top band) in dimer form (mw -100 kD) and the cross¬ over scFv (the lower band) in monomer form (mw ~25 kD) (Fig. 7C, lane 2). IMC-11F8, an IgG, yielded a single protein band of ~ 150 kD (Fig. 7C, lane 1). Under reducing conditions, the di-diabody also gave rise to two bands: the top band at ~ 50 kD (representing the Fc fusion in monomer form), and the lower band at ~ 25 kD (the cross-over scFv) (Fig. 7D, lane 2). As controls, EVIC-11F8 gave two major bands: the IgG heavy chain (50 kD) and the IgG light chain (25 kD) (Fig. 7D, lane 1).
The di-diabodv binds to both EGFR and IGFR
[0109] Two assays were used to demonstrate the di-diabody was capable of binding to both EGFR and IGFR. A cross-linking assay was used to determine whether the di- diabody could bind to both targets simultaneously. Various antibodies were first incubated with a biotin-labeled IGFR in solution and then transferred to a 96-well plate coated with EGFR, followed by incubation with streptoavidin-FJRP to measure the plate-bound biotin activity, i.e., the amount of IGFR that was cross-linked to the immobilized EGFR by the antibody. As shown in Fig. 8 A, only the di-diabody, but not the monospecific BVIC-11F8 or EVIC-A12, was able to cross-link IGFR in solution with the immobilized EGFR.
[0110] A direct binding assay was used to compare antigen-binding efficiency of the di-diabody with its monospecific counterparts. IMC-Al 2 and BVIC-11F8 bound only to their respective targets, whereas the di-diabody reacted to both immobilized EGFR and IGFR, with moderately lower efficiencies as compared to its monospecific counterparts (Fig. 2B and 2C). The ED50 values {i.e., the antibody concentrations that yield 50% of maximum binding) to EGFR were 0.05 nM for BVIC-11F8 and 0.1 - 0.2 nM for the Di-diabody, and to IGFR were 0.1 nM for BVIC-A12 and 0.25 - 0.5 nM for the Di-diabody.
Inhibition tumor cell proliferation in vitro by the di-diabodv
[0111] The efficacy of the di-diabody in inhibiting tumor cell proliferation in vitro was established using a well-characterized tumor cell line, DiFi, which is known to depend on EGFR for survival and growth. The anti-EGFR antibody (BvIC-11F8) significantly inhibited the proliferation of DiFi cells, whereas the anti-IGFR antibody (DVIC-A12), and the control antibody (EvIC-1121; anti-KDR) had little effect (Fig. 9). Proliferation of the DiFi cells was also inhibited by the di-diabody, although at higher concentrations than BVIC-11F8.
- 35 -
NYOl 9247S7 vl The IC50 values were approximately 1 nM for IMC-11F8 (alone), InM for the coadministered combination of IMC-11F8 and IMC-Al 2, and 25 nM for the di-diabody.
Blockage of EGF and IGF signal transduction pathways
[0112] The di-diabody was assayed for efficacy in blocking EGF and IGF-stimulated receptor phosphorylation and downstream signal transduction. Incubation of MCF-7 cells with EGF or IGF results in significant phosphorylation of the respective receptor; incubation with a combination of EGF and IGF yields activation of both EGFR and IGFR (Fig. 1OA, lanes 2, 3, and 4). As expected, when the tumor cells were stimulated with both EGF and IGF, treatment with IMC-A12 or IMC-11F8 inhibited phosphorylation of only its corresponding receptor (Fig. 1OA, lanes 6 and 7). hi contrast, the di-diabody, like the mixture of both EVIC-11F8 and EVΪC-12, significantly blocked activation of both receptors (Fig. 1OA, lanes 5 and 8). The control antibody, IMC-1121, showed no effect on the phosphorylation of EGFR and IGFR (Fig. 1OA, lane 9).
[0113] The effect of IMC-11F8, EVIC-A12 and the di-diabody on Akt and ρ44/p42 MAP kinases (the two major downstream signal transduction molecules associated with both EGFR and IGFR) were also studied in MCF-7 cells (Fig. 10B). Stimulation with IGF results in significant phosphorylation of Akt (Fig. 1OB, lane 2) whereas EGF causes strong phosphorylation of p44/p42 MAPK (Fig. 1OB, lane 3). As expected, combination of IGF and EGF leads to activation of both Akt and ρ44/p42 MAPK (Fig. 1OB, lane 4). In the presence of both EGF and IGF, IMC-11F8 significantly inhibited the activation of MAPK but only moderately reduced the activation of Akt (Fig. 1OB, lane 7), whereas EVIC-A12 strongly reduced Akt phosphorylation but was less effective in p44/p42 MAPK activation (Fig. 1OB, lane 6). The di-diabody, effectively blocked phosphorylation of both Akt and p44/p42 MAPK induced by EGF and IGF (Fig. 1OB, lane 5) as did the combination of BVIC-A12 and IMC-11F8 (Fig. 10, lane 8).
/GFJ? internalization and degradation
[0114] IMC-A12 has been shown to induce rapid and efficient internalization and degradation of cell surface expressed IGFR (Burtrum et al., 2003, Cancer Res. 63:8912). The di-diabody retained the receptor modulation activity of EVIC-A12 on tumor cells, triggering significant IGFR internalization and degradation in MCF-7 cells after incubation at 37°C for
- 36 -
NYOl 924757 vl 4 h (Fig. 1 IA and B). Incubation with the di-diabody also led to significant degradation of IGFR in BxPC3 cells (Fig. HC). This IGFR modulation effect of the di-diabody is both dose-dependent - with the maximum effect, similar to that of IMC-A12, achieved at an antibody concentration of 100 nM (Fig. 1 IA), and time-dependent - greater than 90% of the IGFR were degraded in BxPC3 cells after 4 h incubation at 370C (Fig. HC). IGFR modulation was not observed upon incubation with ligand (IGF), IMC-11F8, or IMC-1121 (anti-VEGFR-2).
Mediation of ADCC on tumor cells
[0115] In addition to blocking growth signals by interfering with growth factor/receptor interaction and down-regulating receptor surface expression, antitumor IgG antibodies can also cause direct tumor cell killing via mediating effective ADCC. The ability of the di-diabody to mediate tumor-cell killing in the presence of human efffector cells was examined. As shown in Fig. 12, BVIC-11F8 demonstrated lytic activity against both A431 and BxPC3 cells but was ineffective against MCF-7 cells. EVIC-Al 2 was effective only towards MCF-7 cells but failed to kill A431 and BxPC3 cells. The di-diabody, mediated killing of all the three tumor lines.
Inhibition of growth of human tumor xenografts in nude mice
[0116] Antitumor activity of the di-diabody was examined and compared to the monospecific parent antibodies. Nude mice bearing established xenografts of- 200 to 300 mm3 were treated with the various antibodies twice a week by intraperitoneal injection, hi the first model of BxPC3 pancreatic tumor xenografts (Fig. 13A), both IMC- 11F8 and BVIC- Al 2 alone (at 40 mg/kg) yielded significant (77% and 58%, respectively) tumor growth inhibition at 6 weeks after initiation of treatment. The di-diabody (at 40 mg/kg) demonstrated a similar antitumor activity (52% tumor growth inhibition) to the individual parent antibodies when given at the same dose (40 mg/kg). The combination of both BVIC- 11F8 and BVIC-A12 (40 mg/kg of each antibody) resulted in the best antitumor activity although with a total antibody that was twice that of the di-diabody.
[0117] Consistent with the BxPC3 result, HT29 xenografts were less responsive to treatment with individual anti-EGFR or anti-IGFR antibodies than to treatment with the di- diabody (Fig. 13B). Treatment with either BVIC-A12 or BVIC-C225, an anti-EGFR antibody
- 37 -
NYOl 924757 vl that is functionally equivalent to IMC-11F8 regarding both in vitro and in vivo antitumor activity (Marie, P. et al., 2004, Proc. Amer. Assoc. Cancer Res. 45, Abstract #5353) resulted in tumor inhibition of 47% and 35% at day 40 post treatment, respectively (p<0.03 compared to the saline and the human IgG groups). The di-diabody yielded a tumor growth inhibition rate of 58%, which is comparable to that achieved by the combination of both IMC-C225 and IMC-A12 (63% tumor growth inhibition). There is no statistically significant difference in overall tumor inhibition, however, between groups treated with the di-diabody, the antibody combination, or the individual antibody at the end of the study.
- 38 -
NYOl 924757 vl

Claims

What is claimed is:
1. An antigen-binding protein comprising a complex of two first polypeptides and two second polypeptides, said first polypeptide having a first diabody polypeptide, and said second polypeptide having a second diabody polypeptide located to the N terminus of one of more heavy chain constant domains capable of stable self association, said first and second diabody polypeptides capable of stable association to form a heterodimer having two antigen binding sites.
2. The antigen-binding protein of Claim 1, wherein said antigen binding sites of the heterodimer have the same specificity.
3. The antigen-binding protein of Claim 1 , wherein said antigen binding sites of the heterodimer have different specificities.
4. The antigen-binding protein of Claim 3, wherein said specificities are for epitopes which reside on the same antigen.
5. The antigen-binding protein of Claim 3, wherein said specificities are for epitopes which reside on different antigens.
6. The antigen-binding protein of Claim 1 , wherein said second polypeptide consists essentially of a second diabody polypeptide, a hinge region, and a CR2 domain..
7. The antigen-binding protein of Claim 1 , wherein said second polypeptide consists essentially of a second diabody polypeptide, a hinge region, and a CH3 domain.
8. The antigen-binding protein of Claim 1, wherein said second polypeptide consists essentially of a second diabody polypeptide, a hinge region, a CH2 domain, and a CR3 domain.
9. The antigen-binding protein of Claim 1, wherein said constant domains are mammalian constant domains.
10. The antigen-binding protein of Claim 1, wherein said constant domains are human constant domains.
- 39 -
NYOl 924757 vl
11. The antigen-binding protein of Claim 1 which has a constant domain capable of binding to an Fc receptor.
12. The antigen-binding protein of Claim 1 which is capable of effecting complement mediated cytoxicity (CMC).
13. The antigen-binding protein of Claim 1 which is capable of effecting antibody dependent cell-mediated cytoxicity (ADCC).
14. The antigen-binding protein of Claim 1 which is linked to an anti-tumor agent.
15. The antigen-binding protein of Claim 1 which neutralizes activation of a VEGF receptor.
16. The antigen-binding protein of Claim 15, wherein the VEGF receptor is a human VEGF receptor.
17. The antigen-binding protein of Claim 15, wherein the VEGF receptor is VEGFR-I.
18. The antigen-binding protein of Claim 15, wherein the VEGF receptor is VEGFR-2.
19. The antigen-binding protein of Claim 1, wherein at least one of the antigen binding sites is specific for KDR.
20. The antigen-binding protein of Claim 19, which neutralizes activation of KDR.
21. The antigen-binding protein of Claim 19, wherein one or both of said antigen- binding sites is the variable region of IMC- 1121.
22. The antigen-binding protein of Claim 1, wherein at least one of the antigen binding sites is specific for FLT-I.
23. The antigen-binding protein of Claim 22, which neutralizes activation of FLTl.
- 40 -
NYOl 924757 vl
24. The antigen-binding protein of Claim 22, wherein one or both of said antigen- binding sites is the variable region of IMC- 18Fl.
25. The antigen-binding protein of Claim 1, wherein at least one of the antigen binding sites is specific for FLT-4.
26. The antigen-binding protein of Claim 25, which neutralizes activation of FLT- 4.
27. The antigen-binding protein of Claim 1, wherein at least one of the antigen binding sites is specific for EGFR.
28. The antigen-binding protein of Claim 27, which neutralizes activation of EGFR.
29. The antigen-binding protein of Claim 27, wherein one or both of said antigen- binding sites is the variable region of IMC-C225.
30. The antigen-binding protein of Claim 27, wherein one or both of said antigen- binding sites is the variable region of IMC- 18Fl.
31. The antigen-binding protein of Claim 1 , wherein at least one of the antigen binding sites is specific for HER2.
32. The antigen-binding protein of Claim 31 , which neutralizes activation of HER2.
33. The antigen-binding protein of Claim 1, wherein at least one of the antigen binding sites is specific for IGFR.
34. The antigen-binding protein of Claim 33, which neutralizes activation of IGFR.
35. The antigen-binding protein of Claim 33, wherein one or both of said antigen- binding sites is the variable region of IMC-A12.
36. The antigen-binding protein of Claim 33, wherein one or both of said antigen- binding sites is the variable region of IMC-2F8.
- 41 -
NYOl 924757 vl
37. The antigen-binding protein of Claim 1, wherein at least one of the antigen binding sites is specific for RON.
38. The antigen-binding protein of Claim 37, which neutralizes activation of RON.
39. The antigen-binding protein of Claim 37, wherein one or both of said antigen- binding sites is the variable region of IMC-41A10 or the variable region of IMC-41A12.
40. The antigen-binding protein of Claim 1, wherein at least one of the antigen binding sites is specific for FGFR.
41. The antigen-binding protein of Claim 40, which neutralizes activation of FGFR.
42. The antigen-binding protein of Claim 1 , wherein at least one of the antigen binding sites is specific for PDGFRα.
43. The antigen-binding protein of Claim 42, which neutralizes activation of PDGFRα.
44. The antigen-binding protein of Claim 1 , wherein at least one of the antigen binding sites is specific for a receptor tyrosine kinase.
45. The antigen-binding protein of Claim 1 , wherein one of the antigen binding sites is specific for KDR and the other antigen-binding site is specific for FLT-I.
46. The antigen-binding protein of Claim 1 , wherein one of the antigen binding sites is specific for EGFR and the other antigen-binding site is specific for IGFR.
47. The antigen-binding protein of Claim 1, wherein one of the antigen binding sites is specific for EGFR and the other antigen-binding site is specific for RON.
48. A method of making an antigen-binding protein, which comprises coexpressing in a host cell a recombinant DNA construct encoding a first polypeptide having a first diabody polypeptide, and
- 42 -
NYOl 924757 vl a recombinant DNA construct encoding a second polypeptide having a second diabody polypeptide located to the N terminus of one of more heavy chain constant domains capable of stable self association, said first and second diabody polypeptides capable of associating to form a heterodimer having two antigen binding sites, for a time and in a manner sufficient to allow expression of said polypeptides and formation of said antigen binding protein; and recovering said antigen binding protein.
49. The method of Claim 48, wherein said constructs are on the same DNA expression vector.
50. The method of Claim 48, wherein said constructs are on different DNA expression vectors.
51. The method of Claim 48, wherein said host cell is a bacterial cell, a yeast cell, or a mammalian cell.
52. The method of Claim 48, wherein said antigen-binding protein is secreted from the host cell.
53. A method of neutralizing the activation of a receptor tyrosine kinase, which comprises contacting a cell with an antigen-binding protein of Claim 1, wherein at least one of the antigen-binding sites of the antigen-binding protein is specific for the receptor tyrosine kinase, in an amount sufficient to neutralize activation of said receptor.
54. The method of Claim 53, wherein the receptor tyrosine kinase is VEGFR-I .
55. The method of Claim 53 , wherein the receptor tyrosine kinase is VEGFR-2.
56. The method of Claim 53, wherein the receptor tyrosine kinase is EGFR.
57. The method of Claim 53, wherein the receptor tyrosine kinase is IGFR.
58. The method of Claim 53, wherein the receptor tyrosine kinase is RON.
- 43 -
NYOl 924757 vl
59. A method of inhibiting angiogenesis which comprises treating a cell that expresses a VEGF receptor with an antigen-binding protein of Claim 1, wherein at least one of the antigen-binding sites is specific for the VEGF receptor.
60. A method of inhibiting tumor growth which comprises treating a cell that expresses a VEGF receptor with an antigen-binding protein of Claim 1, wherein at least one of the antigen-binding sites is specific for the VEGF receptor.
61. The method of Claim 60 or Claim 60, wherein the second antigen-binding site is specific for a different VEGF receptor.
62. The method of Claim 60 or Claim 60, wherein the second antigen-binding site is specific for EGFR.
63. A method of inhibiting tumor growth which comprises treating a cell that expresses EGFR with an antigen-binding protein of Claim 1, wherein at least one of the antigen-binding sites is specific for EGFR.
64. The method of Claim 65, wherein the second antigen-binding site is specific for IGFR.
65. The method of Claim 65, wherein the second antigen-binding site is specific for RON.
66. A method of inhibiting tumor growth which comprises treating a cell that expresses a RTK with an antigen-binding protein of Claim 1, wherein at least one of the antigen-binding sites is specific for the RTK, wherein the RTK is selected from the group consisting of VEGFR-I, VEGFR-2, VEGFR-3, EGFR, HER2, IGFR, RON, FGFR, PDGFR, NGFR, Tek, and Tie2.
- 44 -
NYOl 924757 vl
PCT/US2005/025472 2004-07-17 2005-07-18 Novel tetravalent bispecific antibody WO2006020258A2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP05773142A EP1786918A4 (en) 2004-07-17 2005-07-18 Novel tetravalent bispecific antibody
JP2007521716A JP2008512352A (en) 2004-07-17 2005-07-18 Novel tetravalent bispecific antibody

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US58868804P 2004-07-17 2004-07-17
US60/588,688 2004-07-17

Publications (2)

Publication Number Publication Date
WO2006020258A2 true WO2006020258A2 (en) 2006-02-23
WO2006020258A3 WO2006020258A3 (en) 2006-10-12

Family

ID=35907983

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/025472 WO2006020258A2 (en) 2004-07-17 2005-07-18 Novel tetravalent bispecific antibody

Country Status (3)

Country Link
EP (1) EP1786918A4 (en)
JP (1) JP2008512352A (en)
WO (1) WO2006020258A2 (en)

Cited By (301)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007092453A2 (en) 2006-02-03 2007-08-16 Imclone Systems Incorporated Igf-ir antagonists as adjuvants for treatment of prostate cancer
EP1928506A2 (en) * 2005-08-19 2008-06-11 Abbott Laboratories Dual variable domain immunoglobin and uses thereof
EP2056869A2 (en) * 2006-08-18 2009-05-13 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
WO2009068630A1 (en) * 2007-11-27 2009-06-04 Ablynx N.V. Immunoglobulin constructs
EP2100618A2 (en) 2005-06-17 2009-09-16 Imclone LLC PDGFR-alpha antagonists for treatment of metastatic bone cancer
WO2010028797A1 (en) * 2008-09-10 2010-03-18 F. Hoffmann-La Roche Ag Multivalent antibodies
WO2010028798A1 (en) * 2008-09-10 2010-03-18 F. Hoffmann-La Roche Ag Multivalent antibodies
WO2010028795A1 (en) * 2008-09-10 2010-03-18 F. Hoffmann-La Roche Ag Multivalent antibodies
WO2010034441A1 (en) * 2008-09-26 2010-04-01 F. Hoffmann-La Roche Ag Bispecific anti-egfr/anti-igf-1r antibodies
US7740850B2 (en) 2007-04-17 2010-06-22 ImClone, LLC PDGFRβ-specific antibodies
WO2010093055A1 (en) * 2009-02-10 2010-08-19 Daiichi Sankyo Company, Limited Anti-mst1r antibodies and uses thereof
WO2010112193A1 (en) 2009-04-02 2010-10-07 Roche Glycart Ag Multispecific antibodies comprising full length antibodies and single chain fab fragments
WO2010112194A1 (en) 2009-04-02 2010-10-07 F. Hoffmann-La Roche Ag Antigen-binding polypeptides and multispecific antibodies comprising them
US7811562B2 (en) 2004-12-03 2010-10-12 Schering Corporation Biomarkers for pre-selection of patients for anti-IGF1R therapy
WO2010115589A1 (en) 2009-04-07 2010-10-14 Roche Glycart Ag Trivalent, bispecific antibodies
WO2010115551A1 (en) 2009-04-07 2010-10-14 Roche Glycart Ag Bispecific anti-erbb-1/anti-c-met antibodies
WO2010115552A1 (en) 2009-04-07 2010-10-14 Roche Glycart Ag Bispecific anti-erbb-3/anti-c-met antibodies
WO2010136482A1 (en) * 2009-05-28 2010-12-02 Glaxo Group Limited Antigen-binding proteins
CN102149825A (en) * 2008-07-08 2011-08-10 雅培制药有限公司 Prostaglandin E2 dual variable domain immunoglobulins and uses thereof
WO2011117330A1 (en) 2010-03-26 2011-09-29 Roche Glycart Ag Bispecific antibodies
WO2011117329A1 (en) 2010-03-26 2011-09-29 F. Hoffmann-La Roche Ag Bispecific, bivalent anti-vegf/anti-ang-2 antibodies
WO2011138391A1 (en) 2010-05-06 2011-11-10 Novartis Ag Compositions and methods of use for therapeutic low density lipoprotein - related protein 6 (lrp6) multivalent antibodies
WO2011138392A1 (en) 2010-05-06 2011-11-10 Novartis Ag Compositions and methods of use for therapeutic low density lipoprotein -related protein 6 (lrp6) antibodies
US8062886B2 (en) 2003-11-12 2011-11-22 Schering Corporation Plasmid system for multigene expression
US8094026B1 (en) 2011-05-02 2012-01-10 Robert M Green Organized retail crime detection security system and method
WO2012006341A2 (en) 2010-07-06 2012-01-12 Aveo Pharmaceuticals, Inc. Anti-ron antibodies
WO2012025530A1 (en) 2010-08-24 2012-03-01 F. Hoffmann-La Roche Ag Bispecific antibodies comprising a disulfide stabilized - fv fragment
WO2012069557A1 (en) * 2010-11-24 2012-05-31 Glaxo Group Limited Multispecific antigen binding proteins targeting hgf
US8221765B2 (en) 2008-05-22 2012-07-17 Bristol-Myers Squibb Company Multivalent fibronectin based scaffold domain proteins
US8268314B2 (en) 2008-10-08 2012-09-18 Hoffmann-La Roche Inc. Bispecific anti-VEGF/anti-ANG-2 antibodies
EP2500353A3 (en) * 2005-08-19 2012-10-10 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US8343501B2 (en) 2008-11-24 2013-01-01 Bristol-Myers Squibb Company Bispecific EGFR/IGFIR binding molecules
US8409577B2 (en) 2006-06-12 2013-04-02 Emergent Product Development Seattle, Llc Single chain multivalent binding proteins with effector function
WO2013067355A1 (en) 2011-11-04 2013-05-10 Novartis Ag Low density lipoprotein-related protein 6 (lrp6) - half life extender constructs
US8470332B2 (en) 2006-11-22 2013-06-25 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins for tyrosine kinases receptors, including IGF-IR
EP2626371A1 (en) 2007-07-31 2013-08-14 MedImmune, LLC Multispecific epitope binding proteins and uses thereof
US8524244B2 (en) 2008-02-14 2013-09-03 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins that bind EGFR
WO2013150043A1 (en) 2012-04-05 2013-10-10 F. Hoffmann-La Roche Ag Bispecific antibodies against human tweak and human il17 and uses thereof
US8586714B2 (en) 2009-09-01 2013-11-19 Abbvie, Inc. Dual variable domain immunoglobulins and uses thereof
WO2014006124A1 (en) 2012-07-04 2014-01-09 F. Hoffmann-La Roche Ag Covalently linked antigen-antibody conjugates
US8716450B2 (en) 2009-10-15 2014-05-06 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US8722855B2 (en) 2009-10-28 2014-05-13 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US8735546B2 (en) 2010-08-03 2014-05-27 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
WO2014099997A1 (en) 2012-12-18 2014-06-26 Novartis Ag Compositions and methods that utilize a peptide tag that binds to hyaluronan
WO2014130635A1 (en) 2013-02-20 2014-08-28 Novartis Ag Effective targeting of primary human leukemia using anti-cd123 chimeric antigen receptor engineered t cells
WO2014130657A1 (en) 2013-02-20 2014-08-28 The Trustees Of The University Of Pennsylvania Treatment of cancer using humanized anti-egfrviii chimeric antigen receptor
US20140274801A1 (en) * 2013-03-14 2014-09-18 Elwha Llc Compositions, methods, and computer systems related to making and administering modified t cells
WO2014153270A1 (en) 2013-03-16 2014-09-25 Novartis Ag Treatment of cancer using humanized anti-cd19 chimeric antigen receptor
US8853366B2 (en) 2001-01-17 2014-10-07 Emergent Product Development Seattle, Llc Binding domain-immunoglobulin fusion proteins
US8969289B2 (en) 2010-05-03 2015-03-03 Bristol-Myers Squibb Company Serum albumin binding molecules
US8987418B2 (en) 2013-03-15 2015-03-24 Abbvie Inc. Dual specific binding proteins directed against IL-1β and/or IL-17
US9029508B2 (en) 2008-04-29 2015-05-12 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9035027B2 (en) 2008-06-03 2015-05-19 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9046513B2 (en) 2010-08-26 2015-06-02 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9045551B2 (en) 2012-11-01 2015-06-02 Abbvie Inc. Anti-DLL4/VEGF dual variable domain immunoglobulin and uses thereof
WO2015090229A1 (en) 2013-12-20 2015-06-25 Novartis Ag Regulatable chimeric antigen receptor
WO2015090230A1 (en) 2013-12-19 2015-06-25 Novartis Ag Human mesothelin chimeric antigen receptors and uses thereof
WO2015101586A1 (en) 2014-01-03 2015-07-09 F. Hoffmann-La Roche Ag Bispecific anti-hapten/anti-blood brain barrier receptor antibodies, complexes thereof and their use as blood brain barrier shuttles
WO2015101587A1 (en) 2014-01-03 2015-07-09 F. Hoffmann-La Roche Ag Covalently linked helicar-anti-helicar antibody conjugates and uses thereof
WO2015101589A1 (en) 2014-01-03 2015-07-09 F. Hoffmann-La Roche Ag Covalently linked polypeptide toxin-antibody conjugates
WO2015112626A1 (en) 2014-01-21 2015-07-30 June Carl H Enhanced antigen presenting ability of car t cells by co-introduction of costimulatory molecules
US9101609B2 (en) 2008-04-11 2015-08-11 Emergent Product Development Seattle, Llc CD37 immunotherapeutic and combination with bifunctional chemotherapeutic thereof
US9109026B2 (en) 2008-06-03 2015-08-18 Abbvie, Inc. Dual variable domain immunoglobulins and uses thereof
US9120870B2 (en) 2011-12-30 2015-09-01 Abbvie Inc. Dual specific binding proteins directed against IL-13 and IL-17
WO2015138920A1 (en) 2014-03-14 2015-09-17 Novartis Ag Antibody molecules to lag-3 and uses thereof
WO2015142675A2 (en) 2014-03-15 2015-09-24 Novartis Ag Treatment of cancer using chimeric antigen receptor
WO2015142661A1 (en) 2014-03-15 2015-09-24 Novartis Ag Regulatable chimeric antigen receptor
WO2015157252A1 (en) 2014-04-07 2015-10-15 BROGDON, Jennifer Treatment of cancer using anti-cd19 chimeric antigen receptor
WO2015198243A2 (en) 2014-06-25 2015-12-30 Novartis Ag Compositions and methods for long acting proteins
WO2015198217A2 (en) 2013-02-08 2015-12-30 Novartis Ag Compositions and methods for long-acting antibodies targeting il-17
WO2015198240A2 (en) 2014-06-25 2015-12-30 Novartis Ag Compositions and methods for long acting proteins
WO2016014565A2 (en) 2014-07-21 2016-01-28 Novartis Ag Treatment of cancer using humanized anti-bcma chimeric antigen receptor
WO2016014553A1 (en) 2014-07-21 2016-01-28 Novartis Ag Sortase synthesized chimeric antigen receptors
WO2016014530A1 (en) 2014-07-21 2016-01-28 Novartis Ag Combinations of low, immune enhancing. doses of mtor inhibitors and cars
WO2016025880A1 (en) 2014-08-14 2016-02-18 Novartis Ag Treatment of cancer using gfr alpha-4 chimeric antigen receptor
US9266967B2 (en) 2007-12-21 2016-02-23 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
WO2016044605A1 (en) 2014-09-17 2016-03-24 Beatty, Gregory Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
CN105481981A (en) * 2016-01-27 2016-04-13 中国人民解放军第二军医大学 Bispecific antibody targeting VEGF and application thereof
WO2016061142A1 (en) 2014-10-14 2016-04-21 Novartis Ag Antibody molecules to pd-l1 and uses thereof
WO2016090034A2 (en) 2014-12-03 2016-06-09 Novartis Ag Methods for b cell preconditioning in car therapy
WO2016126608A1 (en) 2015-02-02 2016-08-11 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
US9441034B2 (en) 2008-03-27 2016-09-13 Zymogenetics, Inc. Compositions and methods for inhibiting PDGFRβ and VEGF-A
WO2016164708A1 (en) 2015-04-10 2016-10-13 Adimab, Llc Methods for purifying heterodimeric multispecific antibodies from parental homodimeric antibody species
WO2016164731A2 (en) 2015-04-08 2016-10-13 Novartis Ag Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car) - expressing cell
WO2016168595A1 (en) 2015-04-17 2016-10-20 Barrett David Maxwell Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
WO2016172583A1 (en) 2015-04-23 2016-10-27 Novartis Ag Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
US9499855B2 (en) 2013-03-14 2016-11-22 Elwha Llc Compositions, methods, and computer systems related to making and administering modified T cells
WO2016187349A1 (en) 2015-05-18 2016-11-24 Tcr2, Inc. Compositions and methods for tcr reprogramming using fusion proteins
WO2017015427A1 (en) 2015-07-21 2017-01-26 Novartis Ag Methods for improving the efficacy and expansion of immune cells
WO2017019894A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to lag-3
WO2017019897A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to tim-3
US9562089B2 (en) 2010-05-26 2017-02-07 Bristol-Myers Squibb Company Fibronectin based scaffold proteins having improved stability
US9567403B2 (en) 2013-08-06 2017-02-14 Bio-Thera Solutions, Ltd. Bispecific antibodies which bind EGFR and VEGF
WO2017027392A1 (en) 2015-08-07 2017-02-16 Novartis Ag Treatment of cancer using chimeric cd3 receptor proteins
US9587237B2 (en) 2013-03-14 2017-03-07 Elwha Llc Compositions, methods, and computer systems related to making and administering modified T cells
US9605061B2 (en) 2010-07-29 2017-03-28 Xencor, Inc. Antibodies with modified isoelectric points
US9605084B2 (en) 2013-03-15 2017-03-28 Xencor, Inc. Heterodimeric proteins
US9650446B2 (en) 2013-01-14 2017-05-16 Xencor, Inc. Heterodimeric proteins
WO2017091786A1 (en) 2015-11-23 2017-06-01 Novartis Ag Optimized lentiviral transfer vectors and uses thereof
US9676845B2 (en) 2009-06-16 2017-06-13 Hoffmann-La Roche, Inc. Bispecific antigen binding proteins
WO2017106656A1 (en) 2015-12-17 2017-06-22 Novartis Ag Antibody molecules to pd-1 and uses thereof
WO2017106810A2 (en) 2015-12-17 2017-06-22 Novartis Ag Combination of c-met inhibitor with antibody molecule to pd-1 and uses thereof
US9688758B2 (en) 2012-02-10 2017-06-27 Genentech, Inc. Single-chain antibodies and other heteromultimers
WO2017114497A1 (en) 2015-12-30 2017-07-06 Novartis Ag Immune effector cell therapies with enhanced efficacy
US9701759B2 (en) 2013-01-14 2017-07-11 Xencor, Inc. Heterodimeric proteins
WO2017125897A1 (en) 2016-01-21 2017-07-27 Novartis Ag Multispecific molecules targeting cll-1
US9718883B2 (en) 2003-09-10 2017-08-01 Amgen Fremont Inc. Antibodies to M-CSF
US9738722B2 (en) 2013-01-15 2017-08-22 Xencor, Inc. Rapid clearance of antigen complexes using novel antibodies
WO2017149515A1 (en) 2016-03-04 2017-09-08 Novartis Ag Cells expressing multiple chimeric antigen receptor (car) molecules and uses therefore
US9765153B2 (en) 2012-07-04 2017-09-19 Hoffmann-La Roche Inc. Anti-biotin antibodies and methods of use
WO2017165683A1 (en) 2016-03-23 2017-09-28 Novartis Ag Cell secreted minibodies and uses thereof
WO2017172981A2 (en) 2016-03-29 2017-10-05 University Of Southern California Chimeric antigen receptors targeting cancer
WO2017181119A2 (en) 2016-04-15 2017-10-19 Novartis Ag Compositions and methods for selective protein expression
US9822186B2 (en) 2014-03-28 2017-11-21 Xencor, Inc. Bispecific antibodies that bind to CD38 and CD3
WO2017210617A2 (en) 2016-06-02 2017-12-07 Porter, David, L. Therapeutic regimens for chimeric antigen receptor (car)- expressing cells
US9840554B2 (en) 2015-06-15 2017-12-12 Abbvie Inc. Antibodies against platelet-derived growth factor (PDGF)
US9850320B2 (en) 2014-11-26 2017-12-26 Xencor, Inc. Heterodimeric antibodies to CD3 X CD20
US9856327B2 (en) 2014-11-26 2018-01-02 Xencor, Inc. Heterodimeric antibodies to CD3 X CD123
WO2018013918A2 (en) 2016-07-15 2018-01-18 Novartis Ag Treatment and prevention of cytokine release syndrome using a chimeric antigen receptor in combination with a kinase inhibitor
WO2018023025A1 (en) 2016-07-28 2018-02-01 Novartis Ag Combination therapies of chimeric antigen receptors adn pd-1 inhibitors
US9884921B2 (en) 2014-07-01 2018-02-06 Pfizer Inc. Bispecific heterodimeric diabodies and uses thereof
WO2018026819A2 (en) 2016-08-01 2018-02-08 Novartis Ag Treatment of cancer using a chimeric antigen receptor in combination with an inhibitor of a pro-m2 macrophage molecule
WO2018026953A1 (en) 2016-08-02 2018-02-08 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
US9925272B2 (en) 2012-07-04 2018-03-27 Hoffmann-La Roche Inc. Anti-theophylline antibodies and methods of use
WO2018067993A1 (en) 2016-10-07 2018-04-12 TCR2 Therapeutics Inc. Compositions and methods for t-cell receptors reprogramming using fusion proteins
WO2018067992A1 (en) 2016-10-07 2018-04-12 Novartis Ag Chimeric antigen receptors for the treatment of cancer
US9982036B2 (en) 2011-02-28 2018-05-29 Hoffmann-La Roche Inc. Dual FC antigen binding proteins
WO2018098365A2 (en) 2016-11-22 2018-05-31 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
US9994646B2 (en) 2009-09-16 2018-06-12 Genentech, Inc. Coiled coil and/or tether containing protein complexes and uses thereof
WO2018111340A1 (en) 2016-12-16 2018-06-21 Novartis Ag Methods for determining potency and proliferative function of chimeric antigen receptor (car)-t cells
WO2018140725A1 (en) 2017-01-26 2018-08-02 Novartis Ag Cd28 compositions and methods for chimeric antigen receptor therapy
WO2018144535A1 (en) 2017-01-31 2018-08-09 Novartis Ag Treatment of cancer using chimeric t cell receptor proteins having multiple specificities
WO2018141910A1 (en) 2017-02-02 2018-08-09 Amgen Research (Munich) Gmbh Low ph pharmaceutical composition comprising t cell engaging antibody constructs
WO2018160731A1 (en) 2017-02-28 2018-09-07 Novartis Ag Shp inhibitor compositions and uses for chimeric antigen receptor therapy
US10093733B2 (en) 2014-12-11 2018-10-09 Abbvie Inc. LRP-8 binding dual variable domain immunoglobulin proteins
US10106624B2 (en) 2013-03-15 2018-10-23 Xencor, Inc. Heterodimeric proteins
US10106612B2 (en) 2012-06-27 2018-10-23 Hoffmann-La Roche Inc. Method for selection and production of tailor-made highly selective and multi-specific targeting entities containing at least two different binding entities and uses thereof
WO2018201056A1 (en) 2017-04-28 2018-11-01 Novartis Ag Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
WO2018201051A1 (en) 2017-04-28 2018-11-01 Novartis Ag Bcma-targeting agent, and combination therapy with a gamma secretase inhibitor
US10131710B2 (en) 2013-01-14 2018-11-20 Xencor, Inc. Optimized antibody variable regions
US10138293B2 (en) 2007-12-21 2018-11-27 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US10143748B2 (en) 2005-07-25 2018-12-04 Aptevo Research And Development Llc B-cell reduction using CD37-specific and CD20-specific binding molecules
WO2018232020A1 (en) 2017-06-13 2018-12-20 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
WO2018237157A1 (en) 2017-06-22 2018-12-27 Novartis Ag Antibody molecules to cd73 and uses thereof
WO2019006007A1 (en) 2017-06-27 2019-01-03 Novartis Ag Dosage regimens for anti-tim-3 antibodies and uses thereof
WO2019018730A1 (en) 2017-07-20 2019-01-24 Novartis Ag Dosage regimens of anti-lag-3 antibodies and uses thereof
US10227410B2 (en) 2015-12-07 2019-03-12 Xencor, Inc. Heterodimeric antibodies that bind CD3 and PSMA
US10227411B2 (en) 2015-03-05 2019-03-12 Xencor, Inc. Modulation of T cells with bispecific antibodies and FC fusions
US10251952B2 (en) 2014-06-26 2019-04-09 Hoffmann-La Roche Inc. Humanized anti-tau(pS422) antibody brain shuttles and use thereof
WO2019079569A1 (en) 2017-10-18 2019-04-25 Novartis Ag Compositions and methods for selective protein degradation
WO2019084288A1 (en) 2017-10-25 2019-05-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2019089798A1 (en) 2017-10-31 2019-05-09 Novartis Ag Anti-car compositions and methods
WO2019099838A1 (en) 2017-11-16 2019-05-23 Novartis Ag Combination therapies
US10316088B2 (en) 2016-06-28 2019-06-11 Xencor, Inc. Heterodimeric antibodies that bind somatostatin receptor 2
US10323099B2 (en) 2013-10-11 2019-06-18 Hoffmann-La Roche Inc. Multispecific domain exchanged common variable light chain antibodies
EP3514179A1 (en) 2014-01-24 2019-07-24 Dana-Farber Cancer Institute, Inc. Antibody molecules to pd-1 and uses thereof
WO2019152660A1 (en) 2018-01-31 2019-08-08 Novartis Ag Combination therapy using a chimeric antigen receptor
US20190248918A1 (en) * 2014-03-21 2019-08-15 X-Body, Inc. Bi-specific antigen-binding polypeptides
US10428155B2 (en) 2014-12-22 2019-10-01 Xencor, Inc. Trispecific antibodies
US10442851B2 (en) 2014-03-20 2019-10-15 Bristol-Myers Squibb Company Serum albumin-binding fibronectin type III domains
WO2019200229A1 (en) 2018-04-13 2019-10-17 Novartis Ag Dosage regimens for anti-pd-l1 antibodies and uses thereof
WO2019210153A1 (en) 2018-04-27 2019-10-31 Novartis Ag Car t cell therapies with enhanced efficacy
US10465000B2 (en) 2013-12-20 2019-11-05 Hoffmann-La Roche Inc. Humanized anti-Tau(pS422) antibodies and methods of use
WO2019213282A1 (en) 2018-05-01 2019-11-07 Novartis Ag Biomarkers for evaluating car-t cells to predict clinical outcome
US10487155B2 (en) 2013-01-14 2019-11-26 Xencor, Inc. Heterodimeric proteins
WO2019227003A1 (en) 2018-05-25 2019-11-28 Novartis Ag Combination therapy with chimeric antigen receptor (car) therapies
WO2019226658A1 (en) 2018-05-21 2019-11-28 Compass Therapeutics Llc Multispecific antigen-binding compositions and methods of use
WO2019226617A1 (en) 2018-05-21 2019-11-28 Compass Therapeutics Llc Compositions and methods for enhancing the killing of target cells by nk cells
WO2019232244A2 (en) 2018-05-31 2019-12-05 Novartis Ag Antibody molecules to cd73 and uses thereof
US10501543B2 (en) 2016-10-14 2019-12-10 Xencor, Inc. IL15/IL15Rα heterodimeric Fc-fusion proteins
WO2019237035A1 (en) 2018-06-08 2019-12-12 Intellia Therapeutics, Inc. Compositions and methods for immunooncology
WO2019241426A1 (en) 2018-06-13 2019-12-19 Novartis Ag Bcma chimeric antigen receptors and uses thereof
WO2019246293A2 (en) 2018-06-19 2019-12-26 Atarga, Llc Antibody molecules to complement component 5 and uses thereof
US10519242B2 (en) 2013-03-15 2019-12-31 Xencor, Inc. Targeting regulatory T cells with heterodimeric proteins
US10526417B2 (en) 2014-11-26 2020-01-07 Xencor, Inc. Heterodimeric antibodies that bind CD3 and CD38
WO2020010079A2 (en) 2018-07-02 2020-01-09 Amgen Inc. Anti-steap1 antigen-binding protein
WO2020012337A1 (en) 2018-07-10 2020-01-16 Novartis Ag 3-(5-amino-1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and their use in the treatment of i karos family zinc finger 2 (ikzf2)-dependent diseases
US10544187B2 (en) 2013-03-15 2020-01-28 Xencor, Inc. Targeting regulatory T cells with heterodimeric proteins
WO2020021465A1 (en) 2018-07-25 2020-01-30 Advanced Accelerator Applications (Italy) S.R.L. Method of treatment of neuroendocrine tumors
WO2020047452A2 (en) 2018-08-31 2020-03-05 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2020047501A1 (en) 2018-08-30 2020-03-05 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
WO2020047449A2 (en) 2018-08-31 2020-03-05 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
EP3623380A1 (en) 2013-03-15 2020-03-18 Michael C. Milone Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
WO2020069405A1 (en) 2018-09-28 2020-04-02 Novartis Ag Cd22 chimeric antigen receptor (car) therapies
WO2020069409A1 (en) 2018-09-28 2020-04-02 Novartis Ag Cd19 chimeric antigen receptor (car) and cd22 car combination therapies
US10611825B2 (en) 2011-02-28 2020-04-07 Hoffmann La-Roche Inc. Monovalent antigen binding proteins
US10633457B2 (en) 2014-12-03 2020-04-28 Hoffmann-La Roche Inc. Multispecific antibodies
EP3660042A1 (en) 2014-07-31 2020-06-03 Novartis AG Subset-optimized chimeric antigen receptor-containing t-cells
US10683345B2 (en) 2012-07-13 2020-06-16 Roche Glycart Ag Bispecific anti-VEGF/anti-ANG-2 antibodies and their use in the treatment of ocular vascular diseases
WO2020128972A1 (en) 2018-12-20 2020-06-25 Novartis Ag Dosing regimen and pharmaceutical combination comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
WO2020128898A1 (en) 2018-12-20 2020-06-25 Novartis Ag Pharmaceutical combinations
US10730954B2 (en) 2017-05-12 2020-08-04 Harpoon Therapeutics, Inc. MSLN targeting trispecific proteins and methods of use
WO2020165833A1 (en) 2019-02-15 2020-08-20 Novartis Ag 3-(1-oxo-5-(piperidin-4-yl)isoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2020165834A1 (en) 2019-02-15 2020-08-20 Novartis Ag Substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2020165868A1 (en) 2019-02-15 2020-08-20 Perkinelmer Cellular Technologies Germany Gmbh Low-power microscope-objective pre-scan and high-power microscope-objective scan in x,y and z-direction for imaging objects such as cells using a microscope
WO2020172553A1 (en) 2019-02-22 2020-08-27 Novartis Ag Combination therapies of egfrviii chimeric antigen receptors and pd-1 inhibitors
WO2020176397A1 (en) 2019-02-25 2020-09-03 Novartis Ag Mesoporous silica particles compositions for viral delivery
US10766946B2 (en) 2015-09-23 2020-09-08 Bristol-Myers Squibb Company Fast-off rate serum albumin binding fibronectin type III domains
EP3712171A1 (en) 2014-08-19 2020-09-23 Novartis AG Treatment of cancer using a cd123 chimeric antigen receptor
US10787518B2 (en) 2016-06-14 2020-09-29 Xencor, Inc. Bispecific checkpoint inhibitor antibodies
US10793632B2 (en) 2016-08-30 2020-10-06 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
WO2020205523A1 (en) 2019-03-29 2020-10-08 Atarga, Llc Anti fgf23 antibody
EP3722316A1 (en) 2014-07-21 2020-10-14 Novartis AG Treatment of cancer using a cd33 chimeric antigen receptor
WO2020210678A1 (en) 2019-04-12 2020-10-15 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
US10815311B2 (en) 2018-09-25 2020-10-27 Harpoon Therapeutics, Inc. DLL3 binding proteins and methods of use
WO2020219742A1 (en) 2019-04-24 2020-10-29 Novartis Ag Compositions and methods for selective protein degradation
US10822402B2 (en) 2015-06-24 2020-11-03 Hoffmann-La Roche Inc. Humanized anti-tau(pS422) antibodies and methods of use
US10844134B2 (en) 2016-11-23 2020-11-24 Harpoon Therapeutics, Inc. PSMA targeting trispecific proteins and methods of use
US10849973B2 (en) 2016-11-23 2020-12-01 Harpoon Therapeutics, Inc. Prostate specific membrane antigen binding protein
US10851178B2 (en) 2011-10-10 2020-12-01 Xencor, Inc. Heterodimeric human IgG1 polypeptides with isoelectric point modifications
US10858417B2 (en) 2013-03-15 2020-12-08 Xencor, Inc. Heterodimeric proteins
WO2021003432A1 (en) 2019-07-02 2021-01-07 Fred Hutchinson Cancer Research Center Recombinant ad35 vectors and related gene therapy improvements
US10927180B2 (en) 2017-10-13 2021-02-23 Harpoon Therapeutics, Inc. B cell maturation antigen binding proteins
WO2021035170A1 (en) 2019-08-21 2021-02-25 Precision Biosciences, Inc. Compositions and methods for tcr reprogramming using fusion proteins
US10934337B2 (en) 2019-03-15 2021-03-02 Cartesian Therapeutics, Inc. Anti-BCMA chimeric antigen receptors
US10954311B2 (en) 2015-05-21 2021-03-23 Harpoon Therapeutics, Inc. Trispecific binding proteins and methods of use
US10968276B2 (en) 2013-03-12 2021-04-06 Xencor, Inc. Optimized anti-CD3 variable regions
US10982006B2 (en) 2018-04-04 2021-04-20 Xencor, Inc. Heterodimeric antibodies that bind fibroblast activation protein
US10981992B2 (en) 2017-11-08 2021-04-20 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
WO2021079188A1 (en) 2019-10-21 2021-04-29 Novartis Ag Combination therapies with venetoclax and tim-3 inhibitors
WO2021079195A1 (en) 2019-10-21 2021-04-29 Novartis Ag Tim-3 inhibitors and uses thereof
US11014989B2 (en) 2015-01-26 2021-05-25 Cellectis Anti-CLL1 specific single-chain chimeric antigen receptors (scCARs) for cancer immunotherapy
WO2021108661A2 (en) 2019-11-26 2021-06-03 Novartis Ag Chimeric antigen receptors and uses thereof
WO2021123902A1 (en) 2019-12-20 2021-06-24 Novartis Ag Combination of anti tim-3 antibody mbg453 and anti tgf-beta antibody nis793, with or without decitabine or the anti pd-1 antibody spartalizumab, for treating myelofibrosis and myelodysplastic syndrome
US11046769B2 (en) 2018-11-13 2021-06-29 Compass Therapeutics Llc Multispecific binding constructs against checkpoint molecules and uses thereof
US11053316B2 (en) 2013-01-14 2021-07-06 Xencor, Inc. Optimized antibody variable regions
WO2021144657A1 (en) 2020-01-17 2021-07-22 Novartis Ag Combination comprising a tim-3 inhibitor and a hypomethylating agent for use in treating myelodysplastic syndrome or chronic myelomonocytic leukemia
WO2021146636A1 (en) 2020-01-17 2021-07-22 Becton, Dickinson And Company Methods and compositions for single cell secretomics
US11084863B2 (en) 2017-06-30 2021-08-10 Xencor, Inc. Targeted heterodimeric Fc fusion proteins containing IL-15 IL-15alpha and antigen binding domains
WO2021163618A1 (en) 2020-02-14 2021-08-19 Novartis Ag Method of predicting response to chimeric antigen receptor therapy
WO2021173995A2 (en) 2020-02-27 2021-09-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2021173985A2 (en) 2020-02-27 2021-09-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2021169977A1 (en) 2020-02-28 2021-09-02 南京北恒生物科技有限公司 Novel chimeric antigen receptor and use thereof
US11136403B2 (en) 2017-10-13 2021-10-05 Harpoon Therapeutics, Inc. Trispecific proteins and methods of use
WO2021222347A1 (en) 2020-04-29 2021-11-04 Amgen Inc. Pharmaceutical formulation
WO2021222355A1 (en) 2020-04-29 2021-11-04 Amgen Inc. Pharmaceutical formulation
WO2021231655A1 (en) 2020-05-12 2021-11-18 Lyell Immunopharma, Inc. Chimeric antigen receptor spacers
US11180563B2 (en) 2020-02-21 2021-11-23 Harpoon Therapeutics, Inc. FLT3 binding proteins and methods of use
WO2021238877A1 (en) 2020-05-27 2021-12-02 南京北恒生物科技有限公司 Engineered immune cell and use thereof
WO2021252920A1 (en) 2020-06-11 2021-12-16 Novartis Ag Zbtb32 inhibitors and uses thereof
WO2021249462A1 (en) 2020-06-11 2021-12-16 南京北恒生物科技有限公司 Engineered immune cell expressing nk inhibitory molecule and use thereof
WO2021260528A1 (en) 2020-06-23 2021-12-30 Novartis Ag Dosing regimen comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
WO2022011358A1 (en) * 2020-07-10 2022-01-13 Biomolecular Holdings Llc Tetrahedral antibodies
WO2022012591A1 (en) 2020-07-15 2022-01-20 南京北恒生物科技有限公司 Engineered immune cell for allotransplantation
WO2022013787A1 (en) 2020-07-16 2022-01-20 Novartis Ag Anti-betacellulin antibodies, fragments thereof, and multi-specific binding molecules
WO2022022745A1 (en) 2020-07-31 2022-02-03 南京北恒生物科技有限公司 Novel co-stimulatory domain and uses thereof
WO2022026592A2 (en) 2020-07-28 2022-02-03 Celltas Bio, Inc. Antibody molecules to coronavirus and uses thereof
WO2022029573A1 (en) 2020-08-03 2022-02-10 Novartis Ag Heteroaryl substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2022033537A1 (en) 2020-08-13 2022-02-17 南京北恒生物科技有限公司 Engineered immune cell and use thereof
WO2022040586A2 (en) 2020-08-21 2022-02-24 Novartis Ag Compositions and methods for in vivo generation of car expressing cells
WO2022043558A1 (en) 2020-08-31 2022-03-03 Advanced Accelerator Applications International Sa Method of treating psma-expressing cancers
WO2022043557A1 (en) 2020-08-31 2022-03-03 Advanced Accelerator Applications International Sa Method of treating psma-expressing cancers
WO2022046651A1 (en) 2020-08-24 2022-03-03 Amgen Inc. Pharmaceutical formulation comprising a bite, bispecific antibody, and methionine
WO2022052981A1 (en) 2020-09-10 2022-03-17 南京北恒生物科技有限公司 Chimeric antigen receptor comprising novel co-stimulatory domain and use thereof
US11312770B2 (en) 2017-11-08 2022-04-26 Xencor, Inc. Bispecific and monospecific antibodies using novel anti-PD-1 sequences
US11319355B2 (en) 2017-12-19 2022-05-03 Xencor, Inc. Engineered IL-2 Fc fusion proteins
WO2022095802A1 (en) 2020-11-03 2022-05-12 南京北恒生物科技有限公司 Chimeric antigen receptor targeting cd7 and use thereof
WO2022095803A1 (en) 2020-11-03 2022-05-12 南京北恒生物科技有限公司 Cd7-targeting humanized antibody and use thereof
WO2022097065A2 (en) 2020-11-06 2022-05-12 Novartis Ag ANTIBODY Fc VARIANTS
WO2022104061A1 (en) 2020-11-13 2022-05-19 Novartis Ag Combination therapies with chimeric antigen receptor (car)-expressing cells
WO2022105826A1 (en) 2020-11-23 2022-05-27 南京北恒生物科技有限公司 Nkg2a-targeting antibody and use thereof
US11352426B2 (en) 2015-09-21 2022-06-07 Aptevo Research And Development Llc CD3 binding polypeptides
US11358999B2 (en) 2018-10-03 2022-06-14 Xencor, Inc. IL-12 heterodimeric Fc-fusion proteins
EP4015525A2 (en) 2018-03-23 2022-06-22 GammaDelta Therapeutics Limited Lymphocytes expressing heterologous targeting constructs
WO2022152168A1 (en) 2021-01-12 2022-07-21 南京北恒生物科技有限公司 Ror1-targeting antibody and use thereof
EP4036109A2 (en) 2014-12-29 2022-08-03 Novartis AG Methods of making chimeric antigen receptor-expressing cells
WO2022162569A1 (en) 2021-01-29 2022-08-04 Novartis Ag Dosage regimes for anti-cd73 and anti-entpd2 antibodies and uses thereof
WO2022166365A1 (en) 2021-02-03 2022-08-11 南京北恒生物科技有限公司 Novel chimeric antigen receptor and use thereof
US11421022B2 (en) 2012-06-27 2022-08-23 Hoffmann-La Roche Inc. Method for making antibody Fc-region conjugates comprising at least one binding entity that specifically binds to a target and uses thereof
WO2022182891A1 (en) 2021-02-25 2022-09-01 Lyell Immunopharma, Inc. Ror1 targeting chimeric antigen receptor
US11453716B2 (en) 2016-05-20 2022-09-27 Harpoon Therapeutics, Inc. Single domain serum albumin binding protein
WO2022215011A1 (en) 2021-04-07 2022-10-13 Novartis Ag USES OF ANTI-TGFβ ANTIBODIES AND OTHER THERAPEUTIC AGENTS FOR THE TREATMENT OF PROLIFERATIVE DISEASES
US11472890B2 (en) 2019-03-01 2022-10-18 Xencor, Inc. Heterodimeric antibodies that bind ENPP3 and CD3
WO2022229853A1 (en) 2021-04-27 2022-11-03 Novartis Ag Viral vector production system
US11505595B2 (en) 2018-04-18 2022-11-22 Xencor, Inc. TIM-3 targeted heterodimeric fusion proteins containing IL-15/IL-15RA Fc-fusion proteins and TIM-3 antigen binding domains
WO2022243846A1 (en) 2021-05-18 2022-11-24 Novartis Ag Combination therapies
WO2022256359A1 (en) 2021-06-01 2022-12-08 Amgen Inc. Accelerated method of making lyophilized protein formualtions
US11524991B2 (en) 2018-04-18 2022-12-13 Xencor, Inc. PD-1 targeted heterodimeric fusion proteins containing IL-15/IL-15Ra Fc-fusion proteins and PD-1 antigen binding domains and uses thereof
US11535668B2 (en) 2017-02-28 2022-12-27 Harpoon Therapeutics, Inc. Inducible monovalent antigen binding protein
WO2023278585A1 (en) 2021-06-30 2023-01-05 Amgen Inc. Method of reconstituting lyophilized formulation
WO2023021477A1 (en) 2021-08-20 2023-02-23 Novartis Ag Methods of making chimeric antigen receptor–expressing cells
US11591401B2 (en) 2020-08-19 2023-02-28 Xencor, Inc. Anti-CD28 compositions
US11607453B2 (en) 2017-05-12 2023-03-21 Harpoon Therapeutics, Inc. Mesothelin binding proteins
WO2023044483A2 (en) 2021-09-20 2023-03-23 Voyager Therapeutics, Inc. Compositions and methods for the treatment of her2 positive cancer
US11623958B2 (en) 2016-05-20 2023-04-11 Harpoon Therapeutics, Inc. Single chain variable fragment CD3 binding proteins
WO2023092004A1 (en) 2021-11-17 2023-05-25 Voyager Therapeutics, Inc. Compositions and methods for the treatment of tau-related disorders
US11718679B2 (en) 2017-10-31 2023-08-08 Compass Therapeutics Llc CD137 antibodies and PD-1 antagonists and uses thereof
WO2023150778A1 (en) 2022-02-07 2023-08-10 Visterra, Inc. Anti-idiotype antibody molecules and uses thereof
US11739144B2 (en) 2021-03-09 2023-08-29 Xencor, Inc. Heterodimeric antibodies that bind CD3 and CLDN6
US11752207B2 (en) 2017-07-11 2023-09-12 Compass Therapeutics Llc Agonist antibodies that bind human CD137 and uses thereof
EP4268831A2 (en) 2018-09-12 2023-11-01 Fred Hutchinson Cancer Center Reducing cd33 expression to selectively protect therapeutic cells
WO2023209568A1 (en) 2022-04-26 2023-11-02 Novartis Ag Multispecific antibodies targeting il-13 and il-18
WO2023212559A1 (en) 2022-04-26 2023-11-02 Amgen Inc. Lyophilization method
WO2023214325A1 (en) 2022-05-05 2023-11-09 Novartis Ag Pyrazolopyrimidine derivatives and uses thereof as tet2 inhibitors
WO2023220695A2 (en) 2022-05-13 2023-11-16 Voyager Therapeutics, Inc. Compositions and methods for the treatment of her2 positive cancer
US11859012B2 (en) 2021-03-10 2024-01-02 Xencor, Inc. Heterodimeric antibodies that bind CD3 and GPC3
WO2024030976A2 (en) 2022-08-03 2024-02-08 Voyager Therapeutics, Inc. Compositions and methods for crossing the blood brain barrier
EP4324518A2 (en) 2014-01-31 2024-02-21 Novartis AG Antibody molecules to tim-3 and uses thereof
WO2024040020A1 (en) 2022-08-15 2024-02-22 Absci Corporation Quantitative affinity activity specific cell enrichment
US11919956B2 (en) 2020-05-14 2024-03-05 Xencor, Inc. Heterodimeric antibodies that bind prostate specific membrane antigen (PSMA) and CD3
WO2024056809A1 (en) 2022-09-15 2024-03-21 Novartis Ag Treatment of autoimmune disorders using chimeric antigen receptor therapy

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2007006057A (en) * 2004-11-18 2007-12-10 Imclone Systems Inc Antibodies against vascular endothelial growth factor receptor-1.
US9493578B2 (en) 2009-09-02 2016-11-15 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
AU2010343049A1 (en) * 2009-12-29 2012-07-19 Emergent Product Development Seattle, Llc Polypeptide heterodimers and uses thereof
EP3713961A2 (en) 2017-11-20 2020-09-30 Compass Therapeutics LLC Cd137 antibodies and tumor antigen-targeting antibodies and uses thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
HUP0600225A3 (en) * 2001-06-13 2010-01-28 Genmab As Human monoclonal antibodies to epidermal growth factor receptor (egfr)

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP1786918A4 *

Cited By (438)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8853366B2 (en) 2001-01-17 2014-10-07 Emergent Product Development Seattle, Llc Binding domain-immunoglobulin fusion proteins
US9718883B2 (en) 2003-09-10 2017-08-01 Amgen Fremont Inc. Antibodies to M-CSF
US10280219B2 (en) 2003-09-10 2019-05-07 Amgen Fremont Inc. Antibodies to M-CSF
US8062886B2 (en) 2003-11-12 2011-11-22 Schering Corporation Plasmid system for multigene expression
US7811562B2 (en) 2004-12-03 2010-10-12 Schering Corporation Biomarkers for pre-selection of patients for anti-IGF1R therapy
EP2100618A2 (en) 2005-06-17 2009-09-16 Imclone LLC PDGFR-alpha antagonists for treatment of metastatic bone cancer
EP2100614A2 (en) 2005-06-17 2009-09-16 Imclone LLC PDGFR-alpha antagonists for treatment of metastatic bone cancer
EP2505205A1 (en) 2005-06-17 2012-10-03 Imclone LLC Anti-PDGFR Alpha Antibodies
US10143748B2 (en) 2005-07-25 2018-12-04 Aptevo Research And Development Llc B-cell reduction using CD37-specific and CD20-specific binding molecules
US10307481B2 (en) 2005-07-25 2019-06-04 Aptevo Research And Development Llc CD37 immunotherapeutics and uses thereof
EP1928506A4 (en) * 2005-08-19 2009-10-21 Abbott Lab Dual variable domain immunoglobin and uses thereof
EP2520588A1 (en) * 2005-08-19 2012-11-07 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
EP2500357A3 (en) * 2005-08-19 2012-10-24 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
EP2500359A3 (en) * 2005-08-19 2012-10-17 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
EP2500358A3 (en) * 2005-08-19 2012-10-17 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
EP2495257A3 (en) * 2005-08-19 2012-10-17 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
EP2500356A3 (en) * 2005-08-19 2012-10-24 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
EP2500353A3 (en) * 2005-08-19 2012-10-10 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
EP2500354A3 (en) * 2005-08-19 2012-10-24 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
EP2500355A3 (en) * 2005-08-19 2012-10-24 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
EP1928506A2 (en) * 2005-08-19 2008-06-11 Abbott Laboratories Dual variable domain immunoglobin and uses thereof
EP2500352A1 (en) * 2005-08-19 2012-09-19 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US7972600B2 (en) 2006-02-03 2011-07-05 Imclone Llc IGF-IR antagonists as adjuvants for treatment of prostate cancer
WO2007092453A2 (en) 2006-02-03 2007-08-16 Imclone Systems Incorporated Igf-ir antagonists as adjuvants for treatment of prostate cancer
US8409577B2 (en) 2006-06-12 2013-04-02 Emergent Product Development Seattle, Llc Single chain multivalent binding proteins with effector function
EP2056869A2 (en) * 2006-08-18 2009-05-13 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
EP2056869A4 (en) * 2006-08-18 2009-10-21 Abbott Lab Dual variable domain immunoglobulin and uses thereof
US11149077B2 (en) 2006-11-22 2021-10-19 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins for tyrosine kinases receptors, including IGF-IR
US8470332B2 (en) 2006-11-22 2013-06-25 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins for tyrosine kinases receptors, including IGF-IR
US10221232B2 (en) 2006-11-22 2019-03-05 Bristol-Myers Squibb Company Methods of treating cancer by administering IGF-IR binding molecules
US7740850B2 (en) 2007-04-17 2010-06-22 ImClone, LLC PDGFRβ-specific antibodies
EP2626371A1 (en) 2007-07-31 2013-08-14 MedImmune, LLC Multispecific epitope binding proteins and uses thereof
US8975382B2 (en) 2007-11-27 2015-03-10 Ablynx N.V. Amino acid sequences directed against HER2 and polypeptides comprising the same for the treatment of cancers and/or tumors
US9969805B2 (en) 2007-11-27 2018-05-15 Ablynx N.V. Amino acid sequences directed against HER2 and polypeptides comprising the same for the treatment of cancers and/or tumors
WO2009068630A1 (en) * 2007-11-27 2009-06-04 Ablynx N.V. Immunoglobulin constructs
US9266967B2 (en) 2007-12-21 2016-02-23 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US10138293B2 (en) 2007-12-21 2018-11-27 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US10927163B2 (en) 2007-12-21 2021-02-23 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US9234028B2 (en) 2008-02-14 2016-01-12 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins that bind EGFR
US9920108B2 (en) 2008-02-14 2018-03-20 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins that bind EGFR
US10781247B2 (en) 2008-02-14 2020-09-22 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins that bind EGFR
US8524244B2 (en) 2008-02-14 2013-09-03 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins that bind EGFR
US9441034B2 (en) 2008-03-27 2016-09-13 Zymogenetics, Inc. Compositions and methods for inhibiting PDGFRβ and VEGF-A
US9708390B2 (en) 2008-03-27 2017-07-18 Zymogenetics, Inc. Compositions and methods for inhibiting PDGFRbeta and VEGF-A
US9101609B2 (en) 2008-04-11 2015-08-11 Emergent Product Development Seattle, Llc CD37 immunotherapeutic and combination with bifunctional chemotherapeutic thereof
US9029508B2 (en) 2008-04-29 2015-05-12 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9902762B2 (en) 2008-05-22 2018-02-27 Bristol-Myers Squibb Company Multivalent fibronectin based scaffold domain proteins
US8221765B2 (en) 2008-05-22 2012-07-17 Bristol-Myers Squibb Company Multivalent fibronectin based scaffold domain proteins
US10774130B2 (en) 2008-05-22 2020-09-15 Bristol-Myers Squibb Company Method of treating cancer by administering multivalent fibronectin based scaffold domain proteins
US8728483B2 (en) 2008-05-22 2014-05-20 Bristol-Myers Squibb Company Multivalent fibronectin based scaffold domain proteins
US9109026B2 (en) 2008-06-03 2015-08-18 Abbvie, Inc. Dual variable domain immunoglobulins and uses thereof
US9035027B2 (en) 2008-06-03 2015-05-19 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US8822645B2 (en) 2008-07-08 2014-09-02 Abbvie Inc. Prostaglandin E2 dual variable domain immunoglobulins and uses thereof
CN102149825A (en) * 2008-07-08 2011-08-10 雅培制药有限公司 Prostaglandin E2 dual variable domain immunoglobulins and uses thereof
WO2010028798A1 (en) * 2008-09-10 2010-03-18 F. Hoffmann-La Roche Ag Multivalent antibodies
WO2010028797A1 (en) * 2008-09-10 2010-03-18 F. Hoffmann-La Roche Ag Multivalent antibodies
WO2010028795A1 (en) * 2008-09-10 2010-03-18 F. Hoffmann-La Roche Ag Multivalent antibodies
WO2010034441A1 (en) * 2008-09-26 2010-04-01 F. Hoffmann-La Roche Ag Bispecific anti-egfr/anti-igf-1r antibodies
US8703130B2 (en) 2008-10-08 2014-04-22 Hoffmann-La Roche, Inc. Bispecific anti-VEGF/anti-ANG-2 antibodies
US9708396B2 (en) 2008-10-08 2017-07-18 Hoffmann-La Roche Inc. Bispecific anti-VEGF/anti-ANG-2 antibodies
US8268314B2 (en) 2008-10-08 2012-09-18 Hoffmann-La Roche Inc. Bispecific anti-VEGF/anti-ANG-2 antibodies
EP2792687A1 (en) 2008-10-08 2014-10-22 F. Hoffmann-La Roche AG Bispecific anti-VEGF/anti-ANG-2 antibodies
EP2781526A1 (en) 2008-10-08 2014-09-24 F. Hoffmann-La Roche AG Bispecific anti-VEGF/anti-ANG-2 antibodies
US8343501B2 (en) 2008-11-24 2013-01-01 Bristol-Myers Squibb Company Bispecific EGFR/IGFIR binding molecules
US10954286B2 (en) 2008-11-24 2021-03-23 Bristol-Myers Squibb Company Bispecific EGFR/IGFIR binding molecules
US9017655B2 (en) 2008-11-24 2015-04-28 Bristol-Myers Squibb Company Bispecific EGFR/IGFIR binding molecules
US10183987B2 (en) 2008-11-24 2019-01-22 Bristol-Myers Squibb Company Polynucleotides encoding bispecific EGFR/IGF-IR binding molecules
US9771411B2 (en) 2008-11-24 2017-09-26 Bristol-Myers Squibb Company Method of treating cancer by administering EGFR and EGFR/IGFIR binding molecules
CN102438702A (en) * 2009-02-10 2012-05-02 第一三共株式会社 Anti-mst1r antibodies and uses thereof
US9403909B2 (en) 2009-02-10 2016-08-02 Daiichi Sankyo Company, Limited Anti-MST1R antibodies and uses thereof
CN102438702B (en) * 2009-02-10 2016-03-02 第一三共株式会社 Anti-MST1R antibody and uses thereof
WO2010093055A1 (en) * 2009-02-10 2010-08-19 Daiichi Sankyo Company, Limited Anti-mst1r antibodies and uses thereof
WO2010112194A1 (en) 2009-04-02 2010-10-07 F. Hoffmann-La Roche Ag Antigen-binding polypeptides and multispecific antibodies comprising them
WO2010112193A1 (en) 2009-04-02 2010-10-07 Roche Glycart Ag Multispecific antibodies comprising full length antibodies and single chain fab fragments
US9890204B2 (en) 2009-04-07 2018-02-13 Hoffmann-La Roche Inc. Trivalent, bispecific antibodies
WO2010115552A1 (en) 2009-04-07 2010-10-14 Roche Glycart Ag Bispecific anti-erbb-3/anti-c-met antibodies
WO2010115553A1 (en) 2009-04-07 2010-10-14 Roche Glycart Ag Bispecific anti-erbb-2/anti-c-met antibodies
WO2010115551A1 (en) 2009-04-07 2010-10-14 Roche Glycart Ag Bispecific anti-erbb-1/anti-c-met antibodies
WO2010115589A1 (en) 2009-04-07 2010-10-14 Roche Glycart Ag Trivalent, bispecific antibodies
WO2010136482A1 (en) * 2009-05-28 2010-12-02 Glaxo Group Limited Antigen-binding proteins
US20120177651A1 (en) * 2009-05-28 2012-07-12 Neil James Clarke Antigen-binding proteins
US10640555B2 (en) 2009-06-16 2020-05-05 Hoffmann-La Roche Inc. Bispecific antigen binding proteins
US9676845B2 (en) 2009-06-16 2017-06-13 Hoffmann-La Roche, Inc. Bispecific antigen binding proteins
US11673945B2 (en) 2009-06-16 2023-06-13 Hoffmann-La Roche Inc. Bispecific antigen binding proteins
US8586714B2 (en) 2009-09-01 2013-11-19 Abbvie, Inc. Dual variable domain immunoglobulins and uses thereof
US9994646B2 (en) 2009-09-16 2018-06-12 Genentech, Inc. Coiled coil and/or tether containing protein complexes and uses thereof
US8716450B2 (en) 2009-10-15 2014-05-06 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US8722855B2 (en) 2009-10-28 2014-05-13 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
WO2011117329A1 (en) 2010-03-26 2011-09-29 F. Hoffmann-La Roche Ag Bispecific, bivalent anti-vegf/anti-ang-2 antibodies
US10106600B2 (en) 2010-03-26 2018-10-23 Roche Glycart Ag Bispecific antibodies
WO2011117330A1 (en) 2010-03-26 2011-09-29 Roche Glycart Ag Bispecific antibodies
US10221438B2 (en) 2010-05-03 2019-03-05 Bristol-Myers Squibb Company Serum albumin binding molecules
US9540424B2 (en) 2010-05-03 2017-01-10 Bristol-Myers Squibb Company Serum albumin binding molecules
US10934572B2 (en) 2010-05-03 2021-03-02 Bristol-Myers Squibb Company Serum albumin binding molecules
US8969289B2 (en) 2010-05-03 2015-03-03 Bristol-Myers Squibb Company Serum albumin binding molecules
EP3345926A1 (en) 2010-05-06 2018-07-11 Novartis AG Compositions and methods of use for therapeutic low density lipoprotein-related protein 6 (lrp6) antibodies
EP4234698A2 (en) 2010-05-06 2023-08-30 Novartis AG Compositions and methods of use for therapeutic low density lipoprotein-related protein 6 (lrp6) antibodies
WO2011138391A1 (en) 2010-05-06 2011-11-10 Novartis Ag Compositions and methods of use for therapeutic low density lipoprotein - related protein 6 (lrp6) multivalent antibodies
WO2011138392A1 (en) 2010-05-06 2011-11-10 Novartis Ag Compositions and methods of use for therapeutic low density lipoprotein -related protein 6 (lrp6) antibodies
US9562089B2 (en) 2010-05-26 2017-02-07 Bristol-Myers Squibb Company Fibronectin based scaffold proteins having improved stability
US10273286B2 (en) 2010-05-26 2019-04-30 Bristol-Myers Squibb Company Fibronectin based scaffold proteins having improved stability
US11161893B2 (en) 2010-05-26 2021-11-02 Bristol-Myers Squibb Company Fibronectin based scaffold proteins having improved stability
US8829164B2 (en) 2010-07-06 2014-09-09 Aveo Pharmaceuticals, Inc. Anti-ron antibodies
WO2012006341A2 (en) 2010-07-06 2012-01-12 Aveo Pharmaceuticals, Inc. Anti-ron antibodies
US8603478B2 (en) 2010-07-06 2013-12-10 Aveo Pharmaceuticals, Inc. Anti-RON antibodies
WO2012006341A3 (en) * 2010-07-06 2012-03-22 Aveo Pharmaceuticals, Inc. Anti-ron antibodies
US9605061B2 (en) 2010-07-29 2017-03-28 Xencor, Inc. Antibodies with modified isoelectric points
US9493560B2 (en) 2010-08-03 2016-11-15 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US8735546B2 (en) 2010-08-03 2014-05-27 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9879095B2 (en) 2010-08-24 2018-01-30 Hoffman-La Roche Inc. Bispecific antibodies comprising a disulfide stabilized-Fv fragment
WO2012025530A1 (en) 2010-08-24 2012-03-01 F. Hoffmann-La Roche Ag Bispecific antibodies comprising a disulfide stabilized - fv fragment
US9046513B2 (en) 2010-08-26 2015-06-02 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
WO2012069557A1 (en) * 2010-11-24 2012-05-31 Glaxo Group Limited Multispecific antigen binding proteins targeting hgf
US9982036B2 (en) 2011-02-28 2018-05-29 Hoffmann-La Roche Inc. Dual FC antigen binding proteins
US10611825B2 (en) 2011-02-28 2020-04-07 Hoffmann La-Roche Inc. Monovalent antigen binding proteins
US10793621B2 (en) 2011-02-28 2020-10-06 Hoffmann-La Roche Inc. Nucleic acid encoding dual Fc antigen binding proteins
US8094026B1 (en) 2011-05-02 2012-01-10 Robert M Green Organized retail crime detection security system and method
US10851178B2 (en) 2011-10-10 2020-12-01 Xencor, Inc. Heterodimeric human IgG1 polypeptides with isoelectric point modifications
WO2013067355A1 (en) 2011-11-04 2013-05-10 Novartis Ag Low density lipoprotein-related protein 6 (lrp6) - half life extender constructs
EP3252075A1 (en) 2011-11-04 2017-12-06 Novartis AG Low density lipoprotein-related protein 6 (lrp6) - half life extender constructs
EP3290442A1 (en) 2011-11-04 2018-03-07 Novartis AG Low density lipoprotein-related protein 6 (lrp6) half-life extender constructs
US9120870B2 (en) 2011-12-30 2015-09-01 Abbvie Inc. Dual specific binding proteins directed against IL-13 and IL-17
US9688758B2 (en) 2012-02-10 2017-06-27 Genentech, Inc. Single-chain antibodies and other heteromultimers
US9714292B2 (en) 2012-04-05 2017-07-25 Hoffmann-La Roche Inc. Bispecific antibodies against human TWEAK and human IL17 and uses thereof
WO2013150043A1 (en) 2012-04-05 2013-10-10 F. Hoffmann-La Roche Ag Bispecific antibodies against human tweak and human il17 and uses thereof
US11421022B2 (en) 2012-06-27 2022-08-23 Hoffmann-La Roche Inc. Method for making antibody Fc-region conjugates comprising at least one binding entity that specifically binds to a target and uses thereof
US11407836B2 (en) 2012-06-27 2022-08-09 Hoffmann-La Roche Inc. Method for selection and production of tailor-made highly selective and multi-specific targeting entities containing at least two different binding entities and uses thereof
US10106612B2 (en) 2012-06-27 2018-10-23 Hoffmann-La Roche Inc. Method for selection and production of tailor-made highly selective and multi-specific targeting entities containing at least two different binding entities and uses thereof
US10517945B2 (en) 2012-07-04 2019-12-31 Hoffman-La Roche Inc. Covalently linked antigen-antibody conjugates
US9765153B2 (en) 2012-07-04 2017-09-19 Hoffmann-La Roche Inc. Anti-biotin antibodies and methods of use
WO2014006124A1 (en) 2012-07-04 2014-01-09 F. Hoffmann-La Roche Ag Covalently linked antigen-antibody conjugates
US9925272B2 (en) 2012-07-04 2018-03-27 Hoffmann-La Roche Inc. Anti-theophylline antibodies and methods of use
US10683345B2 (en) 2012-07-13 2020-06-16 Roche Glycart Ag Bispecific anti-VEGF/anti-ANG-2 antibodies and their use in the treatment of ocular vascular diseases
US9944720B2 (en) 2012-11-01 2018-04-17 Abbvie Inc. Anti-DLL4/VEGF dual variable domain immunoglobulin and uses thereof
US9045551B2 (en) 2012-11-01 2015-06-02 Abbvie Inc. Anti-DLL4/VEGF dual variable domain immunoglobulin and uses thereof
US9163093B2 (en) 2012-11-01 2015-10-20 Abbvie Inc. Anti-DLL4/VEGF dual variable domain immunoglobulin and uses thereof
WO2014099997A1 (en) 2012-12-18 2014-06-26 Novartis Ag Compositions and methods that utilize a peptide tag that binds to hyaluronan
US11718667B2 (en) 2013-01-14 2023-08-08 Xencor, Inc. Optimized antibody variable regions
US10472427B2 (en) 2013-01-14 2019-11-12 Xencor, Inc. Heterodimeric proteins
US9701759B2 (en) 2013-01-14 2017-07-11 Xencor, Inc. Heterodimeric proteins
US10738132B2 (en) 2013-01-14 2020-08-11 Xencor, Inc. Heterodimeric proteins
US10738133B2 (en) 2013-01-14 2020-08-11 Xencor, Inc. Heterodimeric proteins
US10487155B2 (en) 2013-01-14 2019-11-26 Xencor, Inc. Heterodimeric proteins
US9650446B2 (en) 2013-01-14 2017-05-16 Xencor, Inc. Heterodimeric proteins
US11053316B2 (en) 2013-01-14 2021-07-06 Xencor, Inc. Optimized antibody variable regions
US11634506B2 (en) 2013-01-14 2023-04-25 Xencor, Inc. Heterodimeric proteins
US10131710B2 (en) 2013-01-14 2018-11-20 Xencor, Inc. Optimized antibody variable regions
US9738722B2 (en) 2013-01-15 2017-08-22 Xencor, Inc. Rapid clearance of antigen complexes using novel antibodies
WO2015198217A2 (en) 2013-02-08 2015-12-30 Novartis Ag Compositions and methods for long-acting antibodies targeting il-17
EP3744736A1 (en) 2013-02-20 2020-12-02 Novartis AG Effective targeting of primary human leukemia using anti-cd123 chimeric antigen receptor engineered t cells
EP3626741A1 (en) 2013-02-20 2020-03-25 The Trustees Of The University Of Pennsylvania Treatment of cancer using humanized anti-egfrviii chimeric antigen receptor
WO2014130657A1 (en) 2013-02-20 2014-08-28 The Trustees Of The University Of Pennsylvania Treatment of cancer using humanized anti-egfrviii chimeric antigen receptor
WO2014130635A1 (en) 2013-02-20 2014-08-28 Novartis Ag Effective targeting of primary human leukemia using anti-cd123 chimeric antigen receptor engineered t cells
US10968276B2 (en) 2013-03-12 2021-04-06 Xencor, Inc. Optimized anti-CD3 variable regions
US9499855B2 (en) 2013-03-14 2016-11-22 Elwha Llc Compositions, methods, and computer systems related to making and administering modified T cells
US20140274801A1 (en) * 2013-03-14 2014-09-18 Elwha Llc Compositions, methods, and computer systems related to making and administering modified t cells
US9662354B2 (en) 2013-03-14 2017-05-30 Elwha Llc Compositions, methods, and computer systems related to making and administering modified T cells
US9587237B2 (en) 2013-03-14 2017-03-07 Elwha Llc Compositions, methods, and computer systems related to making and administering modified T cells
US8987418B2 (en) 2013-03-15 2015-03-24 Abbvie Inc. Dual specific binding proteins directed against IL-1β and/or IL-17
US11814423B2 (en) 2013-03-15 2023-11-14 Xencor, Inc. Heterodimeric proteins
US10287364B2 (en) 2013-03-15 2019-05-14 Xencor, Inc. Heterodimeric proteins
US10858417B2 (en) 2013-03-15 2020-12-08 Xencor, Inc. Heterodimeric proteins
US10544187B2 (en) 2013-03-15 2020-01-28 Xencor, Inc. Targeting regulatory T cells with heterodimeric proteins
EP3623380A1 (en) 2013-03-15 2020-03-18 Michael C. Milone Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
US9605084B2 (en) 2013-03-15 2017-03-28 Xencor, Inc. Heterodimeric proteins
US10106624B2 (en) 2013-03-15 2018-10-23 Xencor, Inc. Heterodimeric proteins
US10519242B2 (en) 2013-03-15 2019-12-31 Xencor, Inc. Targeting regulatory T cells with heterodimeric proteins
US9062108B2 (en) 2013-03-15 2015-06-23 Abbvie Inc. Dual specific binding proteins directed against IL-1 and/or IL-17
US11299554B2 (en) 2013-03-15 2022-04-12 Xencor, Inc. Heterodimeric proteins
EP4067382A1 (en) 2013-03-16 2022-10-05 Novartis AG Treatment of cancer using humanized anti-cd19 chimeric antigen receptor
WO2014153270A1 (en) 2013-03-16 2014-09-25 Novartis Ag Treatment of cancer using humanized anti-cd19 chimeric antigen receptor
EP3539986A1 (en) 2013-03-16 2019-09-18 Novartis AG Treatment of cancer using humanized anti-cd19 chimeric antigen receptor
US9567403B2 (en) 2013-08-06 2017-02-14 Bio-Thera Solutions, Ltd. Bispecific antibodies which bind EGFR and VEGF
US10323099B2 (en) 2013-10-11 2019-06-18 Hoffmann-La Roche Inc. Multispecific domain exchanged common variable light chain antibodies
EP4026909A1 (en) 2013-12-19 2022-07-13 Novartis AG Human mesothelin chimeric antigen receptors and uses thereof
WO2015090230A1 (en) 2013-12-19 2015-06-25 Novartis Ag Human mesothelin chimeric antigen receptors and uses thereof
US10465000B2 (en) 2013-12-20 2019-11-05 Hoffmann-La Roche Inc. Humanized anti-Tau(pS422) antibodies and methods of use
WO2015090229A1 (en) 2013-12-20 2015-06-25 Novartis Ag Regulatable chimeric antigen receptor
US10407511B2 (en) 2014-01-03 2019-09-10 Hoffmann-La Roche Inc. Covalently linked helicar-anti-helicar antibody conjugates and uses thereof
WO2015101586A1 (en) 2014-01-03 2015-07-09 F. Hoffmann-La Roche Ag Bispecific anti-hapten/anti-blood brain barrier receptor antibodies, complexes thereof and their use as blood brain barrier shuttles
US10561737B2 (en) 2014-01-03 2020-02-18 Hoffmann-La Roche Inc. Bispecific anti-hapten/anti-blood brain barrier receptor antibodies, complexes thereof and their use as blood brain barrier shuttles
WO2015101589A1 (en) 2014-01-03 2015-07-09 F. Hoffmann-La Roche Ag Covalently linked polypeptide toxin-antibody conjugates
WO2015101587A1 (en) 2014-01-03 2015-07-09 F. Hoffmann-La Roche Ag Covalently linked helicar-anti-helicar antibody conjugates and uses thereof
US10519249B2 (en) 2014-01-03 2019-12-31 Hoffmann-La Roche Inc. Covalently linked polypeptide toxin-antibody conjugates
WO2015112626A1 (en) 2014-01-21 2015-07-30 June Carl H Enhanced antigen presenting ability of car t cells by co-introduction of costimulatory molecules
EP4303229A2 (en) 2014-01-21 2024-01-10 Novartis AG Enhanced antigen presenting ability of car t cells by co-introduction of costimulatory molecules
EP3514179A1 (en) 2014-01-24 2019-07-24 Dana-Farber Cancer Institute, Inc. Antibody molecules to pd-1 and uses thereof
EP4324518A2 (en) 2014-01-31 2024-02-21 Novartis AG Antibody molecules to tim-3 and uses thereof
EP3660050A1 (en) 2014-03-14 2020-06-03 Novartis AG Antibody molecules to lag-3 and uses thereof
WO2015138920A1 (en) 2014-03-14 2015-09-17 Novartis Ag Antibody molecules to lag-3 and uses thereof
WO2015142675A2 (en) 2014-03-15 2015-09-24 Novartis Ag Treatment of cancer using chimeric antigen receptor
WO2015142661A1 (en) 2014-03-15 2015-09-24 Novartis Ag Regulatable chimeric antigen receptor
EP3811970A1 (en) 2014-03-15 2021-04-28 Novartis AG Regulatable chimeric antigen receptor
US10442851B2 (en) 2014-03-20 2019-10-15 Bristol-Myers Squibb Company Serum albumin-binding fibronectin type III domains
US11203630B2 (en) 2014-03-20 2021-12-21 Bristol-Myers Squibb Company Serum albumin-binding fibronectin type III domains
US11814441B2 (en) * 2014-03-21 2023-11-14 X-Body, Inc. Bi-specific antigen-binding polypeptides
US20190248918A1 (en) * 2014-03-21 2019-08-15 X-Body, Inc. Bi-specific antigen-binding polypeptides
US11840579B2 (en) 2014-03-28 2023-12-12 Xencor, Inc. Bispecific antibodies that bind to CD38 and CD3
US10858451B2 (en) 2014-03-28 2020-12-08 Xencor, Inc. Bispecific antibodies that bind to CD38 and CD3
US9822186B2 (en) 2014-03-28 2017-11-21 Xencor, Inc. Bispecific antibodies that bind to CD38 and CD3
WO2015157252A1 (en) 2014-04-07 2015-10-15 BROGDON, Jennifer Treatment of cancer using anti-cd19 chimeric antigen receptor
EP3888674A1 (en) 2014-04-07 2021-10-06 Novartis AG Treatment of cancer using anti-cd19 chimeric antigen receptor
WO2015198243A2 (en) 2014-06-25 2015-12-30 Novartis Ag Compositions and methods for long acting proteins
WO2015198240A2 (en) 2014-06-25 2015-12-30 Novartis Ag Compositions and methods for long acting proteins
US10251952B2 (en) 2014-06-26 2019-04-09 Hoffmann-La Roche Inc. Humanized anti-tau(pS422) antibody brain shuttles and use thereof
US9884921B2 (en) 2014-07-01 2018-02-06 Pfizer Inc. Bispecific heterodimeric diabodies and uses thereof
WO2016014565A2 (en) 2014-07-21 2016-01-28 Novartis Ag Treatment of cancer using humanized anti-bcma chimeric antigen receptor
WO2016014553A1 (en) 2014-07-21 2016-01-28 Novartis Ag Sortase synthesized chimeric antigen receptors
WO2016014530A1 (en) 2014-07-21 2016-01-28 Novartis Ag Combinations of low, immune enhancing. doses of mtor inhibitors and cars
EP3722316A1 (en) 2014-07-21 2020-10-14 Novartis AG Treatment of cancer using a cd33 chimeric antigen receptor
EP4205749A1 (en) 2014-07-31 2023-07-05 Novartis AG Subset-optimized chimeric antigen receptor-containing cells
EP3660042A1 (en) 2014-07-31 2020-06-03 Novartis AG Subset-optimized chimeric antigen receptor-containing t-cells
WO2016025880A1 (en) 2014-08-14 2016-02-18 Novartis Ag Treatment of cancer using gfr alpha-4 chimeric antigen receptor
EP3712171A1 (en) 2014-08-19 2020-09-23 Novartis AG Treatment of cancer using a cd123 chimeric antigen receptor
WO2016044605A1 (en) 2014-09-17 2016-03-24 Beatty, Gregory Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
EP3967709A1 (en) 2014-09-17 2022-03-16 Novartis AG Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
WO2016061142A1 (en) 2014-10-14 2016-04-21 Novartis Ag Antibody molecules to pd-l1 and uses thereof
EP4245376A2 (en) 2014-10-14 2023-09-20 Novartis AG Antibody molecules to pd-l1 and uses thereof
US9856327B2 (en) 2014-11-26 2018-01-02 Xencor, Inc. Heterodimeric antibodies to CD3 X CD123
US11859011B2 (en) 2014-11-26 2024-01-02 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US11673972B2 (en) 2014-11-26 2023-06-13 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US11945880B2 (en) 2014-11-26 2024-04-02 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US9850320B2 (en) 2014-11-26 2017-12-26 Xencor, Inc. Heterodimeric antibodies to CD3 X CD20
US11225528B2 (en) 2014-11-26 2022-01-18 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US10526417B2 (en) 2014-11-26 2020-01-07 Xencor, Inc. Heterodimeric antibodies that bind CD3 and CD38
US10913803B2 (en) 2014-11-26 2021-02-09 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US10889653B2 (en) 2014-11-26 2021-01-12 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US11352442B2 (en) 2014-11-26 2022-06-07 Xencor, Inc. Heterodimeric antibodies that bind CD3 and CD38
US11111315B2 (en) 2014-11-26 2021-09-07 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US10259887B2 (en) 2014-11-26 2019-04-16 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
WO2016090034A2 (en) 2014-12-03 2016-06-09 Novartis Ag Methods for b cell preconditioning in car therapy
US10633457B2 (en) 2014-12-03 2020-04-28 Hoffmann-La Roche Inc. Multispecific antibodies
US10093733B2 (en) 2014-12-11 2018-10-09 Abbvie Inc. LRP-8 binding dual variable domain immunoglobulin proteins
US10428155B2 (en) 2014-12-22 2019-10-01 Xencor, Inc. Trispecific antibodies
EP4036109A2 (en) 2014-12-29 2022-08-03 Novartis AG Methods of making chimeric antigen receptor-expressing cells
US11014989B2 (en) 2015-01-26 2021-05-25 Cellectis Anti-CLL1 specific single-chain chimeric antigen receptors (scCARs) for cancer immunotherapy
WO2016126608A1 (en) 2015-02-02 2016-08-11 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
US11091548B2 (en) 2015-03-05 2021-08-17 Xencor, Inc. Modulation of T cells with bispecific antibodies and Fc fusions
US10227411B2 (en) 2015-03-05 2019-03-12 Xencor, Inc. Modulation of T cells with bispecific antibodies and FC fusions
EP4056588A1 (en) 2015-04-08 2022-09-14 Novartis AG Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car)- expressing cell
WO2016164731A2 (en) 2015-04-08 2016-10-13 Novartis Ag Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car) - expressing cell
US10787500B2 (en) 2015-04-10 2020-09-29 Adimab, Llc Methods for purifying heterodimeric multispecific antibodies from parental homodimeric antibody species
WO2016164708A1 (en) 2015-04-10 2016-10-13 Adimab, Llc Methods for purifying heterodimeric multispecific antibodies from parental homodimeric antibody species
WO2016168595A1 (en) 2015-04-17 2016-10-20 Barrett David Maxwell Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
EP4234685A2 (en) 2015-04-17 2023-08-30 Novartis AG Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
WO2016172583A1 (en) 2015-04-23 2016-10-27 Novartis Ag Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
EP3770168A1 (en) 2015-05-18 2021-01-27 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
US10442849B2 (en) 2015-05-18 2019-10-15 Tcr2 Therabeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US11028142B2 (en) 2015-05-18 2021-06-08 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US10358474B2 (en) 2015-05-18 2019-07-23 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
EP3466967A1 (en) 2015-05-18 2019-04-10 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
WO2016187349A1 (en) 2015-05-18 2016-11-24 Tcr2, Inc. Compositions and methods for tcr reprogramming using fusion proteins
US10358473B2 (en) 2015-05-18 2019-07-23 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US10954311B2 (en) 2015-05-21 2021-03-23 Harpoon Therapeutics, Inc. Trispecific binding proteins and methods of use
US9840554B2 (en) 2015-06-15 2017-12-12 Abbvie Inc. Antibodies against platelet-derived growth factor (PDGF)
US10822402B2 (en) 2015-06-24 2020-11-03 Hoffmann-La Roche Inc. Humanized anti-tau(pS422) antibodies and methods of use
WO2017015427A1 (en) 2015-07-21 2017-01-26 Novartis Ag Methods for improving the efficacy and expansion of immune cells
EP3964528A1 (en) 2015-07-29 2022-03-09 Novartis AG Combination therapies comprising antibody molecules to lag-3
WO2017019894A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to lag-3
WO2017019897A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to tim-3
EP3878465A1 (en) 2015-07-29 2021-09-15 Novartis AG Combination therapies comprising antibody molecules to tim-3
WO2017027392A1 (en) 2015-08-07 2017-02-16 Novartis Ag Treatment of cancer using chimeric cd3 receptor proteins
US11352426B2 (en) 2015-09-21 2022-06-07 Aptevo Research And Development Llc CD3 binding polypeptides
US11434275B2 (en) 2015-09-23 2022-09-06 Bristol-Myers Squibb Company Fast-off rate serum albumin binding fibronectin type III domains
US10766946B2 (en) 2015-09-23 2020-09-08 Bristol-Myers Squibb Company Fast-off rate serum albumin binding fibronectin type III domains
WO2017091786A1 (en) 2015-11-23 2017-06-01 Novartis Ag Optimized lentiviral transfer vectors and uses thereof
US11623957B2 (en) 2015-12-07 2023-04-11 Xencor, Inc. Heterodimeric antibodies that bind CD3 and PSMA
US10227410B2 (en) 2015-12-07 2019-03-12 Xencor, Inc. Heterodimeric antibodies that bind CD3 and PSMA
WO2017106656A1 (en) 2015-12-17 2017-06-22 Novartis Ag Antibody molecules to pd-1 and uses thereof
WO2017106810A2 (en) 2015-12-17 2017-06-22 Novartis Ag Combination of c-met inhibitor with antibody molecule to pd-1 and uses thereof
EP4219689A2 (en) 2015-12-30 2023-08-02 Novartis AG Immune effector cell therapies with enhanced efficacy
WO2017114497A1 (en) 2015-12-30 2017-07-06 Novartis Ag Immune effector cell therapies with enhanced efficacy
EP3851457A1 (en) 2016-01-21 2021-07-21 Novartis AG Multispecific molecules targeting cll-1
WO2017125897A1 (en) 2016-01-21 2017-07-27 Novartis Ag Multispecific molecules targeting cll-1
CN105481981A (en) * 2016-01-27 2016-04-13 中国人民解放军第二军医大学 Bispecific antibody targeting VEGF and application thereof
WO2017149515A1 (en) 2016-03-04 2017-09-08 Novartis Ag Cells expressing multiple chimeric antigen receptor (car) molecules and uses therefore
WO2017165683A1 (en) 2016-03-23 2017-09-28 Novartis Ag Cell secreted minibodies and uses thereof
WO2017172981A2 (en) 2016-03-29 2017-10-05 University Of Southern California Chimeric antigen receptors targeting cancer
EP4219721A2 (en) 2016-04-15 2023-08-02 Novartis AG Compositions and methods for selective protein expression
WO2017181119A2 (en) 2016-04-15 2017-10-19 Novartis Ag Compositions and methods for selective protein expression
US11453716B2 (en) 2016-05-20 2022-09-27 Harpoon Therapeutics, Inc. Single domain serum albumin binding protein
US11623958B2 (en) 2016-05-20 2023-04-11 Harpoon Therapeutics, Inc. Single chain variable fragment CD3 binding proteins
WO2017210617A2 (en) 2016-06-02 2017-12-07 Porter, David, L. Therapeutic regimens for chimeric antigen receptor (car)- expressing cells
US11492407B2 (en) 2016-06-14 2022-11-08 Xencor, Inc. Bispecific checkpoint inhibitor antibodies
US11236170B2 (en) 2016-06-14 2022-02-01 Xencor, Inc. Bispecific checkpoint inhibitor antibodies
US10787518B2 (en) 2016-06-14 2020-09-29 Xencor, Inc. Bispecific checkpoint inhibitor antibodies
US11225521B2 (en) 2016-06-28 2022-01-18 Xencor, Inc. Heterodimeric antibodies that bind somatostatin receptor 2
US10316088B2 (en) 2016-06-28 2019-06-11 Xencor, Inc. Heterodimeric antibodies that bind somatostatin receptor 2
WO2018013918A2 (en) 2016-07-15 2018-01-18 Novartis Ag Treatment and prevention of cytokine release syndrome using a chimeric antigen receptor in combination with a kinase inhibitor
WO2018023025A1 (en) 2016-07-28 2018-02-01 Novartis Ag Combination therapies of chimeric antigen receptors adn pd-1 inhibitors
WO2018026819A2 (en) 2016-08-01 2018-02-08 Novartis Ag Treatment of cancer using a chimeric antigen receptor in combination with an inhibitor of a pro-m2 macrophage molecule
US11242376B2 (en) 2016-08-02 2022-02-08 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
WO2018026953A1 (en) 2016-08-02 2018-02-08 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
US10793632B2 (en) 2016-08-30 2020-10-06 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
US11085021B2 (en) 2016-10-07 2021-08-10 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
WO2018067992A1 (en) 2016-10-07 2018-04-12 Novartis Ag Chimeric antigen receptors for the treatment of cancer
EP3848392A1 (en) 2016-10-07 2021-07-14 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
US10208285B2 (en) 2016-10-07 2019-02-19 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
WO2018067993A1 (en) 2016-10-07 2018-04-12 TCR2 Therapeutics Inc. Compositions and methods for t-cell receptors reprogramming using fusion proteins
US11377638B2 (en) 2016-10-07 2022-07-05 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US10550185B2 (en) 2016-10-14 2020-02-04 Xencor, Inc. Bispecific heterodimeric fusion proteins containing IL-15-IL-15Rα Fc-fusion proteins and PD-1 antibody fragments
US10501543B2 (en) 2016-10-14 2019-12-10 Xencor, Inc. IL15/IL15Rα heterodimeric Fc-fusion proteins
US11851491B2 (en) 2016-11-22 2023-12-26 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
WO2018098365A2 (en) 2016-11-22 2018-05-31 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
US10849973B2 (en) 2016-11-23 2020-12-01 Harpoon Therapeutics, Inc. Prostate specific membrane antigen binding protein
US10844134B2 (en) 2016-11-23 2020-11-24 Harpoon Therapeutics, Inc. PSMA targeting trispecific proteins and methods of use
WO2018111340A1 (en) 2016-12-16 2018-06-21 Novartis Ag Methods for determining potency and proliferative function of chimeric antigen receptor (car)-t cells
EP4043485A1 (en) 2017-01-26 2022-08-17 Novartis AG Cd28 compositions and methods for chimeric antigen receptor therapy
WO2018140725A1 (en) 2017-01-26 2018-08-02 Novartis Ag Cd28 compositions and methods for chimeric antigen receptor therapy
WO2018144535A1 (en) 2017-01-31 2018-08-09 Novartis Ag Treatment of cancer using chimeric t cell receptor proteins having multiple specificities
WO2018141910A1 (en) 2017-02-02 2018-08-09 Amgen Research (Munich) Gmbh Low ph pharmaceutical composition comprising t cell engaging antibody constructs
WO2018160731A1 (en) 2017-02-28 2018-09-07 Novartis Ag Shp inhibitor compositions and uses for chimeric antigen receptor therapy
US11535668B2 (en) 2017-02-28 2022-12-27 Harpoon Therapeutics, Inc. Inducible monovalent antigen binding protein
WO2018201051A1 (en) 2017-04-28 2018-11-01 Novartis Ag Bcma-targeting agent, and combination therapy with a gamma secretase inhibitor
WO2018201056A1 (en) 2017-04-28 2018-11-01 Novartis Ag Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
US10730954B2 (en) 2017-05-12 2020-08-04 Harpoon Therapeutics, Inc. MSLN targeting trispecific proteins and methods of use
US11607453B2 (en) 2017-05-12 2023-03-21 Harpoon Therapeutics, Inc. Mesothelin binding proteins
WO2018232020A1 (en) 2017-06-13 2018-12-20 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
WO2018237157A1 (en) 2017-06-22 2018-12-27 Novartis Ag Antibody molecules to cd73 and uses thereof
WO2019006007A1 (en) 2017-06-27 2019-01-03 Novartis Ag Dosage regimens for anti-tim-3 antibodies and uses thereof
US11084863B2 (en) 2017-06-30 2021-08-10 Xencor, Inc. Targeted heterodimeric Fc fusion proteins containing IL-15 IL-15alpha and antigen binding domains
US11752207B2 (en) 2017-07-11 2023-09-12 Compass Therapeutics Llc Agonist antibodies that bind human CD137 and uses thereof
WO2019018730A1 (en) 2017-07-20 2019-01-24 Novartis Ag Dosage regimens of anti-lag-3 antibodies and uses thereof
US10927180B2 (en) 2017-10-13 2021-02-23 Harpoon Therapeutics, Inc. B cell maturation antigen binding proteins
US11136403B2 (en) 2017-10-13 2021-10-05 Harpoon Therapeutics, Inc. Trispecific proteins and methods of use
WO2019079569A1 (en) 2017-10-18 2019-04-25 Novartis Ag Compositions and methods for selective protein degradation
WO2019084288A1 (en) 2017-10-25 2019-05-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
US11718679B2 (en) 2017-10-31 2023-08-08 Compass Therapeutics Llc CD137 antibodies and PD-1 antagonists and uses thereof
WO2019089798A1 (en) 2017-10-31 2019-05-09 Novartis Ag Anti-car compositions and methods
US11312770B2 (en) 2017-11-08 2022-04-26 Xencor, Inc. Bispecific and monospecific antibodies using novel anti-PD-1 sequences
US10981992B2 (en) 2017-11-08 2021-04-20 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
WO2019099838A1 (en) 2017-11-16 2019-05-23 Novartis Ag Combination therapies
US11319355B2 (en) 2017-12-19 2022-05-03 Xencor, Inc. Engineered IL-2 Fc fusion proteins
WO2019152660A1 (en) 2018-01-31 2019-08-08 Novartis Ag Combination therapy using a chimeric antigen receptor
EP4015525A2 (en) 2018-03-23 2022-06-22 GammaDelta Therapeutics Limited Lymphocytes expressing heterologous targeting constructs
US10982006B2 (en) 2018-04-04 2021-04-20 Xencor, Inc. Heterodimeric antibodies that bind fibroblast activation protein
WO2019200229A1 (en) 2018-04-13 2019-10-17 Novartis Ag Dosage regimens for anti-pd-l1 antibodies and uses thereof
US11524991B2 (en) 2018-04-18 2022-12-13 Xencor, Inc. PD-1 targeted heterodimeric fusion proteins containing IL-15/IL-15Ra Fc-fusion proteins and PD-1 antigen binding domains and uses thereof
US11505595B2 (en) 2018-04-18 2022-11-22 Xencor, Inc. TIM-3 targeted heterodimeric fusion proteins containing IL-15/IL-15RA Fc-fusion proteins and TIM-3 antigen binding domains
WO2019210153A1 (en) 2018-04-27 2019-10-31 Novartis Ag Car t cell therapies with enhanced efficacy
WO2019213282A1 (en) 2018-05-01 2019-11-07 Novartis Ag Biomarkers for evaluating car-t cells to predict clinical outcome
WO2019226617A1 (en) 2018-05-21 2019-11-28 Compass Therapeutics Llc Compositions and methods for enhancing the killing of target cells by nk cells
WO2019226658A1 (en) 2018-05-21 2019-11-28 Compass Therapeutics Llc Multispecific antigen-binding compositions and methods of use
WO2019227003A1 (en) 2018-05-25 2019-11-28 Novartis Ag Combination therapy with chimeric antigen receptor (car) therapies
WO2019232244A2 (en) 2018-05-31 2019-12-05 Novartis Ag Antibody molecules to cd73 and uses thereof
WO2019237035A1 (en) 2018-06-08 2019-12-12 Intellia Therapeutics, Inc. Compositions and methods for immunooncology
WO2019241426A1 (en) 2018-06-13 2019-12-19 Novartis Ag Bcma chimeric antigen receptors and uses thereof
WO2019246293A2 (en) 2018-06-19 2019-12-26 Atarga, Llc Antibody molecules to complement component 5 and uses thereof
WO2020010079A2 (en) 2018-07-02 2020-01-09 Amgen Inc. Anti-steap1 antigen-binding protein
US11530274B2 (en) 2018-07-02 2022-12-20 Amgen Inc. Anti-STEAP1 antigen-binding protein
WO2020012337A1 (en) 2018-07-10 2020-01-16 Novartis Ag 3-(5-amino-1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and their use in the treatment of i karos family zinc finger 2 (ikzf2)-dependent diseases
WO2020021465A1 (en) 2018-07-25 2020-01-30 Advanced Accelerator Applications (Italy) S.R.L. Method of treatment of neuroendocrine tumors
WO2020047501A1 (en) 2018-08-30 2020-03-05 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
WO2020047452A2 (en) 2018-08-31 2020-03-05 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2020047449A2 (en) 2018-08-31 2020-03-05 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
EP4268831A2 (en) 2018-09-12 2023-11-01 Fred Hutchinson Cancer Center Reducing cd33 expression to selectively protect therapeutic cells
US11807692B2 (en) 2018-09-25 2023-11-07 Harpoon Therapeutics, Inc. DLL3 binding proteins and methods of use
US10815311B2 (en) 2018-09-25 2020-10-27 Harpoon Therapeutics, Inc. DLL3 binding proteins and methods of use
WO2020069405A1 (en) 2018-09-28 2020-04-02 Novartis Ag Cd22 chimeric antigen receptor (car) therapies
WO2020069409A1 (en) 2018-09-28 2020-04-02 Novartis Ag Cd19 chimeric antigen receptor (car) and cd22 car combination therapies
US11358999B2 (en) 2018-10-03 2022-06-14 Xencor, Inc. IL-12 heterodimeric Fc-fusion proteins
US11046769B2 (en) 2018-11-13 2021-06-29 Compass Therapeutics Llc Multispecific binding constructs against checkpoint molecules and uses thereof
WO2020128972A1 (en) 2018-12-20 2020-06-25 Novartis Ag Dosing regimen and pharmaceutical combination comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
WO2020128898A1 (en) 2018-12-20 2020-06-25 Novartis Ag Pharmaceutical combinations
WO2020165834A1 (en) 2019-02-15 2020-08-20 Novartis Ag Substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2020165868A1 (en) 2019-02-15 2020-08-20 Perkinelmer Cellular Technologies Germany Gmbh Low-power microscope-objective pre-scan and high-power microscope-objective scan in x,y and z-direction for imaging objects such as cells using a microscope
WO2020165833A1 (en) 2019-02-15 2020-08-20 Novartis Ag 3-(1-oxo-5-(piperidin-4-yl)isoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2020172553A1 (en) 2019-02-22 2020-08-27 Novartis Ag Combination therapies of egfrviii chimeric antigen receptors and pd-1 inhibitors
WO2020176397A1 (en) 2019-02-25 2020-09-03 Novartis Ag Mesoporous silica particles compositions for viral delivery
US11472890B2 (en) 2019-03-01 2022-10-18 Xencor, Inc. Heterodimeric antibodies that bind ENPP3 and CD3
US11220535B2 (en) 2019-03-15 2022-01-11 Cartesian Therapeutics, Inc. Anti-BCMA chimeric antigen receptors
US10934337B2 (en) 2019-03-15 2021-03-02 Cartesian Therapeutics, Inc. Anti-BCMA chimeric antigen receptors
WO2020205523A1 (en) 2019-03-29 2020-10-08 Atarga, Llc Anti fgf23 antibody
WO2020210678A1 (en) 2019-04-12 2020-10-15 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2020219742A1 (en) 2019-04-24 2020-10-29 Novartis Ag Compositions and methods for selective protein degradation
WO2021003432A1 (en) 2019-07-02 2021-01-07 Fred Hutchinson Cancer Research Center Recombinant ad35 vectors and related gene therapy improvements
WO2021035170A1 (en) 2019-08-21 2021-02-25 Precision Biosciences, Inc. Compositions and methods for tcr reprogramming using fusion proteins
WO2021079188A1 (en) 2019-10-21 2021-04-29 Novartis Ag Combination therapies with venetoclax and tim-3 inhibitors
WO2021079195A1 (en) 2019-10-21 2021-04-29 Novartis Ag Tim-3 inhibitors and uses thereof
WO2021108661A2 (en) 2019-11-26 2021-06-03 Novartis Ag Chimeric antigen receptors and uses thereof
WO2021123902A1 (en) 2019-12-20 2021-06-24 Novartis Ag Combination of anti tim-3 antibody mbg453 and anti tgf-beta antibody nis793, with or without decitabine or the anti pd-1 antibody spartalizumab, for treating myelofibrosis and myelodysplastic syndrome
WO2021123996A1 (en) 2019-12-20 2021-06-24 Novartis Ag Uses of anti-tgf-beta antibodies and checkpoint inhibitors for the treatment of proliferative diseases
WO2021144657A1 (en) 2020-01-17 2021-07-22 Novartis Ag Combination comprising a tim-3 inhibitor and a hypomethylating agent for use in treating myelodysplastic syndrome or chronic myelomonocytic leukemia
WO2021146636A1 (en) 2020-01-17 2021-07-22 Becton, Dickinson And Company Methods and compositions for single cell secretomics
WO2021163618A1 (en) 2020-02-14 2021-08-19 Novartis Ag Method of predicting response to chimeric antigen receptor therapy
US11180563B2 (en) 2020-02-21 2021-11-23 Harpoon Therapeutics, Inc. FLT3 binding proteins and methods of use
WO2021173995A2 (en) 2020-02-27 2021-09-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2021173985A2 (en) 2020-02-27 2021-09-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2021169977A1 (en) 2020-02-28 2021-09-02 南京北恒生物科技有限公司 Novel chimeric antigen receptor and use thereof
WO2021222347A1 (en) 2020-04-29 2021-11-04 Amgen Inc. Pharmaceutical formulation
WO2021222355A1 (en) 2020-04-29 2021-11-04 Amgen Inc. Pharmaceutical formulation
WO2021231655A1 (en) 2020-05-12 2021-11-18 Lyell Immunopharma, Inc. Chimeric antigen receptor spacers
US11919956B2 (en) 2020-05-14 2024-03-05 Xencor, Inc. Heterodimeric antibodies that bind prostate specific membrane antigen (PSMA) and CD3
WO2021238877A1 (en) 2020-05-27 2021-12-02 南京北恒生物科技有限公司 Engineered immune cell and use thereof
WO2021249462A1 (en) 2020-06-11 2021-12-16 南京北恒生物科技有限公司 Engineered immune cell expressing nk inhibitory molecule and use thereof
WO2021252920A1 (en) 2020-06-11 2021-12-16 Novartis Ag Zbtb32 inhibitors and uses thereof
WO2021260528A1 (en) 2020-06-23 2021-12-30 Novartis Ag Dosing regimen comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
WO2022011358A1 (en) * 2020-07-10 2022-01-13 Biomolecular Holdings Llc Tetrahedral antibodies
WO2022012591A1 (en) 2020-07-15 2022-01-20 南京北恒生物科技有限公司 Engineered immune cell for allotransplantation
WO2022013787A1 (en) 2020-07-16 2022-01-20 Novartis Ag Anti-betacellulin antibodies, fragments thereof, and multi-specific binding molecules
WO2022026592A2 (en) 2020-07-28 2022-02-03 Celltas Bio, Inc. Antibody molecules to coronavirus and uses thereof
WO2022022745A1 (en) 2020-07-31 2022-02-03 南京北恒生物科技有限公司 Novel co-stimulatory domain and uses thereof
WO2022029573A1 (en) 2020-08-03 2022-02-10 Novartis Ag Heteroaryl substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2022033537A1 (en) 2020-08-13 2022-02-17 南京北恒生物科技有限公司 Engineered immune cell and use thereof
US11591401B2 (en) 2020-08-19 2023-02-28 Xencor, Inc. Anti-CD28 compositions
US11919958B2 (en) 2020-08-19 2024-03-05 Xencor, Inc. Anti-CD28 compositions
WO2022040586A2 (en) 2020-08-21 2022-02-24 Novartis Ag Compositions and methods for in vivo generation of car expressing cells
WO2022046651A1 (en) 2020-08-24 2022-03-03 Amgen Inc. Pharmaceutical formulation comprising a bite, bispecific antibody, and methionine
WO2022043558A1 (en) 2020-08-31 2022-03-03 Advanced Accelerator Applications International Sa Method of treating psma-expressing cancers
WO2022043557A1 (en) 2020-08-31 2022-03-03 Advanced Accelerator Applications International Sa Method of treating psma-expressing cancers
WO2022052981A1 (en) 2020-09-10 2022-03-17 南京北恒生物科技有限公司 Chimeric antigen receptor comprising novel co-stimulatory domain and use thereof
WO2022095802A1 (en) 2020-11-03 2022-05-12 南京北恒生物科技有限公司 Chimeric antigen receptor targeting cd7 and use thereof
WO2022095803A1 (en) 2020-11-03 2022-05-12 南京北恒生物科技有限公司 Cd7-targeting humanized antibody and use thereof
WO2022097065A2 (en) 2020-11-06 2022-05-12 Novartis Ag ANTIBODY Fc VARIANTS
WO2022104061A1 (en) 2020-11-13 2022-05-19 Novartis Ag Combination therapies with chimeric antigen receptor (car)-expressing cells
WO2022105826A1 (en) 2020-11-23 2022-05-27 南京北恒生物科技有限公司 Nkg2a-targeting antibody and use thereof
WO2022152168A1 (en) 2021-01-12 2022-07-21 南京北恒生物科技有限公司 Ror1-targeting antibody and use thereof
WO2022162569A1 (en) 2021-01-29 2022-08-04 Novartis Ag Dosage regimes for anti-cd73 and anti-entpd2 antibodies and uses thereof
WO2022166365A1 (en) 2021-02-03 2022-08-11 南京北恒生物科技有限公司 Novel chimeric antigen receptor and use thereof
WO2022182891A1 (en) 2021-02-25 2022-09-01 Lyell Immunopharma, Inc. Ror1 targeting chimeric antigen receptor
US11739144B2 (en) 2021-03-09 2023-08-29 Xencor, Inc. Heterodimeric antibodies that bind CD3 and CLDN6
US11859012B2 (en) 2021-03-10 2024-01-02 Xencor, Inc. Heterodimeric antibodies that bind CD3 and GPC3
WO2022215011A1 (en) 2021-04-07 2022-10-13 Novartis Ag USES OF ANTI-TGFβ ANTIBODIES AND OTHER THERAPEUTIC AGENTS FOR THE TREATMENT OF PROLIFERATIVE DISEASES
WO2022229853A1 (en) 2021-04-27 2022-11-03 Novartis Ag Viral vector production system
WO2022243846A1 (en) 2021-05-18 2022-11-24 Novartis Ag Combination therapies
WO2022256359A1 (en) 2021-06-01 2022-12-08 Amgen Inc. Accelerated method of making lyophilized protein formualtions
WO2023278585A1 (en) 2021-06-30 2023-01-05 Amgen Inc. Method of reconstituting lyophilized formulation
WO2023021477A1 (en) 2021-08-20 2023-02-23 Novartis Ag Methods of making chimeric antigen receptor–expressing cells
WO2023044483A2 (en) 2021-09-20 2023-03-23 Voyager Therapeutics, Inc. Compositions and methods for the treatment of her2 positive cancer
WO2023092004A1 (en) 2021-11-17 2023-05-25 Voyager Therapeutics, Inc. Compositions and methods for the treatment of tau-related disorders
WO2023150778A1 (en) 2022-02-07 2023-08-10 Visterra, Inc. Anti-idiotype antibody molecules and uses thereof
WO2023209568A1 (en) 2022-04-26 2023-11-02 Novartis Ag Multispecific antibodies targeting il-13 and il-18
WO2023212559A1 (en) 2022-04-26 2023-11-02 Amgen Inc. Lyophilization method
WO2023214325A1 (en) 2022-05-05 2023-11-09 Novartis Ag Pyrazolopyrimidine derivatives and uses thereof as tet2 inhibitors
WO2023220695A2 (en) 2022-05-13 2023-11-16 Voyager Therapeutics, Inc. Compositions and methods for the treatment of her2 positive cancer
WO2024030976A2 (en) 2022-08-03 2024-02-08 Voyager Therapeutics, Inc. Compositions and methods for crossing the blood brain barrier
WO2024040020A1 (en) 2022-08-15 2024-02-22 Absci Corporation Quantitative affinity activity specific cell enrichment
WO2024056809A1 (en) 2022-09-15 2024-03-21 Novartis Ag Treatment of autoimmune disorders using chimeric antigen receptor therapy

Also Published As

Publication number Publication date
EP1786918A2 (en) 2007-05-23
WO2006020258A3 (en) 2006-10-12
EP1786918A4 (en) 2009-02-11
JP2008512352A (en) 2008-04-24

Similar Documents

Publication Publication Date Title
EP1786918A2 (en) Novel tetravalent bispecific antibody
AU2007215013A1 (en) Functional antibodies
US20040259156A1 (en) Bispecific immunoglobulin-like antigen binding proteins and method of production
CN113260379B (en) Protease cleavable bispecific antibodies and uses thereof
KR20200104328A (en) Bispecific antigen binding molecule
US20040242851A1 (en) Bispecific antibodies that bind to vegf receptors
KR20180129684A (en) Anti-Human Interleukin-2 Antibodies and Uses thereof
WO2004003211A1 (en) Bispecific antibodies that bind to vegf receptors
EP4155319A1 (en) 4-1bb binding protein and application thereof
CN112500491B (en) Bispecific antibody for specifically neutralizing helper T cell TGF-beta signal, pharmaceutical composition and application thereof
IL299232A (en) Bispecific antibody and use thereof
TW202202530A (en) Binding protein having h2l2 and hcab structures
US20230051266A1 (en) Anti-bcma/anti-4-1bb bispecific antibodies and uses thereof
CA3228682A1 (en) Anti-b7-h4 antibody, and preparation method therefor and use thereof
AU2022318255A1 (en) Combinations of antigen binding molecules
JP2024516098A (en) Multispecific antibodies targeting BCMA
AU2020271467A1 (en) Antibodies against programmed death-ligand 1 and uses thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2007521716

Country of ref document: JP

NENP Non-entry into the national phase in:

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2005773142

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2005773142

Country of ref document: EP