WO2014012085A2 - Bispecific asymmetric heterodimers comprising anti-cd3 constructs - Google Patents

Bispecific asymmetric heterodimers comprising anti-cd3 constructs Download PDF

Info

Publication number
WO2014012085A2
WO2014012085A2 PCT/US2013/050411 US2013050411W WO2014012085A2 WO 2014012085 A2 WO2014012085 A2 WO 2014012085A2 US 2013050411 W US2013050411 W US 2013050411W WO 2014012085 A2 WO2014012085 A2 WO 2014012085A2
Authority
WO
WIPO (PCT)
Prior art keywords
cell
polypeptide
construct
heteromultimer
multispecific heteromultimer
Prior art date
Application number
PCT/US2013/050411
Other languages
French (fr)
Other versions
WO2014012085A9 (en
WO2014012085A3 (en
Inventor
Surjit Bhimarao Dixit
Thomas SPRETER VON KREUDENSTEIN
Gordon Yiu Kon NG
Original Assignee
Zymeworks Inc.
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
Priority to KR20157003872A priority Critical patent/KR20150036606A/en
Priority to RU2015102193A priority patent/RU2650868C2/en
Priority to EP13816697.0A priority patent/EP2872170A4/en
Priority to CN201380037369.3A priority patent/CN104640562A/en
Priority to CA2878843A priority patent/CA2878843A1/en
Priority to BR112015000798A priority patent/BR112015000798A2/en
Application filed by Zymeworks Inc. filed Critical Zymeworks Inc.
Priority to AU2013289883A priority patent/AU2013289883B2/en
Priority to JP2015521877A priority patent/JP2015528003A/en
Publication of WO2014012085A2 publication Critical patent/WO2014012085A2/en
Publication of WO2014012085A3 publication Critical patent/WO2014012085A3/en
Publication of WO2014012085A9 publication Critical patent/WO2014012085A9/en
Priority to HK15108247.7A priority patent/HK1207575A1/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
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2809Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/14Ectoparasiticides, e.g. scabicides
    • 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
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • 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
    • C07K16/2887Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
    • 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
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/35Valency
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/524CH2 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/526CH3 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/528CH4 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • 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
    • C07K2317/64Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising a combination of variable region and constant region components
    • 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
    • C07K2317/71Decreased effector function due to an Fc-modification
    • 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
    • C07K2317/72Increased effector function due to an Fc-modification
    • 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
    • C07K2317/74Inducing cell proliferation
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/31Fusion polypeptide fusions, other than Fc, for prolonged plasma life, e.g. albumin

Definitions

  • the field of the invention is the rational design of multispecific scaffolds comprising a CD3 binding domain for custom development of biotherapeutics.
  • bispecific antibodies and other fused multispecific therapeutics exhibit dual or multiple target specificities and an opportunity to create drugs with novel modes of action.
  • the development of such multivalent and multispecific therapeutic proteins with favorable pharmacokinetics and functional activity has been a challenge.
  • T cells thymus derived cells
  • B cells bone marrow derived cells
  • T cells exhibit immunological specificity and are directly involved in cell-mediated immune responses (such as graft rejection).
  • T cells act against or in response to a variety of foreign structures (antigens). In many instances these foreign antigens are expressed on host cells as a result of infection. However, foreign antigens can also come from the host having been altered by neoplasia or infection.
  • T cell activation is a complex phenomenon that depends on the participation of a variety of cell surface molecules expressed on the responding WSGF
  • the antigen-specific T cell population For example, the antigen-specific T cell population.
  • the antigen-specific T cell population For example, the antigen-specific T cell population.
  • CD3 gamma and delta
  • multispecific heteromultimers comprising a CD3 binding domain.
  • a bispecific asymmetric heterodimer comprising anti-CD3 constructs.
  • isolated multispecific heteromultimer constructs comprising: a first polypeptide construct comprising a first heavy chain polypeptide and a CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second heavy chain polypeptide which is different from said first heavy chain polypeptide, and an antigen binding polypeptide construct that binds to a target antigen on at least one B cell; wherein: at least one of said CD3 binding polypeptide construct and said antigen binding polypeptide construct comprises a single chain Fv region; wherein said multispecific heteromultimer construct simultaneously engages said at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell; and said first and second heavy chain polypeptides form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said hetero
  • said stable mammalian cell is transfected at least a first DNA
  • the first or second polypeptide construct is devoid of at least one of immunoglobulin light chain, and immunoglobulin first constant (CH1 ) region.
  • the isolated heteromultimer constructs described herein wherein the heterodimer Fc region comprises a variant CH2 domain or hinge comprising amino acid modifications that prevents functionally effective binding to all the Fcgamma receptors.
  • the isolated multispecific heteromultimer described herein wherein wherien said variant CH2 domain or hinge comprising amino acid modification also prevents functionally effective binding to complement proteins (C1 q complex).
  • the heterodimer Fc region comprises a variant CH2 domain or hinge comprising amino acid modifications that enhance binding to the FcyRllb receptor.
  • heteromultimer construct comprising: a first polypeptide construct comprising a first heavy chain polypeptide and a CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second heavy chain polypeptide which is different from said first heavy chain polypeptide, and an antigen binding polypeptide construct that binds to a target antigen on at least one B cell;
  • At least one of said CD3 binding polypeptide construct and said antigen binding polypeptide construct optionally comprises a single chain Fv region; said first and second heavy chain polypeptides form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc, wherein: said heterodimeric Fc is formed with stability at least comparable to a
  • said expression product comprises greater than 70% of said multispecific heteromultimer, and less than 10% monomers or homodimers of said first or second polypeptide constructs; and said multispecific heteromultimer construct binds said at least one B cell with a valency greater than one, and said multispecific heteromultimer simultaneously engages said at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell.
  • said multispecific heteromultimer construct binds said at least one B cell with a valency of two.
  • an isolated multispecific heteromultimer construct comprising: a first polypeptide construct comprising a first heavy chain polypeptide and a CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second heavy chain polypeptide which is different from said first heavy chain polypeptide, and a steric modulator construct which exhibits negligible receptor binding; wherein: said multispecific heteromultimer construct simultaneously engages at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell; and said first and second heavy chain polypeptides form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc, wherein: said heterodimeric Fc is formed with stability at least comparable to a native homodimeric Fc, and said heterodimeric F
  • first polypeptide construct comprising a first heavy chain polypeptide and a CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second heavy chain polypeptide which is different from said first heavy chain polypeptide, and wherein said second polypeptide construct does not comprise an antigen binding polypeptide construct; wherein: said multispecific heteromultimer construct simultaneously engages at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell; and said first and second heavy chain polypeptides form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc, wherein: said heterodimeric
  • Fc is formed with stability at least comparable to a native homodimeric Fc, and said heterodimeric Fc is formed with purity such that when said multispecific heteromultimer construct is coexpressed from a stable mammalian cell in an expression product, said expression product comprises at least about 75% of said multispecific heteromultimer, and less than 10% monomers or
  • heterodimer Fc region comprises a variant CH2 domain comprising amino acid modifications to promote selective binding of a
  • the variant CH2 domain selectively binds Fcgammallb receptor greater than to wild-type CH2 domain. In certain embodiments, the variant CH2 domain selectively binds atleast one of
  • Fcgammallla and Fcgammalla receptor greater than a wild-type CH2 domain.
  • the heterodimer Fc region is formed with a purity greater than about 90%. In certain embodiments, the heterodimer Fc region is formed with a purity of about 95%
  • the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications L351Y, F405A, and Y407V
  • the variant CH3 sequence of the second transporter polypeptide comprises the amino acid modifications T366L, K392M, and T394W
  • the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications L351Y, F405A, and Y407V
  • the variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications T366L, K392L, and T394W.
  • the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications T350V, L351Y, F405A, and Y407V
  • the variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications T350V, T366L, K392M, and T394W
  • the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications T350V, L351Y, F405A, and Y407V
  • the variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications T350V, T366L, K392L, and T394W.
  • the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications T366L, N390R, K392R, and T394W
  • the variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications L351 Y, S400E, F405A, and Y407V
  • the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications T350V, T366L, N390R, K392R, and T394W
  • the variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications T350V, L351Y, S400E, F405A, and Y407V.
  • heterodimer Fc is aglycosylated.
  • the isolated multispecific heteromultimer described herein wherein the antigen binding polypeptide construct that binds to a target antigen on at least one B cell comprises at least one target antigen binding domain derived from an antibody, a fibronectin, an affibody, anticalin, cysteine knot protein, DARPin, avimer, Kunitz domain or variant or derivative thereof.
  • said antibody is a heavy chain antibody devoid of light chains.
  • said antigen binding polypeptide construct comprises at least one CD19 binding domain.
  • said antigen binding polypeptide construct comprises at least one CD20 binding domain.
  • an isolated multispecific heteromultimer construct comprising: a first polypeptide construct comprising a first transporter polypeptide fused to at least one CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second transporter polypeptide which is different from said first transporter polypeptide, fused to at least one antigen binding polypeptide construct that binds to a target antigen on at least one B cell;
  • first and second transporter polypeptides are derived from a protein by segmentation of said protein, each transporter polypeptide comprising an amino acid sequence with at least 90% identity to a segment of said protein, and wherein said transporter polypeptides self-assemble to form a quasi-native structure of said monomeric protein.
  • each transporter polypeptide is an albumin derivative.
  • said albumin is human serum albumin.
  • least one transporter polypeptide is an allo-
  • isolated multispecific heteromultimer constructs comprising: a first polypeptide construct comprising a first transporter polypeptide fused to at least one CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second transporter polypeptide which is different from said first transporter polypeptide, fused to at least one antigen binding polypeptide construct that binds to a target antigen on at least one B cell;
  • first and second transporter polypeptides are obtained by segmentation of albumin, and each transporter polypeptide comprising an amino acid sequence with at least 90% identity to a segment of albumin such that said transporter polypeptides self-assemble to form quasi-native albumin, and wherein said first cargo polypeptide does not have any binding domain present in said second cargo polypeptide.
  • a heteromultimer described herein wherein said multispecific heteromultimer construct simultaneously engages said at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell.
  • said antigen binding polypeptide construct that binds to a target antigen on at least one B cell comprises at least one target antigen binding domain derived from an antibody, a fibronectin, an affibody, anticalin, cysteine knot protein, DARPin, avimer, Kunitz domain or variant or derivative thereof.
  • the antigen binding polypeptide construct comprises at least one CD19 binding domain.
  • a multispecific heteromultimer described herein wherein said antigen binding polypeptide construct comprises at least one CD20 binding domain.
  • the at least one CD3 binding polypeptide construct comprises at least one CD3 binding domain derived from a CD3 specific
  • the at least one CD3 binding domain comprises at least one amino acid modification that reduces immunogenicity as compared to a corresponding CD3 binding domain not comprising said modification.
  • the isolated multispecific heteromultimer described herein wherein said at least one CD3 binding domain comprises at least one amino acid modification that increases its stability as measured by T m , as compared to a corresponding CD3 binding domain not comprising said modification.
  • the at least one CD3 binding polypeptide construct comprises at least one CD3 binding domain derived from a CD3 specific antibody is a heavy chain antibody devoid of light chains.
  • the at least one CD3 binding polypeptide construct comprises at least one CD3 binding domain derived from a non-antibody protein scaffold domain.
  • the isolated heteromutlimer construct described herein wherein at least one of said first and second polypeptide constructs further comprises a single-chain Fv polypeptide. In certain embodiments, is provided the isolated heteromutlimer construct described herein, wherein at least one of said first and second polypeptide constructs further comprises a single-chain Fab polypeptide.
  • the isolated heteromutlimer construct described herein where in the CD3 expressing cell is a T-cell.
  • the isolated heteromultimer described herein wherein said heteromultimer binds to the T-cell with sufficient affinity and decorates the T cell at sufficient capacity that induces the T-cell to display B cell killing activity when the T cell and the B cell are bridged.
  • CD3 expressing cell is a human cell.
  • the CD3 expressing cell is a human cell.
  • CD3 expressing cell is a non-human, mammalian cell.
  • the mammalian cell is a primate cell.
  • the primate is a monkey.
  • the CD3 binding polypeptide binds to CD3 constructs across multiple species which include at least one or more of human, rat, mouse and monkey.
  • the at least one B cell is associated with a disease.
  • the disease is a cancer selected from a carcinoma, a sarcoma, leukaemia, lymphoma and glioma.
  • the cancer is at least one of squamous cell carcinoma, adenocarcinoma, transition cell carcinoma, osteosarcoma and soft tissue sarcoma.
  • the at least one B cell is an autoimmune reactive cell that is a lymphoid or myeloid cell.
  • heteromultimer further comprises at least one binding domain that binds at least one of: EpCAM, EGFR, IGFR, HER-2 neu, HER-3, HER-4, PSMA, CEA, MUC-1 (mucin), MUC2, MUC3, MUC4, MUC5, MUC7, CCR4, CCR5, CD19, CD20, CD33, CD30, ganglioside GD3, 9-0-Acetyl-GD3, GM2, Poly SA, GD2, Carboanhydrase IX (MN/CA IX), CD44v6, Sonic Hedgehog (Shh), Wue-1 , Plasma Cell Antigen, (membrane-bound), Melanoma
  • MCSP Chondroitin Sulfate Proteoglycan
  • CCR8 TNF-alpha precursor
  • STEAP mesothelin, A33 Antigen, Prostate Stem Cell Antigen (PSCA), Ly-6; desmoglein 4, E-cadherin neoepitope, Fetal Acetylcholine Receptor, CD25, CA19-9 marker, CA-125 marker and Muellerian Inhibitory Substance (MIS) Receptor type II, sTn (sialylated Tn antigen; TAG-72), FAP (fibroblast activation antigen), endosialin, LG, SAS, EPHA4 CD63, CD3 BsAb immunocytokines TNF, IFNy, IL-2, and TRAIL.
  • MIS Muellerian Inhibitory Substance
  • heteromultimer construct described herein wherein said heteromultimer optionally comprises at least one linker.
  • said at least one linker is a polypeptide comprising from about 1 to about 100 amino acids.
  • multispecific heteromultimer described herein comprising at least a first DNA sequence encoding said first polypeptide construct and at least a second DNA sequence encoding said second polypeptide construct.
  • the method comprising: transfecting at least one mammalian cell with: at least a first DNA sequence encoding said first polypeptide construct and at least a second DNA sequence encoding said second polypeptide construct, such that said at least one first DNA sequence, said at least one second DNA sequence are transfected in said at least one mammalian cell in a pre-determined ratio to generate stable mammalian cells; culturing said stable mammalian cells to produce said expression product comprising said multispecific heteromultimer.
  • said mammalian cell is selected from the group consisting of a VERO, HeLa, HEK, NS0, Chinese Hamster Ovary (CHO), W138, BHK, COS-7, Caco-2 and MDCK cell, and subclasses and variants thereof.
  • composition comprising a
  • multispecific heteromultimer described herein and a suitable excipient.
  • a process for the production of said pharmaceutical composition comprising: culturing a host cell under conditions allowing the expression of a heteromultimer as defined herein; recovering the produced heteromultimer from the culture; and producing the pharmaceutical
  • a proliferative disease a minimal residual cancer, a tumorous disease, an inflammatory disease, an immunological disorder, an autoimmune disease, an infectious disease, viral disease, allergic reactions, parasitic
  • said method comprising administering to a subject in need of such a prevention, treatment or amelioration a pharmaceutical composition described herein.
  • a method of treating cancer in a mammal in need thereof comprising administering to the mammal a composition comprising an effective amount of the pharmaceutical composition described herein, optionally in combination with other pharmaceutically active molecules.
  • said cancer is a solid tumor.
  • said solid tumor is one or more of sarcoma, carcinoma, and lymphoma.
  • the cancer is a hematological cancer.
  • the cancer is one or more of B-cell lymphoma, non-Hodgkin's lymphoma, and leukemia.
  • a method of treating cancer cells comprising providing to said cell a composition comprising a heteromultimer described herein.
  • said heteromultimer is provided in conjugation with another therapeutic agent.
  • composition comprising an effective amount of the pharmaceutical composition described herein.
  • a method of treating a cancer cell regressive after treatment with blinatumomab comprising providing to said cancer cell a composition comprising an effective amount of the pharmaceutical composition described herein.
  • a method of treating an individual suffering from a disease characterized by expression of B cells comprising providing to said individual an effective amount of a composition comprising an effective amount of the pharmaceutical composition described herein.
  • said disease is not responsive to treatment with at least one of an anti-CD19 antibody and an anti-CD20 antibody.
  • said mammal in need thereof, comprising administering to said mammal a composition comprising an effective amount of the pharmaceutical composition provided herein.
  • said autoimmune condition is one or more of multiple sclerosis, rheumatoid arthritis, lupus erytematosus, psoriatic arthritis, psoriasis, vasculitis, uveitis, Crohn's disease, and type 1 diabetes.
  • a method of treating an inflammatory condition in a mammal in need thereof comprising administering to said mammal a composition comprising an effective amount of the pharmaceutical composition comprising an heteromultimer provided herein.
  • kits comprising a heteromultimer as defined herein, and instructions for use thereof.
  • a heteromultimer construct comprising: a first monomer comprising a first transporter polypeptide fused to a first cargo polypeptide that comprises at least one HER2 binding domain; a second monomer comprising a second transporter polypeptide which is different from said first transporter polypeptide, fused to a second cargo polypeptide that comprises at least one HER3 binding domain; wherein said first cargo polypeptide does not have any binding domain present in said second cargo polypeptide; wherein said first and second transporter polypeptide form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimer with stability comparable to a native homodimeric Fc.
  • a heteromultimer construct comprising: a first monomer comprising a first transporter polypeptide fused to a first cargo polypeptide that comprises at least one HER2 binding domain; a second monomer comprising a second transporter polypeptide which is different from said first transporter polypeptide, fused to a second cargo polypeptide polypeptide that comprises at least one HER3 binding domain; wherein said first and second transporter polypeptides are obtained by segmentation of
  • a pharmaceutical composition comprising an insolated multispecific heteromultimer as defined herein, and a suitable excipient. Also provided is a process for the production of such a
  • composition said process comprising: culturing a host cell under conditions allowing the expression of a heteromultimer as defined herein; recovering the produced heteromultimer from the culture; and producing the pharmaceutical composition.
  • host cells comprising nucleic acid encoding a heteromultimer described herein.
  • the nucleic acid encoding the first monomeric protein and the nucleic acid encoding the second monomeric protein are present in a single vector.
  • the nucleic acid encoding the first monomeric protein and the nucleic acid encoding the second monomeric protein are present in separate vectors.
  • kits comprising a heteromultimer as defined herein, and instructions for use thereof.
  • Fig. 1 A-B Fig. 1 A depicts exemplary schematic representation of heteromultimer constructs provided herein.
  • the Immunoglobulin based Anti-CD3 x CD19 constructs demonstrate different aspects of the heteromultimers for instance the cartoon shows the first and second
  • polypeptide constructs wherein the first polypeptide construct comprises a CH3 binding construct (black) and the second polypeptide construct comprises an antigen binding construct (blue).
  • the antigen binding construct is absent or replaced by a steric modulating construct. Also shown is the Fc heteromultimer formed by the variant CH3 regions of the first and
  • FIG. 1 B shows the abil
  • Fig. 2 demonstrates that a heteromultimer described herein (v873) is able to selectively bind and bridge to CD3-expressing Jurkat T cells (lower right panel) and to CD19-expressing Raji B cells (upper right panel).
  • Figure 2 also demonstrates that the one-armed anti-CD3 antibody specifically binds to Jurkat T cells (lower middle panels)and does not cross-react to CD19 expressing B cells, (upper middle panels)and that the one-armed anti-CD19 antibody specifically binds to Raji B cells (upper left panel) and does not cross- react to Jurkat T cells (lower left panel).
  • Fig. 3A-3B Fig. 3A depict the ability of a heteromultimer described herein (v873) to redirect IL-2 activated PBMC to kill target Raji B cells from 3 donors.
  • Fig. 3B demonstrates that a heteromultimer described herein is able to mediate higher redirected T-cell cytotoxicity than a construct lacking the heterodimeric Fc in one of the donors.
  • Fig. 4 shows that heteromultimers described herein are able to bind to CD3-expressing Jurkat T-cells and to CD19-expressing Raji B-cells.
  • Fig. 5A-5B Fig. 5A indicates that at the concentration tested, v1093, which is a heteromultimer described herein, was able to bridge 31 % of total cells, and v873 another heteromultimer construct described herein was able to bridge 25% of total cells.
  • Fig. 5B demonstrates that v1093 is able to bridge Jurkat T cells and Raji B cells to a greater extent than v221 and similar to v891 and v873.
  • Fig. 6 shows antibody therapeutics that can be provided along with a heteromultimer described herein for treatment of certain indications.
  • Fig. 7 shows an SDS-PAGE demonstratin
  • heteromultimer constructs described herein are expressed transiently in
  • CH03E7 cells with a cell viability of > 80 % CH03E7 cells with a cell viability of > 80 %.
  • Fig. 8 shows that a heteromultimer described herein (v873) induces are higher % cytotoxicity to target B cells when compared to negative control human lgG1 (G1 ) when comparing across individual donors.
  • Fig. 9A-9B Fig. 9A shows that human IgG (hlgG) does not bind to Jurkat T-cells and has low level binding to Raji B-cells.
  • Figure 9A also shows that anti-CD19 one arm constructs bind selectively to the Raji B-cells and does not cross-react to Jurkat T cells.
  • Fig. 9B demonstrates FACS assay shows that v873-a heteromultimer described herein, binds selectively to Jurkat T-cells and to Raji B-cells.
  • Fig. 10 shows that v873-a heteromultimer described herein does not bind to the K562 cell line, which does not express CD19 or CD3.
  • Fig. 11 shows that v873-a heteromultimer described herein does not bind to mouse lymphoid cells which does not express CD19 or CD3.
  • Fig 12.A-B show FACS binding curves of heteromultimer constructs described herein (v873, v875) and the construct lacking a heterodimeric Fc (v891 ), to CD3 expressing HPB-ALL and CD3 expressing Jurkat T cells, and to CD19 expressing Raji B cells.
  • Fig. 13A-B Fig. 13A illustrates the FACS binding curves for heteromultimer constructs v875, v1379, v1380, v1381 , and control v891 binding to CD19 expressing Raji cells tested in 0.1 to 300 nM range.
  • Fig. 13B illustrates the FACS binding curve for heteromultimer constructs v875, v1379, v1380, binding to HBP-ALL T cells tested in 0.1 to 300 nM range.
  • Fig. 14 indicates that heteomultimer constructs described herein (v875 and v891 ) facilitate comparable bridging between Raji B-cells and Jurkat T-cells.
  • Use of the control human IgG resulted in 2.5% bridging between Raji and Jurkat cells, while v875 facilitated bridging of 22.9% of total cells, and v891 facilitated bridging of 14.5% of total cells.
  • Fig. 15A-15B Fig. 15A shows the amounl
  • Fig. 15B shows the amount of bridging using a 15:1 ratio of T-cells to B-cells, with heteromultimer concentrations ranging from 0.3 nM to 3 nM.
  • E:T ratios (1 :1 and 15:1 ) tested with v875 resulted in similar total T cell-B cell bridging when expressed as fold over background.
  • Fig 16A-16E depicts the ability of v875, v1379 and v1380 to mediate antibody dependent B cell cytoxicity by redirected CD4+ and CD8+ T cell towards Raji B cells.
  • Fig. 16 B-16E depict representations of the data in Fig. 16A normalized to human IgG, for v875 (Fig. 16B and Fig. 16C), and v1379 and v1380 (Fig. 16D and Fig. 16E), and include % cytotoxicity indicated at each test antibody concentration.
  • Fig. 17A-17B Fig. 17A illustrates that Fc blocking of IL-2 activated PBMC results in a minor (v875) or no (v873) reduction in the % cytotoxicity of target Raji B cells.
  • Figure 17B illustrates that Fc blocking of resting PBMC results in a reduction in the % cytotoxicity of target Raji B cells for v875 and v873.
  • Fig. 18A-18B Fig. 18A illustrates that Fc blocking of IL-2 activated PBMC results in a reduction in the % cytotoxicity of target Raji B cells at all antibody concentrations tested for v875 and v873.
  • Fig. 18B illustrates that Fc blocking of resting PBMC results in a reduction in the % cytotoxicity of target Raji B cells at all antibody concentrations tested for v875 and v873.
  • Fig. 19A-19B Fig. 19A shows that v875 and v873 elicit >30% cytotoxicity to target Raji B cells with IL-2 activiated CD8+ T cells as effectors, and maximal target cell killing is seen at the 3 nM concentration.
  • Fig. 19B shows that v875 and v873 elicit dose dependent (>20%) cytotoxicity to target Raji B cells with resting CD8+ T cells as effectors.
  • Fig. 20A-20B Fig. 20A shows the target Raji B cell cytotoxicty of v875 with IL-2 activated CD4+ and CD8+ T cells.
  • Fig. 20B shows the target Raji B cell cytotoxicty of v875 with resting CD4+ and CD8+ T cells.
  • Fig. 21 shows, relative to untreated media
  • PBMC and total IL-2 activated PBMC activated PBMC.
  • Fig. 22 shows, relative to untreated media and human IgG controls, v875 has a more selective B cell killing by sparing more autologous T cells compared to v873 and v891.
  • Fig. 23A-23D shows the effects of v875 on the viability of CD20+, CD4+, CD8+ subsets in IL-2 activated cell cultures.
  • Fig. 23B shows the effects of v875 on the viability of CD20+, CD4+, CD8+ subsets resting cell cultures.
  • Fig. 23C shows the effects of v1379 and v1380 on the viability of CD20+, CD4+, CD8+ subsets in IL-2 activated cell cultures.
  • Fig. 23D shows shows the effects of v1379 and v1380 on the viability of CD20+, CD4+, CD8+ subsets resting cell cultures.
  • Fig. 24A-24B Results of antibody mediated LDH release in resting effector and Raji B cells shown in Fig. 24A. Results of antibody mediated LDH release in activiated effector are shown in Figure 24B.
  • Fig. 25A-25D Fig 25A illustrates the mediation of ADCC by rituximab and by a heteromultimer described herein with a WT Fc (v875) (ca. 40% max cell lysis).
  • Fig 25B shows that v1379 which is a heteromultimer described herein with a WT Fc can mediate ADCC while v1380, with a
  • FIG. 25C-25D show the results of the CDC assay with v1380 and v1379 (Fig. 25C) and v875 (Fig. 25D) of target Daudi B cells with comparisons to positive control Rituximab.
  • Fig. 26 shows that at 0.3 nM, v875 and v1380 do not induce PBMC proliferation compared to human IgG.
  • the lower panel of Fig. 26 shows the results of the 100 nM antibody concentrations, and shows that v875, v1380 and v891 induce higher cell proliferation relative to human IgG.
  • Fig. 26 (lower panel) also shows that at 100 nM, v875 has a similar proliferative index compared to anti-CD3 OKT3 in all four PBMC populations.
  • Fig. 27A-27E show that v1380 (L234AJJ
  • Fig. 27A-27E The results from the cytokine release assay as shown in Fig. 27A-27E include summary plots of PBMC supernatant TNFa (Fig. 27A) INFy (Fig. 27B), IL-2 (Fig. 27C), IL-4 (Fig. 27D), and IL-10 (Fig. 27E) levels following incubation with test items at 0.3 nM concentrations for 4 days (graph y-axis represents log cytokine levels in pg per mL from 4 donors).
  • Fig. 28A-28B show the results from the average stimulation index induced by v875 at 0.3nM (Fig. 28A) and 100 nM (Fig. 28B) concentrations on purified CD8+ T cells in the absence or presence of purified CD19+ B cells at 4 days incubation time-point.
  • Fig. 29A-29B show the results from the average stimulation index induced by v1380 at 0.3nM (Fig. 29A) and 100nM (Fig. 29B) concentrations on purified CD8+ T cells in the absence or presence of purified CD19+ B cells at 4 days incubation time-point.
  • Fig. 30A-30C show the results from the T:B cell bridging
  • FIG. 30A shows a direct comparison of human IgG and v875 at 200X magnification and illustrates a higher amount of bridging visible between Raji B cell and Jurkat T cells compared to human IgG.
  • Figure 30B and Figure 30C show two fields of view for v875 ( Figure 30B) and human IgG ( Figure 30C) at 400X magnification.
  • Fig. 31 A-31 B shows the SDS-PAGE analysis and relative purity of v875, v1380, v1379 and v891 following protein A and SEC purification, and following 47 day storage at 4°C.
  • Fig. 31 B shows the SDS-PAGE analysis and relative purity of additional exemplary hetermultimers including v875, v1653, v1654, v1655, v1656, v1660, v1800, and v1802 following protein A and SEC purification.
  • Fig. 32 shows the LC-MS results of the Max Ent. molecular weight profiles for v875.
  • Fig. 33A-33C show DSC results for heter
  • Fig. 34A-C show ability of heteromultimers described herein to bridge Raji B and Jurkat T cells (B:T), as well RajkRaji B cell bridging (B:B) and JurkatJurkat T cell bridging (T:T) assessed by FACS.
  • Fig. 34A shows the amount of T:B, B:B and T:T bridging of v875, v1379, v1380, v891 , v1381 , commercial OKT3 and human IgG over three experimental replicates.
  • Fig. 34A shows the amount of T:B, B:B and T:T bridging of v875, v1379, v1380, v891 , v1381 , commercial OKT3 and human IgG over three experimental replicates.
  • 34B shows the amount of T:B, B:B and T:T bridging of variants with engineered anti-CD3 warheads for stability enhancement (v1653, v1654, v1655, v1656, v1660, v1800, v1802) and v875, and human IgG.
  • stability enhancement v1653, v1654, v1655, v1656, v1660, v1800, v1802
  • v875 human IgG.
  • 34C shows the amount of T:B, B:B and T:T bridging of Fc knock-out variants that have either engineered anti-CD3 warheads for stability enhancement (v1666), or have human/cynomolgous monkey cross-reactive anti-CD3 and anti-CD19 scFvs (v4541 , v4543, v4545, v4548) commercial OKT3 anti-CD3 control, v2176 anti- CD19 control and human IgG negative control, and all variants mediate low T:T bridging.
  • engineered anti-CD3 warheads for stability enhancement v1666)
  • human/cynomolgous monkey cross-reactive anti-CD3 and anti-CD19 scFvs v4541 , v4543, v4545, v4548
  • Fig. 35 illustrates binding of heteromultimers described herein to human CD3 (top panel) and binding to the cynomologous CD3 receptor (bottom panel) as determined by ELISA.
  • Fig. 36A-36B illustrates affinity on linear and log scales
  • Fig. 37 depicts the binding of variant 1090 compared to the control 1087 in MALME-3M cells and indicates that v1090 has similar binding as v1087 to target MALME-3M cells.
  • any concentration range, percentage range, ratio range, or integer range is to be understood to include the value of any integer within the recited range and, when appropriate, fractions thereof (such as one tenth and one hundredth of an integer), unless otherwise indicated.
  • “about” means ⁇ 10% of the indicated range, value, sequence, or structure, unless otherwise indicated.
  • the terms “a” and “an” as used herein refer to “one or more” of the enumerated components unless otherwise indicated or dictated by its context. The use of the alternative (e.g., "or”) should be understood to mean either one, both, or any combination thereof of the alternatives.
  • amino acid names and atom names e.g.
  • polypeptide peptide
  • protein protein
  • polypeptide peptide
  • peptide protein
  • the terms apply to naturally occurring amino acid polymers as well as amino acid polymers in which one or more amino acid residues is a non-naturally encoded amino acid.
  • the terms encompass amino acid chains of any length, including full length proteins, wherein the amino acid residues are linked by covalent peptide bonds.
  • nucleotide sequence is intended to indicate a
  • nucleotide sequence may be of genomic, cDNA, RNA, semisynthetic or synthetic origin, or any combination thereof.
  • PCR polymerase chain reaction
  • PCR generally refers to a method for amplification of a desired nucleotide sequence in vitro, as described, for example, in U.S. Pat. No. 4,683,195.
  • the PCR method involves repeated cycles of primer extension synthesis, using oligonucleotide primers capable of hybridising preferentially to a template nucleic acid.
  • Transformation and “transfection” are used interchangeably to refer to the process of introducing DNA into a cell.
  • amino acid refers to naturally occurring and non- naturally occurring amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids
  • Naturally encoded amino acids are the 20 comr
  • arginine asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, praline, serine, threonine, tryptophan, tyrosine, and valine) and pyrrolysine and selenocysteine.
  • Amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an a carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, such as, homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium.
  • Such analogs have modified R groups (such as, norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid.
  • Reference to an amino acid includes, for example, naturally occurring proteogenic L-amino acids; D-amino acids, chemically modified amino acids such as amino acid variants and derivatives; naturally occurring non-proteogenic amino acids such as ⁇ -alanine, ornithine, etc.; and chemically synthesized compounds having properties known in the art to be characteristic of amino acids.
  • non-naturally occurring amino acids include, but are not limited to, a-methyl amino acids (e.g.
  • a-methyl alanine D- amino acids, histidine-like amino acids (e.g., 2-amino-histidine, ⁇ -hydroxy- histidine, homohistidine), amino acids having an extra methylene in the side chain (“homo" amino acids), and amino acids in which a carboxylic acid functional group in the side chain is replaced with a sulfonic acid group (e.g., cysteic acid).
  • non-natural amino acids including synthetic non-native amino acids, substituted amino acids, or one or more D-amino acids into the proteins of the present invention may be advantageous in a number of different ways.
  • D-amino acid-containing peptides, etc. exhibit increased stability in vitro or in vivo compared to L-amino acid-containing counterparts.
  • the construction of peptides, etc., incorporating D-amino acids can be particularly useful when greater intracellular stability is desired or required. More specifically, D-peptides, etc., are resistant to endogenous peptidases and proteases, thereby providing improved bioavailability of the molecule, and
  • peptides, etc. cannot be processed efficiently for major histocompatibility complex class ll-restricted presentation to T helper cells, and are therefore, less likely to induce humoral immune responses in the whole organism.
  • Amino acids may be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the lUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes.
  • Constantly modified variants applies to both amino acid and nucleic acid sequences. With respect to particular nucleic acid sequences, “conservatively modified variants” refers to those nucleic acids which encode identical or essentially identical amino acid sequences, or where the nucleic acid does not encode an amino acid sequence, to essentially identical sequences. Because of the degeneracy of the genetic code, a large number of functionally identical nucleic acids encode any given protein. For instance, the codons GCA, GCC, GCG and GCU all encode the amino acid alanine. Thus, at every position where an alanine is specified by a codon, the codon can be altered to any of the corresponding codons described without altering the encoded polypeptide.
  • nucleic acid variations are "silent variations," which are one species of conservatively modified variations. Every nucleic acid sequence herein which encodes a polypeptide also describes every possible silent variation of the nucleic acid.
  • silent variations are one species of conservatively modified variations.
  • Every nucleic acid sequence herein which encodes a polypeptide also describes every possible silent variation of the nucleic acid.
  • each codon in a nucleic acid except AUG, which is ordinarily the only codon for methionine, and TGG, which is ordinarily the only codon for tryptophan) can be modified to yield a functionally identical molecule.
  • each silent variation of a nucleic acid which encodes a polypeptide is implicit in each described sequence.
  • amino acid sequences one of ordinary skill in the art will recognize that individual substitutions, deletions or additions to a nucleic acid, peptide, polypeptide, or protein sequence which alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a nucleic acid, peptide, polypeptide, or protein sequence which alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a
  • nucleic acids or polypeptide sequences refer to two or more sequences or subsequences that are the same. Sequences are “substantially identical” if they have a percentage of amino acid residues or nucleotides that are the same (i.e., about 50% identity, about 55% identity, 60% identity, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, or about 95% identity over a specified region), when compared and aligned for maximum correspondence over a comparison window, or designated region as measured using one of the following sequence comparison algorithms (or other algorithms available to persons of ordinary skill in the art) or by manual alignment and visual inspection. This definition also refers to the complement of a test sequence. The identity can exist over a region that is at least about 50 amino acids or nucleotides that are the same. Sequences are “substantially identical” if they have a percentage of amino acid residues or nucleotides that are the same (i.e., about 50% identity, about 55% identity, 60% identity,
  • a polynucleotide encoding a polypeptide of the present invention may be obtained by a process comprising the steps of screening a library under stringent hybridization conditions with a labeled probe having a polynucleotide sequence of the invention or a fragment thereof, and isolating full-length cDNA and genomic clones containing said polynucleotide sequence.
  • a derivative, or a variant of a polypeptide is said to share
  • the derivative or variant is at least 75% the same as that of either the peptide or a fragment of the peptide having the same number of amino acid residues as the derivative. . In certain embodiments, the derivative or variant is at least 85% the same as that of either the peptide or a fragment of the peptide having the same number of amino acid residues as the derivative. In certain embodiments, the amino acid sequence of the derivative is at least 90% the same as the peptide or a fragment of the peptide having the same number of amino acid residues as the derivative.
  • the amino acid sequence of the derivative is at least 95% the same as the peptide or a fragment of the peptide having the same number of amino acid residues as the derivative. In certain embodiments, the derivative or variant is at least 99% the same as that of either the peptide or a fragment of the peptide having the same number of amino acid residues as the derivative.
  • bispecific is intended to include any agent, e.g., heteromultimer, monomer, protein, peptide, or protein or peptide complex, which has two different binding specificities.
  • agent e.g., heteromultimer, monomer, protein, peptide, or protein or peptide complex, which has two different binding specificities.
  • the molecule may bind to, or interact with, (a) a cell surface target molecule and (b) an Fc receptor on the surface of an effector cell.
  • the monomer is bispecific formed by attaching to the same transporter polypeptide, two cargo molecules with different binding specificities.
  • the heteromultimer is itself bispecific formed by attaching to the transporter polypeptides, at least two cargo molecules with different specificities.
  • multispecific or “heterospecific" is intended to include any agent, e.g., a protein, peptide, or protein or peptide complex, which has more than two different binding specificities.
  • the molecule may bind to, or interact with, (a) a cell surface target molecule such as but not limited to cell surface antigens, (b) an Fc receptor on the surface of an effector cell, and optionally (c) at least one other component.
  • a cell surface target molecule such as but not limited to cell surface antigens
  • an Fc receptor on the surface of an effector cell and optionally (c) at least one other component.
  • embodiments of the heteromultimers described herein are inclusive of, but not limited to, bispecific, trispecific, tetraspecific, and other multispecific molecules.
  • these molecules are directed to cell surface antigens, such as CD30, and to other targets, such as Fc receptors on effector cells.
  • isolated heteromultimer means a heteromultimer that has been identified and separated and/or recovered from a component of its natural cell culture environment. Contaminant components of its natural environment are materials that would interfere with diagnostic or therapeutic uses for the heteromultimer, and may include enzymes, hormones, and other proteinaceous or non-proteinaceous solutes.
  • the phrase "selectively (or specifically) hybridizes to” refers to the binding, duplexing, or hybridizing of a molecule only to a particular nucleotide sequence under stringent hybridization conditions when that sequence is present in a complex mixture (including but not limited to, total cellular or library DNA or RNA).
  • an “antibody” or “immunoglobulin” refers to a polypeptide substantially encoded by an immunoglobulin gene or
  • immunoglobulin genes or fragments thereof, which specifically bind and recognize an analyte (antigen).
  • the recognized immunoglobulin genes include the kappa, lambda, alpha, gamma, delta, epsilon and mu constant region genes, as well as the myriad immunoglobulin variable region genes.
  • Light chains are classified as either kappa or lambda.
  • Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, which in turn define the immunoglobulin classes, IgG, IgM, IgA, IgD, and IgE, respectively.
  • An exemplary immunoglobulin (antibody) structural unit is composed of two pairs of polypeptide chains, each pair having one "light"
  • variable light chain VL
  • variable heavy chain VH
  • the lgG1 heavy chain comprises of the VH, CH1 , CH2 and CH3 domains respectively from the N to C-terminus.
  • the light chain comprises of the VL and CL domains from N to C terminus.
  • the lgG1 heavy chain comprises a hinge between the CH1 and CH2 domains.
  • the immunoglobulin constructs comprise at least one
  • immunoglobulin domain from IgG, IgM, IgA, IgD, or IgE connected to a therapeutic polypeptide.
  • the immunoglobulin domain from IgG, IgM, IgA, IgD, or IgE connected to a therapeutic polypeptide.
  • the immunoglobulin domain from IgG, IgM, IgA, IgD, or IgE connected to a therapeutic polypeptide.
  • the immunoglobulin based construct such as a diabody, or a nanobody.
  • the immunoglobulin constructs described herein comprise at least one immunoglobulin domain from a heavy chain antibody such as a camelid antibody.
  • the immunoglobulin constructs provided herein comprise at least one immunoglobulin domain from a mammalian antibody such as a bovine antibody, a human antibody, a camelid antibody, a mouse antibody or any chimeric antibody.
  • antigenic determinant is synonymous with “antigen” and “epitope,” and refers to a site (e.g. a contiguous stretch of amino acids or a conformational configuration made up of different regions of non-contiguous amino acids) on a polypeptide macromolecule to which an antigen binding moiety binds, forming an antigen binding moiety-antigen complex.
  • Useful antigenic determinants can be found, for example, on the surfaces of tumor cells, on the surfaces of virus-infected cells, on the surfaces of other diseased cells, on the surface of immune cells, free in blood serum, and/or in the extracellular matrix (ECM).
  • ECM extracellular matrix
  • MCSP, FAP, CEA, EGFR, CD33, CD3 can be any native form the proteins from any vertebrate source, including mammals such as primates (e.g. humans) and rodents (e.g. mice and rats), unless otherwise indicated.
  • the antigen is a human protein.
  • the term encompasses the "full-length", unprocessed protein as well as any form of the protein that results from processing in the cell.
  • the term also encompasses naturally occurring variants of the protein, e.g. splice variants or allelic variants.
  • Exemplary human proteins useful as antigens include, but are not limited to: Melanoma-associated
  • MCSP Chondroitin Sulfate Proteoglycan
  • Proteoglycan 4 (UniProt no. Q6UVK1 (version 70), NCBI RefSeq no. NP
  • FAP Fibroblast Activation Protein
  • Carcinoembroynic antigen also known as Carcinoembryonic antigen-
  • CD33 also known as gp67 or Siglec-3 (UniProt no. P20138, NCBI Accession nos. NP 001076087, NP 001 171079); Epidermal Growth Factor Receptor (EGFR), also known as ErbB-1 or Herl (UniProt no. P0053, NCBI Accession nos. NP 958439, NP 958440), and CD3, particularly the epsilon subunit of CD3 (see UniProt no. P07766 (version 130), NCBI RefSeq no. NP 000724.1 , SEQ ID NO: 265 for the human sequence; or UniProt no.
  • the T cell activating bispecific antigen binding molecule of the invention binds to an epitope of an activating T cell antigen or a target cell antigen that is conserved among the activating T cell antigen or target antigen from different species.
  • binding is selective for the antigen and can be discriminated from unwanted or nonspecific interactions.
  • the ability of an antigen binding moiety to bind to a specific antigenic determinant can be measured either through an enzyme- linked immunosorbent assay (ELISA) or other techniques familiar to one of skill in the art, e.g. surface plasmon resonance (SPR) technique (analyzed on a BIAcore instrument) (Liljeblad et al, Glyco J 17, 323-329 (2000)), and traditional binding assays (Heeley, Endocr Res 28, 217-229 (2002)).
  • ELISA enzyme- linked immunosorbent assay
  • SPR surface plasmon resonance
  • an antigen binding moiety that binds to the antigen, or an antigen binding molecule comprising that antigen binding moiety has a dissociation constant (K D ) of ⁇ 1 ⁇ , ⁇ 100 nM, ⁇ 10 nM, ⁇ 1 nM, ⁇ 0.1 nM, ⁇ 0.01 nM, or ⁇ 0.001 nM (e.g. 10 ⁇ 8 M or less, e.g. from 10 ⁇ 8 M to 10 "13 M, e.g., from 10 "9 M to 10 "13 M).
  • K D dissociation constant
  • Affinity refers to the strength of the sum total of non-covalent interactions between a single binding site of a molecule (e.g., a receptor) and its binding partner (e.g., a ligand). Unless indicated otherwise, as used herein,
  • binding affinity refers to intrinsic binding affinity which reflects a 1 : 1
  • the affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (K D ), which is the ratio of dissociation and association rate constants (k 0ff and k on , respectively).
  • K D dissociation constant
  • equivalent affinities may comprise different rate constants, as long as the ratio of the rate constants remains the same.
  • Affinity can be measured by well established methods known in the art, including those described herein. A particular method for measuring affinity is Surface Plasmon Resonance (SPR).
  • Reduced binding for example reduced binding to an Fc receptor, refers to a decrease in affinity for the respective interaction, as measured for example by SPR.
  • the term includes also reduction of the affinity to zero (or below the detection limit of the analytic method), i.e. complete abolishment of the interaction.
  • increased binding refers to an increase in binding affinity for the respective interaction.
  • an "activating T cell antigen” as used herein refers to an antigenic determinant expressed on the surface of a T lymphocyte, particularly a cytotoxic T lymphocyte, which is capable of inducing T cell activation upon interaction with an antigen binding molecule. Specifically, interaction of an antigen binding molecule with an activating T cell antigen may induce T cell activation by triggering the signaling cascade of the T cell receptor complex. In a particular embodiment the activating T cell antigen is CD3.
  • T cell activation refers to one or more cellular response of a T lymphocyte, particularly a cytotoxic T lymphocyte, selected from: proliferation, differentiation, cytokine secretion, cytotoxic effector molecule release, cytotoxic activity, and expression of activation markers.
  • T cell activating bispecific antigen binding molecules of the invention are capable of inducing T cell activation.
  • Suitable assays to measure T cell activation are known in the art described herein.
  • a "target cell antigen” as used herein refers to an antigenic determinant presented on the surface of a target cell, for example a B cell in a
  • 31 WSGF tumor such as a cancer cell or a cell of the tumor stror
  • first and second with respect to antigen binding moieties etc., are used for convenience of distinguishing when there is more than one of each type of moiety. Use of these terms is not intended to confer a specific order or orientation of the T cell activating bispecific antigen binding molecule unless explicitly so stated.
  • a “Fab molecule” refers to a protein consisting of the VH and CH1 domain of the heavy chain (the “Fab heavy chain”) and the VL and CL domain of the light chain (the “Fab light chain”) of an immunoglobulin.
  • fused is meant that the components (e.g. a Fab molecule and an Fc domain subunit) are linked by peptide bonds, either directly or via one or more peptide linkers.
  • single-chain refers to a molecule comprising amino acid monomers linearly linked by peptide bonds.
  • one of the antigen binding moieties is a single-chain Fab molecule, i.e. a Fab molecule wherein the Fab light chain and the Fab heavy chain are connected by a peptide linker to form a single peptide chain.
  • the C-terminus of the Fab light chain is connected to the N-terminus of the Fab heavy chain in the single-chain Fab molecule.
  • one of the antigen binding moieties is a single- chain Fv molecule
  • crossover Fab molecule also termed “Crossfab” is meant a crossover Fab molecule
  • the crossover Fab molecule comprises a peptide chain composed of the light chain variable region and the heavy chain constant region, and a peptide chain composed of the heavy chain variable region and the light chain constant region.
  • the crossover Fab molecule comprises a peptide chain composed of the light chain variable region and the heavy chain constant region, and a peptide chain composed of the heavy chain variable region and the light chain constant region.
  • peptide chain comprising the heavy chain variable region is referred to herein as the "heavy chain” of the crossover Fab molecule.
  • FR refers to variable domain residues other than hypervariable region (HVR) residues.
  • the FR of a variable domain generally consists of four FR domains: FR1 , FR2, FR3, and FR4. Accordingly, the HVR and FR sequences generally appear in the following sequence in VH (or VL): FR1 -H1 (L1 )-FR2-H2(L2)-FR3-H3(L3)-FR4.
  • the "class" of an antibody or immunoglobulin refers to the type of constant domain or constant region possessed by its heavy chain. There are five major classes of antibodies: IgA, IgD, IgE,
  • IgG, and IgM may be further divided into subclasses (isotypes), e.g., IgGi, lgG 2 , lgG 3 , lgG , IgAi, and lgA 2 .
  • immunoglobulins are called ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ , respectively.
  • Fc domain or "Fc region” herein is used to define a C- terminal region of an immunoglobulin heavy chain that contains at least a portion of the constant region.
  • the term includes native sequence Fc regions and variant Fc regions.
  • the boundaries of the Fc region of an IgG heavy chain might vary slightly, the human IgG heavy chain Fc region is usually defined to extend from Cys226, or from Pro230, to the carboxyl-terminus of the heavy chain.
  • the C-terminal lysine (Lys447) of the Fc region may or may not be present.
  • numbering of amino acid residues in the Fc region or constant region is according to the EU numbering system, also called the EU index, as described in Kabat et al, Sequences of Proteins of Immunological Interest, 5th Ed. Public Health
  • a "subunit" of an Fc domain as used herein refers to one of the two polypeptides forming the dimeric Fc domain, i.e. a polypeptide comprising C-terminal constant regions of an immunoglobulin heavy chain, capable of stable self-association.
  • a subunit of an IgG Fc domain comprises ar
  • a "modification promoting the association of the first and the second subunit of the Fc domain” is a manipulation of the peptide backbone or the post-translational modifications of an Fc domain subunit that reduces or prevents the association of a polypeptide comprising the Fc domain subunit with an identical polypeptide to form a homodimer.
  • a modification promoting association as used herein particularly includes separate modifications made to each of the two Fc domain subunits desired to associate (i.e. the first and the second subunit of the Fc domain), wherein the promote association of the two Fc domain subunits and the formation of heterodimers.
  • a modification promoting association may alter the structure or charge of one or both of the Fc domain subunits so as to make their
  • effector functions refers to those biological activities attributable to the Fc region of an antibody, which vary with the antibody isotype.
  • antibody effector functions include: Clq binding and complement dependent cytotoxicity (CDC), Fc receptor binding, antibody- dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (AD CP), cytokine secretion, immune complex-mediated antigen uptake by antigen presenting cells, down regulation of cell surface receptors (e.g. B cell receptor), and B cell activation.
  • albumin refers collectively to albumin protein or amino acid sequence, or an albumin segment or variant, having one or more functional activities (e.g., biological activities) of albumin.
  • albumin refers to human albumin or segments thereof (see for example, EP 201 239, EP 322 094 WO 97/24445, WO95/23857) especially the mature form of human albumin, or albumin from other vertebrates, or segments thereof, or analogs or variants of these molecules or fragments thereof.
  • albumin refers to a truncated version of albumin.
  • quadsi-albumin refers to a heter
  • the monomeric polypeptides are "segments" that preferentially associate as heteromultimeric pairs to form a quasi-protein.
  • the quasi-albumin has 90% of the activity of the whole albumin. In some embodiments, the quasi-albumin has 75% of the activity of whole- albumin. In an embodiment, the quasi-albumin has 50% of the activity of whole albumin. In some embodiments, the quasi-albumin has 50-75% of the activity of whole albumin. In an embodiment, quasi-albumin has 80% of the activity of whole albumin. In some embodiments, the quasi-albumin has 90% of the structure of whole albumin as determined by molecular modeling. In some embodiments, the quasi-albumin has 80% of the structure of whole albumin as determined by molecular modeling. In some embodiments, the quasi-albumin has 70% of the structure of whole albumin as determined by molecular modeling. In some embodiments, the quasi-albumin has 50% of the structure of whole albumin as determined by molecular modeling. In some embodiments, the quasi-albumin has 50%-75% of the structure of whole albumin as determined by molecular modeling.
  • HSA human serum albumin
  • HA human albumin
  • albumin and serum albumin are broader, and encompass human serum albumin (and fragments and variants thereof) as well as albumin from other species (and fragments and variants thereof).
  • each albumin-based construct described herein is based on a variant of normal HA.
  • variants includes insertions, deletions and substitutions, either conservative or non conservative, where such changes do not substantially alter one or more of the oncotic, useful ligand-binding and non-immunogenic properties of albumin, or the active
  • the isolated heteromultimeric constructs described herein include naturally occurring polymorphic variants of human albumin and fragments of human albumin, for example those fragments disclosed in EP 322 094 (namely HA (Pn), where n is 369 to 419).
  • the albumin is derived from any vertebrate, especially any mammal that includes but is not limited to human, cow, sheep, rat, mouse, rabbit, horse, dog or pig. In certain embodiments, the albumin is derived from non-mammalian albumins including, but are not limited to hen and salmon.
  • an "alloalbumin” is a genetic variant of albumin.
  • the alloalbumin is human alloalbumin (HAA).
  • HAA human alloalbumin
  • Alloalbumins that differ in electrophoretic mobility from albumin have been identified through population genetics surveys in the course of clinical electrophoresis, or in blood donor surveys. As markers of mutation and migration, alloalbumins are of interest to geneticists, biochemists, and anthropologists, but most of these
  • Table 1 List of substitutions comprised by various alloalbumins as compared to HA of SEQ ID NO: 1. Thermostability, half-life information and other HAAs are provided in Krogh-hansen et al. Biochim Biophys Acta 1747, 81 -88(
  • segmentation refers to a precise internal splice of the original protein sequence which results in “segments” of the protein sequence that preferentially associate as heteromultimers to form a quasi-protein.
  • proteins and/or 'quasi-native structures' present the native protein like functional and structural characteristics. Proteins are naturally dynamics molecules and display an ensemble of structural configurations although we ascribe a native structure to it, such as the one obtained by X-ray
  • the reference native protein in this case is the protein from which the transporter polypeptide is derived and the reference native structure is the structure of the monomeric protein from which the transporter polypeptide is derived.
  • the reference native structure is the structure of the monomeric protein from which the transporter polypeptide is derived.
  • two or more different polypeptides self-assemble to form a heteromultimeric structural and exhibit functional characteristics like a native protein which itself is a monomeric entity.
  • heteromultimer constructs comprising transporter polypeptides derived from albumin that self-assemble to form a heteromultimer that exhibits native albumin like functional characteristics such as FcRn binding and structural characteristics. These heteromultimers are referred to as being quasi-native.
  • CD3 complex as described herein is a complex of at least five membrane-bound polypeptides in mature T-lymphocytes that are non- covalently associated with one another and with the T-cell receptor.
  • the CD3 complex includes the gamma, delta, epsilon, zeta, and eta chains (also referred to as gamma, delta, epsilon, zeta, and eta chains (also referred to CD3 complex).
  • CD19 has proved to be a very useful target.
  • CD19 is expressed in the whole B lineage from the pro B cell to the mature B cell, it is not shed, is uniformly expressed on all lymphoma cells, and is absent from stem cells.
  • compositions containing a multispecific heteromultimeric construct described herein can be administered for prophylactic, enhancing, and/or therapeutic treatments.
  • engineered, engineered, engineering are considered to include any manipulation of the peptide backbone or the post- translational modifications of a naturally occurring or recombinant polypeptide or fragment thereof.
  • Engineering includes modifications of the amino acid sequence, of the glycosylation pattern, or of the side chain group of individual amino acids, as well as combinations of these approaches.
  • the engineered proteins are expressed and produced by standard molecular biology
  • isolated nucleic acid molecule or polynucleotide is intended a nucleic acid molecule, DNA or RNA, which has been removed from its native environment.
  • a recombinant polynucleotide encoding a polypeptide contained in a vector is considered isolated.
  • Further examples of an isolated polynucleotide include recombinant polynucleotides maintained in
  • An isolated polynucleotide includes a polynucleotide molecule contained in cells that ordinarily contain the polynucleotide molecule, but the polynucleotide molecule is present extrachromosomally or at a chromosomal location that is different from its natural chromosomal location.
  • Isolated RNA molecules include in vivo or in vitro RNA transcripts, as well as positive and negative strand forms, and double-stranded forms. Isolated polynucleotides or nucleic acids described herein, further include such molecules produced synthetically.
  • a polynucleotide or a nucleic acid in certain embodiments, include a regulatory element such as a promoter, ribosome binding site, or a transcription terminator.
  • nucleic acid or polynucleotide having a nucleotide sequence at least, for example, 95% "identical" to a reference nucleotide sequence of the present invention it is intended that the nucleotide sequence of the
  • polynucleotide is identical to the reference sequence except that the
  • polynucleotide sequence may include up to five point mutations per each 100 nucleotides of the reference nucleotide sequence.
  • up to 5% of the nucleotides in the reference sequence may be deleted or substituted with another nucleotide, or a number of nucleotides up to 5% of the total nucleotides in the reference sequence may be inserted into the reference sequence.
  • These alterations of the reference sequence may occur at the 5' or 3' terminal positions of the reference nucleotide sequence or anywhere between those terminal positions,
  • any particular polynucleotide sequence is at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to a nucleotide sequence of the present invention can be determined conventionally using known computer programs, such as the ones discussed above for polypeptides (e.g. ALIGN-2).
  • expression cassette refers to a
  • the recombinant expression cassette can be incorporated into a plasmid, chromosome, mitochondrial DNA, plastid DNA, virus, or nucleic acid fragment.
  • the recombinant expression cassette portion of an expression vector includes, among other sequences, a nucleic acid sequence to be transcribed and a promoter.
  • the expression cassette of the invention comprises polynucleotide sequences that encode bispecific antigen binding molecules of the invention or fragments thereof.
  • vector or "expression vector” is synonymous with
  • expression construct refers to a DNA molecule that is used to introduce and direct the expression of a specific gene to which it is operably associated in a target cell.
  • the term includes the vector as a self-replicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced.
  • the expression vector of the present invention comprises an expression cassette. Expression vectors allow transcription of large amounts of stable mRNA. Once the expression vector is inside the target cell, the ribonucleic acid molecule or protein that is encoded by the gene is produced by the cellular transcription and/or translation machinery.
  • the expression vector of the invention comprises an expression cassette that comprises polynucleotide sequences that encode bispecific antigen binding molecules of the invention or fragments thereof.
  • host cell refers to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells.
  • Host cells include
  • transformants and “transformed cells,” which include the primary transformed cell and progeny derived therefrom without regard to the number of passages.
  • progeny are not completely identical in nucleic acid content to a parent cell, but may contain mutations. Mutant progeny that have the same function or biological activity as screened or selected for in the
  • Host cells include cultured cells, e.g. mammalian cultured cells, such as CHO cells, BHK cells, NSO cells, SP2/0 cells, YO myeloma cells, P3X63 mouse myeloma cells, PER cells, PER.C6 cells or hybridoma cells, yeast cells, insect cells, and plant cells, to name only a few, but also cells comprised within a transgenic animal, transgenic plant or cultured plant or animal tissue.
  • mammalian cultured cells such as CHO cells, BHK cells, NSO cells, SP2/0 cells, YO myeloma cells, P3X63 mouse myeloma cells, PER cells, PER.C6 cells or hybridoma cells, yeast cells, insect cells, and plant cells, to name only a few, but also cells comprised within a transgenic animal, transgenic plant or cultured plant or animal tissue.
  • An "activating Fc receptor” is an Fc receptor that following engagement by an Fc domain of an antibody elicits signaling events that stimulate the receptor-bearing cell to perform effector functions.
  • Human activating Fc receptors include FcyRllla (CD 16a), FcyRI (CD64), and FcyRlla (CD32).
  • Antibody-dependent cell-mediated cytotoxicity is an immune mechanism leading to the lysis of antibody-coated target cells by immune effector cells.
  • the target cells are cells to which antibodies or derivatives thereof comprising an Fc region specifically bind, generally via the protein part that is N-terminal to the Fc region.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • reduced ADCC is defined as either a reduction in the number of target cells that are lysed in a given time, at a given concentration of antibody in the medium surrounding the target cells, by the mechanism of ADCC defined above, and/or an increase in the concentration of antibody in the medium surrounding the target cells, required to achieve the lysis of a given number of target cells in a given time, by the mechanism of ADCC.
  • ADCC is relative to the ADCC mediated by the same antibody produced by the same type of host cells, using the same standard production, purification, formulation and storage methods (which are known to those skilled in the art), but that has not been engineered.
  • the reduction in ADCC mediated by an antibody comprising in its Fc domain an amino acid substitution that reduces ADCC is relative to the ADCC mediated by the same antibody without this amino acid substitution in the Fc domain.
  • heteromultimer described herein refers to the amount that is necessary to result in a physiological change in the cell or tissue to which it is administered.
  • a "therapeutically effective amount" of an agent e.g. a
  • composition comprising a multispecific heteromultimer described herein, refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result.
  • a therapeutically effective amount of an agent for example eliminates, decreases, delays, minimizes or prevents adverse effects of a disease.
  • mammals include, but are not limited to, domesticated animals (e.g. cows, sheep, cats, dogs, and horses), primates (e.g. humans and non-human primates such as monkeys), rabbits, and rodents (e.g. mice and rats). Particularly, the individual or subject is a human.
  • domesticated animals e.g. cows, sheep, cats, dogs, and horses
  • primates e.g. humans and non-human primates such as monkeys
  • rabbits e.g. mice and rats
  • rodents e.g. mice and rats
  • composition refers to a preparation which is in such form as to permit the biological activity of a multispecific heteromultimer construct contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
  • a "pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical composition, other than an active ingredient, which is nontoxic to a subject.
  • a pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
  • treatment refers to clinical intervention in an attempt to alter the natural course of a disease in the individual being treated, and can be performed either for prophylaxis or during the course of clinical pathology.
  • Desirable effects of treatment include, but are not limited to, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or
  • multispecific heteromultimer constructs described herein are used to delay development of a disease or to slow the progression of a disease.
  • instructions is used to refer to instructions customarily included in commercial packages of therapeutic products that contain information about the indications, usage, dosage, administration, combination therapy, contraindications and/or warnings concerning the use of such therapeutic products.
  • cross-species binding means binding of a binding domain described herein to the same target molecule in humans and other organisims for instance, but not restricted to non-chimpanzee primates.
  • cross-species binding or “interspecies binding” is to be understood as an interspecies reactivity to the same molecule "X” (i.e. the homolog) expressed in different species, but not to a molecule other than "X”.
  • macaque CD3 epsilon can be determined, for instance, by FACS analysis.
  • the FACS analysis is carried out in a way that the respective monoclonal antibody is tested for binding to human and non-chimpanzee primate cells, e.g.
  • macaque PSCA, CD19, C-MET, Endosialin, EpCAM, IGF-1 R or FAPa can be determined, for instance, by FACS analysis.
  • the FACS analysis is carried out in a way that the respective monoclonal antibody is tested for binding to human and non-chimpanzee primate cells, e.g. macaque cells,
  • Endosialin, EpCAM, IGF-1 R or FAPa antigens were endosialin, EpCAM, IGF-1 R or FAPa antigens, respectively.
  • isolated multispecific heteromultimer constructs comprising a first polypeptide construct comprising a first heavy chain polypeptide and a CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; and a second polypeptide construct comprising a second heavy chain polypeptide which is different from said first heavy chain polypeptide, and an antigen binding polypeptide construct that binds to a target antigen on at least one B cell, wherein:at least one of said
  • CD3 binding polypeptide construct and said antigen binding polypeptide construct comprises a single chain Fv region; said multispecific heteromultimer construct simultaneously engages said at least one B cell and said at least one
  • CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell; and said first and second heavy chain polypeptides form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc which is formed with stability at least comparable to a native homodimeric Fc, and with purity such that when said multispecific heteromultimer construct is expressed from a mammalian cell in an expression product, said expression product comprises at least about 70% of said multispecific heteromultimer, and less than 10% monomers or homodimers of said first or second polypeptide constructs.
  • the expression product comprises at least about 75% of said multispecific heteromultimer, and less than 15% monomers or homodimers of said first or second polypeptide constructs. In certain embodiments, the expression product comprises at least about 80% of said multispecific heteromultimer, and less than 10% monomers or homodimers of said first or second polypeptide
  • expression product comprises at least about 90% of said multispecific heteromultimer, and less than 10% monomers or homodimers of said first or second polypeptide constructs.
  • [00173] is the isolated multispecific heteromultimer construct, wherein said first or second polypeptide construct is devoid of at least one of immunoglobulin light chain, and immunoglobulin first constant (CH1 ) region.
  • an isolated multispecific heteromultimer construct comprising: a first polypeptide construct comprising a first heavy chain polypeptide and a CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second heavy chain polypeptide which is different from said first heavy chain polypeptide, and wherein said second polypeptide construct does not comprise an antigen binding polypeptide construct that binds to a target antigen on a B cell; wherein: said multispecific heteromultimer construct simultaneously engages at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell; and said first and second heavy chain polypeptides form a heterodimeric Fc region comprising a variant
  • immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc, wherein: said heterodimeric
  • Fc is formed with stability at least comparable to a native homodimeric Fc, and said heterodimeric Fc is formed with purity such that when said multispecific heteromultimer construct is expressed from a stable mammalian cell in an expression product, said expression product comprises at least about 75% of said multispecific heteromultimer, and less than 10% monomers or
  • the heterodimeric Fc interacts with cell surface receptors such as
  • an isolated multispecific heteromultimer construct comprising: a first polypeptide construct comprising a first heavy chain polypeptide and a CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second heavy chain polypeptide which is different from said first heavy chain polypeptide, and a steric modulator construct which exhibits negligible receptor binding; wherein: said multispecific heteromultimer construct simultaneously engages at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell; and said first and second heavy chain polypeptides form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc, wherein: said heterodimeric Fc is formed with stability at least comparable to a native homodimeric Fc, and said heterodimeric F
  • the steric modulator construct is a polypeptide sequence that helps modulate sterical features of the multispecific
  • heteromultimer as the multimer binds to T and/or B cells.
  • the steric modulator construct comprises a polypeptide domain that is designed de-novo.
  • the steric modulator construct comprises polypeptide domains obtained by engineering a known
  • the steric modulator construct comprises an engineered Fab region or fragment thereof which is engineered to remove binding properties.
  • an isolated multispecific heteromultimer construct comprising: a first polypeptide construct comprising a first heavy chain polypeptide and a CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second heavy chain polypeptide which is different from said first heavy chain polypeptide, and wherein said second polypeptide construct does not comprise an antigen binding polypeptide construct; wherein: said multispecific heteromultimer construct simultaneously engages at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell; and said first and second heavy chain polypeptides form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc, wherein: said heterodimeric
  • Fc is formed with stability at least comparable to a native homodimeric Fc, and said heterodimeric Fc is formed with purity such that when said multispecific heteromultimer construct is coexpressed from a stable mammalian cell in an expression product, said expression product comprises at least about 75% of said multispecific heteromultimer, and less than 10% monomers or
  • heterodimer Fc region comprises a variant CH2 domain or hinge comprising amino acid modifications that prevents functionally effective binding to all the Fcgamma receptors.
  • the multispecific heteromultimer constructs that bind at least one B cell with a valency greater than one, and simultaneously engage said at least one B cell and at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell.
  • binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; and a second polypeptide construct comprising a second heavy chain polypeptide which is different from said first heavy chain polypeptide, and an antigen binding polypeptide construct that binds to a target antigen on at least one B cell; wherein: at least one of said CD3 binding polypeptide construct and said antigen binding polypeptide construct optionally comprises a single chain Fv region; said first and second heavy chain polypeptides form a heterodimeric Fc region comprising a variant
  • immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc with stability at least comparable to a native homodimeric Fc, and with purity such that when said multispecific heteromultimer construct is expressed from a mammalian cell in an expression product, said expression product comprises greater than 75% of said multispecific heteromultimer, and less than 10% monomers or
  • multispecific heteromultimer construct is capable of interacting with the B-cell via the said antigen binding polypeptide construct on the second heavy chain as well as interaction via the said heterodimeric Fc with FcgRllb receptors on the B-cell to show valency greater than one during B-cell engagement.
  • heterodimer Fc region comprises a variant CH2 domain comprising amino acid modifications to promote selective binding of a Fcgamma receptor.
  • the heteromultimer wherein the variant CH2 domain selectively binds at least one of Fcgammallla and Fcgammallb receptor as compared to wild-type CH2 domain.
  • [00181] is an isolated multispecific heteromultimer construct described herein wherein the heterodimer Fc is glycosylated.
  • [00182] is an isolated multispecific heteromultimer described herein, wherein the heterodimer Fc is afucosylated.
  • Provded herein are multispecific heteromultimer comstructs that comprise different antigen binding moieties, fused to one or the other of the two subunits of the Fc domain, thus the two subunits of the Fc domain are typically comprised of two non-identical polypeptide chains.
  • polypeptides is modified to promote the association of the desired polypeptides.
  • the first and second heavy chain are identical to each other.
  • polypeptides of the heteromultimer constructs described herein form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc which is formed with stability at least comparable to a native homodimeric Fc, and with purity such that when said multispecific heteromultimer construct is coexpressed from a mammalian cell as an expression product, said expression product comprises at least about 75% of said multispecific heteromultimer, and less than 10% monomers or
  • the first and second heavy chain are identical to each other.
  • polypeptides of the heteromultimer constructs described herein form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc which is formed with stability at least comparable to a native homodimeric Fc, and with purity such that when said multispecific heteromultimer construct is co-expressed from a mammalian cell as an expression product, said expression product comprises at least about 90% of said multispecific heteromultimer, and less than 10% monomers or
  • the first and secon are identical to each other.
  • polypeptides of the heteromultimer constructs described herein form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc which is formed with stability at least comparable to a native homodimeric Fc, and with purity such that when said multispecific heteromultimer construct is co-expressed from a mammalian cell as an expression product, said expression product comprises at least about 95% of said multispecific heteromultimer, and less than 10% monomers or
  • the isolated multispecific heteromultimer provided herein, wherein the variant CH3 domain has a melting temperature (Tm) of about 73°C or greater.
  • [00191] in certain embodiments is the isolated multispecific heteromultimer described herein, wherein the heterodimer Fc region is formed with a purity greater than about 78%.
  • the isolated multispecific heteromultimer described herein wherein the heterodimer Fc region is formed with a purity of at least about 78% or greater and the Tm is at least about 75°C.
  • the isolated multispecific heteromultimer described herein wherein the heterodimer Fc region is formed with a purity of at least about 75% and the Tm is about 75°C or greater.
  • isolated multispecific heteromultimer constructs wherein: a) the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications L351 Y,
  • the variant CH3 sequence of the second transporter polypeptide comprises the amino acid modifications T366L, K392M, and
  • the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications L351Y, F405A, and Y407V
  • the variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications T366L, K392L, and T394W
  • the variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications T350V, T366L, K392M, and T394W; d) the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications T350V, L351Y, F405A, and Y407V, and the variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications T350V, T366L, K392L, and T394W; e) the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications T366L, N390R, K392R, and T394W, and the variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications L351 Y, S400E, F405A, and Y407V; or f) the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications T350V, T366L
  • the Fc regions of the heteromultimer constructs described herein comprises a variant CH2 domain comprising amino acid modifications to promote selective binding of a Fcgamma receptor.
  • the isolated multispecific heteromultimer described herein comprise a variant CH2 domain that selectively binds a Fcgammallb receptor with an affinity greater than that of the wild-type CH2 domain.
  • the isolated multispecific heteromultimer described herein comprise a variant CH2 domain that selectively binds a FcgammallA and/or FcgammalllA receptor with an affinity greater than that of the wild-type CH2 domain.
  • the Fc regions of the heteromultimer constructs described herein exhibit reduced binding affinity to an Fc receptor
  • the Fc region exhibits less than 50%, alternatively less than 20%, alternatively less than 10% and in some embodiments, less than 5% of the binding affinity to an Fc receptor, as compared to a native lgG1 Fc region, and/or less than 50%, alternatively less than 20%, alternatively less than 10% and in some embodiments less than 5% of the effector function, as compared to a native lgG1 Fc region.
  • the Fc region of a heteromutlimer construct described herein does not substantially bind to an Fc receptor or induce appreciable effector function.
  • the Fc receptor is an Fey receptor.
  • the Fc receptor is a mammalian Fc receptor.
  • the mammalian Fc receptor is a human Fc receptor.
  • the Fc receptor is an activating Fc receptor.
  • the Fc receptor is an activating human Fey receptor, more specifically human FcyRllla, FcyRI or FcyRlla, most specifically human
  • the effector function is one or more function selected from the group consisting of CDC, ADCC, ADCP, and cytokine secretion. In a particular embodiment the effector function is ADCC.
  • the Fc region exhibits binding affinity to neonatal Fc receptor (FcRn). In certain embodiments, the FcRn binding affinity is substantially similar to that of a native lgG1 Fc. In some embodiments, substantially similar binding to FcRn is achieved when the Fc region of a heteromultimer construct described herein exhibits greater than about 70%, or in some embodiments greater than about 80%, and in some particular embodiments greater than about 90% of the binding affinity of a native lgG1 Fc domain to FcRn.
  • the Fc region of a heteromultimer construct described herein is engineered to have reduced binding affinity to an Fc receptor and/or reduced effector function, as compared to a non-engineered Fc domain.
  • the engineered mutations are present in the lower hinge and CH2 domain.
  • the Fc region of a heteromultimer construct described herein is engineered to have reduced binding affinity to an Fc receptor and/or reduced effector function, as compared to a non-engineered Fc domain.
  • the engineered mutations are present in the lower hinge and CH2 domain.
  • the Fc region of a heteromultimer construct described herein is engineered to have reduced binding affinity to an Fc receptor and/or reduced effector function, as compared to a non-engineered Fc domain.
  • the engineered mutations are present in the lower hinge and CH2 domain.
  • 55 WSGF heteromultimer described herein comprises one or mc
  • the same one or more amino acid mutation is present in each of the two subunits of the Fc region. In some embodiments, different amino acid mutations are introduced in each of the two subunits of the Fc region. In one embodiment the amino acid mutation reduces the binding affinity of the Fc region to an Fc receptor. In one embodiment the amino acid mutation reduces the binding affinity of the Fc region to an Fc receptor by at least 2-fold, or in some embodiments at least 5-fold, or in an embodiment at least 10-fold.
  • the combination of these amino acid mutations reduces the binding affinity of the Fc region to an Fc receptor by at least 10-fold, or in some embodiments at least 20-fold, or in certain embodiments at least 50-fold.
  • the binding affinity of the Fc region for the Fc receptor is reduced to an extent where there is no longer any detectable binding for the mutant Fc for the Fc receptor in standard binding assay such as using the SPR instrument.
  • the heteromultimer construct described herein comprising an engineered Fc domain exhibits less than 20%, and in certain embodiments less than 10%, and in select embodiments less than 5% of the binding affinity to an Fc receptor as compared to a corresponding construct comprising an Fc domain which is not engineered to reduce binding to an Fc receptor.
  • the Fc receptor is an Fey receptor.
  • the Fc receptor is a human Fc receptor.
  • the Fc receptor is an activating Fc receptor.
  • the Fc receptor is an activating human Fey receptor which in certain embodiments is one of human FcyRllla, FcyRI and FcyRlla.
  • binding to each of these receptors is reduced.
  • binding affinity to a complement component for instance, but not restricted to C1 q, is also reduced.
  • binding affinity to a complement component for instance, but not restricted to C1 q
  • FcRn 56 WSGF neonatal Fc receptor
  • the reduced effector function can include, but is not limited to, one or more of the following: reduced complement dependent cytotoxicity (CDC), reduced antibody-dependent cell-mediated cytotoxicity (ADCC), reduced antibody-dependent cellular phagocytosis (ADCP), reduced cytokine secretion, reduced immune complex-mediated antigen uptake by antigen- presenting cells, reduced binding to NK cells, reduced binding to macrophages, reduced binding to monocytes, reduced binding to polymorphonuclear cells, reduced direct signaling inducing apoptosis, reduced crosslinking of target- bound antibodies, reduced dendritic cell maturation, or reduced T cell priming.
  • CDC reduced complement dependent cytotoxicity
  • ADCC reduced antibody-dependent cell-mediated cytotoxicity
  • ADCP reduced antibody-dependent cellular phagocytosis
  • reduced immune complex-mediated antigen uptake by antigen- presenting cells reduced binding to NK cells, reduced binding to macrophages, reduced binding to monocytes, reduced binding to polymorphonuclear cells, reduced direct signaling
  • the reduced effector function is one or more selected from the group of reduced CDC, reduced ADCC, reduced ADCP, and reduced cytokine secretion. In certain embodiments the reduced effector function is reduced ADCC. In one embodiment the reduced ADCC is less than 20% of the ADCC induced by a non-engineered Fc domain (or a T cell activating bispecific antigen binding molecule comprising a non-engineered Fc domain). In another embodiment the reduced ADCC is less than 50% of the ADCC induced by a non-engineered Fc domain (or a T cell activating bispecific antigen binding molecule comprising a non-engineered Fc domain). In a further embodiment the reduced ADCC is less than 10% of the ADCC induced by a non-engineered Fc domain (or a T cell activating bispecific antigen binding molecule comprising a non-engineered Fc domain).
  • heterodimer Fc region comprises a variant CH2 domain comprising amino acid modifications to promote selective binding of a Fcgamma receptor as described herein.
  • the multispecific heteromultimer construct comprises a variant CH2 region that binds at least one B cell such that the heteromultimer construct binds B cells with a valency greater than one.
  • isolated multispecific heteromultimer constructs comprising: a first polypeptide construct comprising a first transporter polypeptide fused to at least one CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second transporter polypeptide which is different from said first transporter polypeptide, fused to at least one antigen binding polypeptide construct that binds to a target antigen on at least one B cell;
  • each transporter polypeptide comprises an amino acid sequence with at least 85% identity to a segment of the protein.
  • each transporter polypeptide comprises an amino acid sequence with at least 80% identity to a segment of the protein.
  • each transporter polypeptide comprises an amino acid sequence with at least 95% identity to a segment of the protein.
  • each transporter polypeptide comprises an amino acid sequence with at least 99% identity to a segment of the protein.
  • each transporter polypeptide is an albumin
  • embidments is an albumin based i
  • each transporter polypeptide is an allo-albumin derivative. In certain embodiments is an isolated multispecific heteromultimer described herein wherein each transporter polypeptide is derived from a different alloalbumin. In some embodiments, each transporter polypeptide comprises an amino acid sequence with at least 75% identity to a segment of albumin. In some embodiments, each transporter polypeptide comprises an amino acid sequence with at least 80% identity to a segment of albumin. In some embodiments, each transporter polypeptide comprises an amino acid sequence with at least 90% identity to a segment of albumin. In some other embodiments, each transporter polypeptide comprises an amino acid sequence with at least 95% identity to a segment of albumin. In some other embodiments, each transporter polypeptide comprises an amino acid sequence with at least 99% identity to a segment of albumin.
  • an albumin based isolated multispecific heteromultimer construct comprising: a first monomer comprising a first transporter polypeptide fused to at least one CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second transporter polypeptide which is different from said first transporter polypeptide, fused to at least one antigen binding polypeptide construct that binds to a target antigen on at least one B cell; wherein said first and second transporter polypeptides are obtained by segmentation of albumin, and each transporter polypeptide comprising an amino acid sequence with at least 90% identity to a segment of albumin such that said transporter polypeptides self-assemble to form quasi-native albumin, and wherein said first cargo polypeptide does not have any binding domain present in said second cargo polypeptide.
  • albumin based multispecific heteromultimer constructs as described above, wherein said first transporter polypeptide comprising at least one mutation selected from A194C, L198C, W214C,
  • polypeptide comprises at least one mutation selected from L331 C, A335C,
  • V343C, L346C, A350C, V455C, and N458C are examples of V343C, L346C, A350C, V455C, and N458C.
  • multispecific heteromultimer constructs described herein wherein said multispecific heteromultimer construct simultaneously engages said at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell.
  • heteromultimer constructs comprising transporter polypeptides derived from albumin that self-assemble to form a heteromultimer that exhibits native albumin like functional characteristics such as FcRn binding and structural characteristics.
  • the tumor cells are from a solid tumor. In some embodiments, the
  • heteromultimer constructs described herein home to tumor cells and
  • heteromultimer constructs described herein home to at least one tumor cell, bind simultaneously to said at least one tumor cell and and at least one T-cell in a manner that results in the lysis of said tumor cell.
  • the heteromultimer constructs described herein home to at least one tumor cell, bind simultaneously to said at least one tumor cell and and at least one T-cell such that the binding to said tumor cell is with a higher valency than the binding to said T-cell, and causes the lysis of said tumor cell.
  • CD3 complex binding polypeptide constructs [00212] CD3 complex binding polypeptide constructs:
  • heteromultimer construct comprises at least one CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell.
  • the at least one CD3 binding polypeptide construct comprises at least one CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell.
  • 60 WSGF comprises at least one CD3 binding domain from a Ct
  • the at least one CD3 binding domain comprises at least one amino acid modification that reduces immunogenicity as compared to a corresponding CD3 binding domain not comprising said modification.
  • the at least one CD3 binding domain comprises at least one amino acid modification that increases its stability as measured by T m , as compared to a corresponding CD3 binding domain not comprising said modification. In some embodiments, there is about a 3 degree increase in the T m as compared to the native CD3 binding domain not comprising said at least one modification.
  • the at least one CD3 binding polypeptide construct described herein comprises at least one CD3 binding domain from a CD3 specific antibody wherein said CD3 specific antibody is a heavy chain antibody devoid of light chains.
  • the at least one CD3 binding polypeptide construct described herein comprises at least one CD3 binding domain derived from a non-antibody protein scaffold domain.
  • the CD3 binding polypeptide constructs are CD3 binding Fab constructs (i.e. antigen binding constructs comprising a heavy and a light chain, each comprising a variable and a constant region).
  • said Fab construct is mammalian. In one embodiment said
  • Fab construct is human. In another embodiment said Fab construct is humanized. In yet another embodiment said Fab contruct comprises at least
  • said Fab construct is a single chain Fab (scFab).
  • the CD3 binding polypeptide constructs comprise CD3 binding scFab constructs wherein the C-terminus of the Fab light chain is connected to the N-terminus of the Fab heavy chain by a peptide linker.
  • the peptide linker allows arrangement of the Fab heavy and light chain to form a functional CD3 binding moiety.
  • the peptide linkers suitable for connecting the Fab heavy and light chain include sequences comprising glycine-serine linkers for instance, but not limited to (G m S) n -GG, (SGn)m, (SEGn)m, wherein m and n are between 0-20.
  • the scFab construct is a cross-over construct wherein the constant regions of the Fab light chain and the Fab heavy chain are exchanged.
  • the variable regions of the Fab light chain and the Fab heavy chain are exchanged.
  • the CD3 binding polypeptide constructs comprise CD3 binding Fv constructs (i.e. antigen binding constructs comprising a heavy and a light chain, each comprising a variable region).
  • said Fv construct is mammalian.
  • said Fv construct is human.
  • said Fv construct is humanized.
  • said Fv contruct comprises at least one of human heavy and light chain variable regions.
  • said Fv construct is a single chain Fv (scFv).
  • the CD3 binding polypeptide construct of a multispecific heteromultimer construct described herein bind to at least one component of the CD3 complex.
  • the CD3 binding polypeptide construct binds to at least one of CD3 epsilon, CD3 gamma, CD3 delta or CD3 zeta of the CD3 complex.
  • the CD3 binding polypeptide construct binds the CD3epsilon domain.
  • binding polypeptide construct binds a human CD3 complex.
  • the CD3 binding polypeptide construct exhibits cross- species binding to a least one member of the CD3 complex.
  • multispecific heteromultimer constructs comprising at least one CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell, where in the CD3 expressing cell is a T-cell.
  • the CD3 expressing cell is a human cell.
  • the CD3 expressing cell is a non-human, mammalian cell.
  • the T cell is a cytotoxic T cell. In some embodiments the T cell is a CD4 + or a CD8 + T cell.
  • the construct is capable of activating and redirecting cytotoxic activity of a T cell to a target cell such as a B cell.
  • said redirection is independent of MHC-mediated peptide antigen presentation by the target cell and and/or specificity of the T cell.
  • hetromultimer constructs that are capable of simultaneous binding to a B cell antigen for instance a tumor cell antigen, and an activating T cell antigen.
  • the heteromultimer construct is capable of crosslinking a T cell and a target B cell by simultaneous binding to a B cell antigen for instance CD19 or CD20 and an activating T cell antigen for instance CD3.
  • the simultaneous binding results in lysis of a target B cell, for instance a tumor cell. In one embodiment, such simultaneous binding results in activation of the T cell.
  • such simultaneous binding results in a cellular response of a T lymphocyte, for instance a cytotoxic T lymphocyte, selected from the group of: proliferation, differentiation, cytokine secretion, cytotoxic effector molecule release, cytotoxic activity, and expression of activation markers.
  • a T lymphocyte for instance a cytotoxic T lymphocyte, selected from the group of: proliferation, differentiation, cytokine secretion, cytotoxic effector molecule release, cytotoxic activity, and expression of activation markers.
  • the antigen binding polypeptide construct binds to a target antigen on at least one B cell.
  • the antigen binding polypeptide construct binds at least one member of a B cell CD21 -CD19-CD81 complex.
  • the antigen binding polypeptide construct comprises at least one CD19 binding domain or fragment thereof.
  • the antigen binding polypeptide construct comprises at least one CD20 binding domain.
  • the at least one antigen binding domain is a CD19 or CD20 binding domain which is obtained from a CD19 or CD20 specific antibody, a nanobody, fibronectin, affibody, anticalin, cysteine knot protein, DARPin, avimer, Kunitz domain or variant or derivative thereof.
  • the at least one antigen binding polypeptide construct described herein comprises at least one antigen binding domain which is a CD19 or CD20 binding domain from an antibody which is a heavy chain antibody devoid of light chains.
  • the at least one antigen binding domain is a CD19 or CD20 binding domain that comprises at least one amino acid modification that reduces immunogenicity as compared to a corresponding antigen binding domain not comprising said modification.
  • the at least one antigen binding domain is a CD19 or CD20 binding domain comprising at least one amino acid modification that increases its stability as measured by T m , as compared to a corresponding domain not comprising said modification.
  • polypeptide construct is a Fab construct that binds at least one of CD19 and
  • said Fab construct is mammalian. In one embodiment said Fab construct is human. In another embodiment said Fab construct is humanized. In yet another embodiment said Fab contruct comprises at least one of human heavy and light chain constant regions. In a further embodiment said Fab construct is a single chain Fab (scFab).
  • construct comprises a scFab construct wherein the C-terminus of the Fab light chain is connected to the N-terminus of the Fab heavy chain by a peptide linker.
  • the peptide linker allows arrangement of the Fab heavy and light chain to form a functional CD19 and/or CD20 binding moiety.
  • the peptide linkers suitable for connecting the Fab heavy and light chain include sequences comprising glycine-serine linkers for instance, but not limited to (G m S) n -GG, (SG n ) m , (SEG n ) m , wherein m and n are between 0-20.
  • the scFab construct is a cross-over construct wherein the constant regions of the Fab light chain and the Fab heavy chain are exchanged. In another embodiment of a cross-over Fab, the variable regions of the Fab light chain and the Fab heavy chain are exchanged.
  • polypeptide construct is a Fv construct that binds at least one of CD19 and CD20 on a B cell.
  • said Fv construct is mammalian.
  • said Fv construct is human.
  • said Fv construct is humanized.
  • said Fv contruct comprises at least one of human heavy and light chain variable regions.
  • said Fv construct is a single chain Fv (scFv).
  • the antigen binding polypeptide construct exhibits cross-species binding to a least one antigen expressed on the surface of a B cell.
  • the antigen binding polypeptide construct of a multispecific heteromultimer construct described herein bind to at least one of mammalian CD19 and CD20.
  • binding polypeptide construct binds a human CD19 or CD20.
  • hetromultimer constructs that are capable of simultaneous binding to a B cell antigen for instance a tumor cell antigen, and an activating T cell antigen.
  • the heteromultimer construct is capable of crosslinking a T cell and a target B cell by simultaneous binding to a B cell antigen for instance CD19 or CD20 and an activating T cell antigen for instance CD3.
  • the disease comprises at least one antigen binding polypeptide construct that binds to a target antigen such as a CD19 or CD20 on at least one B cell associated with a disease.
  • the disease is a cancer selected from a carcinoma, a sarcoma, leukaemia, lymphoma and glioma.
  • the cancer is at least one of squamous cell carcinoma, adenocarcinoma, transition cell carcinoma, osteosarcoma and soft tissue sarcoma.
  • the at least one B cell is an autoimmune reactive cell that is a lymphoid or myeloid cell.
  • an albumin or immunoglobulin based multispecific heteromultimer construct described herein further comprises at least one binding domain that binds at least one of: EpCAM, EGFR, IGFR, HER-2 neu, HER-3, HER-4, PSMA, CEA, MUC-1 (mucin), MUC2, MUC3, MUC4, MUC5, MUC7, CCR4, CCR5, CD19, CD20, CD33, CD30, ganglioside GD3, 9-0-Acetyl-GD3, GM2, Poly SA, GD2, Carboanhydrase IX (MN/CA IX), CD44v6, Sonic Hedgehog (Shh), Wue-1 , Plasma Cell Antigen, (membrane- bound), Melanoma Chondroitin Sulfate Proteoglycan (MCSP), CCR8, TNF- alpha precursor, STEAP, mesothelin, A33 Antigen, Prostate Stem Cell Antigen (PSCA), Ly-6;
  • multispecific heteromultimer constructs described herein which are differentially modified during or after translation.
  • the modification is at least one of: glycosylation,
  • the heteromultimer construct is chemically modified by known techniques, including but not limited, to specific chemical cleavage by cyanogen bromide, trypsin, chymotrypsin, papain, V8 protease, NaBH 4 ; acetylation, formylation, oxidation, reduction; and metabolic synthesis in the presence of tunicamycin.
  • Additional post-translational modifications of heteromultimers described herein include, for example, N-linked or O-linked carbohydrate chains, processing of N-terminal or C-terminal ends), attachment of chemical moieties to the amino acid backbone, chemical modifications of N-linked or O- linked carbohydrate chains, and addition or deletion of an N-terminal methionine residue as a result of procaryotic host cell expression.
  • the heteromultimer constructs described herein are modified with a detectable label, such as an enzymatic, fluorescent, isotopic or affinity label to allow for detection and isolation of the protein.
  • examples of suitable enzyme labels include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase;
  • examples of suitable prosthetic group complexes include streptavidin biotin and avidin/biotin;
  • examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol;
  • bioluminescent materials include luciferase, luciferin, and aequorin
  • suitable radioactive material include iodine, carbon, sulfur, tritium, indium, technetium, thallium, gallium, palladium, molybdenum, xenon, fluorine.
  • heteromultimer constructs described herein are attached to macrocyclic chelators that associate with radiometal ions.
  • heteromultimer constructs described herein are modified by either natural processes, such as post-translational
  • polypeptides from heteromultimers described herein are branched, for example, as a result of ubiquitination, and in some embodiments are cyclic, with or without branching. Cyclic, branched, and branched cyclic polypeptides are a result from posttranslation natural processes or made by synthetic methods.
  • Modifications include acetylation, acylation, ADP- ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination.
  • heteromultimeric constructs described herein are attached to solid supports, which are particularly useful for immunoassays or purification of polypeptides that are bound by, that bind to, or associate with albumin fusion proteins of the invention.
  • solid supports include, but are not limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene.
  • RNA for example, in the form of messenger RNA (mRNA).
  • RNA of the present invention may be single stranded or double stranded.
  • a set of expression vectors for expressing a multispecific heteromultimer construct described herein which comprises a first and a second polypeptide construct, said set comprising at least a first DNA sequence encoding said first polypeptide construct and at least a second DNA sequence encoding said second polypeptide construct.
  • polynucleotide sequences encoding a heteromultimer construct described herein or a polypeptide construct thereof with sequence as provided herein.
  • a polynucleotide comprising a sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%,
  • nucleotide sequence shown in Figure 98%, or 99% identical to a nucleotide sequence shown in Figure .
  • kits for producing an expression product containing a multispecific heteromultimer construct as described herein, in stable mammalian cells comprising: transfecting at least one mammalian cell with: at least a first DNA sequence encoding said first polypeptide construct and at least a second DNA sequence encoding said second polypeptide construct, such that said at least one first DNA sequence, said at least one second DNA sequence are transfected in said at least one
  • said predetermined ratio of the at least one first DNA sequence: at least one second DNA sequence is about 1 :1 . In certain other embodiments, said predetermined ratio of the at least one first DNA sequence: at least one second DNA sequence is skewed towards a larger amount of the one first DNA sequence such as about 2:1. In yet other embodiments, said predetermined ratio of the at least one first DNA sequence: at least one second DNA sequence is skewed towards a larger amount of the one first DNA sequence such as about 1 :2.
  • the mammalian cell is selected from the group consisting of a VERO, HeLa, HEK, NSO, Chinese Hamster Ovary (CHO), W138, BHK, COS-7, Caco-2 and MDCK cell, and subclasses and variants thereof.
  • heteromultimers produced as recombinant molecules by secretion from yeast, a microorganism such as a bacterium, or a human or animal cell line.
  • the polypeptides are secreted from the host cells.
  • Embodiments include a cell, such as a yeast cell transformed to express a heteromultimer protein described herein.
  • a cell such as a yeast cell transformed to express a heteromultimer protein described herein.
  • transformed host cells themselves, are provided culture of those cells, preferably a monoclonal (clonally homogeneous) culture, or a culture derived from a monoclonal culture, in a nutrient medium. If the polypeptide is secreted, the medium will contain the polypeptide, with the cells, or without the cells if they have been filtered or centrifuged away.
  • Many expression systems are known and may be used, including bacteria (for example E. coli and Bacillus subtilis), yeasts (for example Saccharomyces cerevisiae, Kluyveromyces lactis and Pichia pastoris, filamentous fungi (for example Aspergillus), plant cells, animal cells and insect cells.
  • a heteromultimer described herein is produced in conventional ways, for example from a coding sequence inserted in the host chromosome or on a free plasmid.
  • the yeasts are transformed with a coding sequence for the
  • Successfully transformed cells i.e., cells that contain a DNA construct of the present invention
  • cells resulting from the introduction of an expression construct can be grown to produce the desired polypeptide.
  • Cells can be harvested and lysed and their DNA content examined for the presence of the DNA using a method such as that described by Southern (1975) J. Mol. Biol. 98, 503 or Berent et al. (1985) Biotech. 3, 208.
  • the presence of the protein in the supernatant can be detected using antibodies.
  • Useful yeast plasmid vectors include pRS403-406 and pRS413-416 and are generally available from Stratagene Cloning Systems, La Jolla, Calif. 92037, USA.
  • Plasmids pRS403, pRS404, pRS405 and pRS406 are Yeast Integrating plasmids (Yips) and incorporate the yeast selectable markers HIS3, 7RP1 , LEU2 and URA3.
  • Plasmids pRS413-416 are Yeast Centromere plasmids (Ycps).
  • a variety of methods have been developed to operably link DNA to vectors via complementary cohesive termini. For instance, complementary homopolymer tracts can be added to the DNA segment to be inserted to the vector DNA. The vector and DNA segment are then joined by hydrogen bonding between the complementary honmopolymeric tails to form
  • Synthetic linkers containing one or more restriction sites provide an alternative method of joining the DNA segment to vectors.
  • the DNA segment, generated by endonuclease restriction digestion, is treated with bacteriophage
  • T4 DNA polymerase or E. coli DNA polymerase 1 enzymes that remove protruding, _single-stranded termini with their 3' 5'-exonucleolytic activities, and fill in recessed 3'-ends with their polymerizing activities.
  • the products of the reaction are DNA segments carrying polymeric linker sequences at their ends. These DNA segments are then cleaved with the appropriate restriction enzyme and ligated to an expression vector that has been cleaved with an enzyme that produces termini compatible with those of the DNA segment.
  • Exemplary genera of yeast contemplated to be useful in the practice of the present invention as hosts for expressing the albumin, fusion proteins are Pichua (formerly classified as Hansenula), Saccharomyces, Kluyveromyces, Aspergillus, Candida, Torulopsis, Torulaspora,
  • Schizosaccharomyces Citeromyces, Pachysolen, Zygosaccharomyces, Debaromyces, Trichoderma, Cephalosporium, Humicola, Mucor, Neurospora, Yarrowia, Metschunikowia, Rhodosporidium, Leucosporidium, Botryoascus, Sporidiobolus, Endomycopsis, and the like.
  • Preferred genera are those selected from the group consisting of Saccharomyces, Schizosaccharomyces, Kluyveromyces, Pichia and Torulaspora. Examples of Saccharomyces spp. are S. cerevisiae, S. italicus and S. rouxii.
  • Kluyveromyces spp. are K. fragilis, K. lactis and K. marxianus.
  • a suitable Torulaspora species is T. delbrueckii.
  • Examples of Pichia (Hansenula) spp. are P. angusta (formerly H. polymorpha), P. anomala
  • Exemplary species of Saccharomyces useful for the synthesis of heteromultimer constructs described herein include S. cerevisiae, S. italicus, S. diastaticus, and Zygosaccharomyces rouxii.
  • Kluyveromyces include K. fragilis and K. lactis.
  • Hansenula include H. polymorpha (now Pichia angust
  • Pichia anomala and Pichia capsulata. Additional preferred exemplary species of Pichia include P. pastoris. Preferred exemplary species of Aspergillusinclude
  • Yarrowia include Y. lipolytica.
  • Many preferred yeast species are available from the ATCC.
  • the following preferred yeast species are available from the ATCC and are useful in the expression of albumin fusion proteins: Saccharomyces cerevisiae, Hansen, teleomorph strain BY4743 yap3 mutant (ATCC Accession
  • Suitable promoters for S. cerevisiae include those associated with the PGKI gene, GAL1 or GALI O genes, CYCI, PH05, TRP1 , ADH1 , ADH2, the genes for glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, triose phosphate isomerase,
  • phosphoglucose isomerase glucokinase
  • alpha-mating factor pheromone [a mating factor pheromone]
  • the PRBI promoter the GUT2 promoter
  • the GPDI promoter the GPDI promoter
  • hybrid promoters involving hybrids of parts of 5' regulatory regions with parts of 5' regulatory regions of other promoters or with upstream activation sites (e.g. the promoter of EP-A-258 067).
  • pombe are the thiamine-repressible promoter from the nmt gene as described by Maundrell (1990) J. Biol. Chem. 265, 10857-10864 and the glucose repressible jbpl gene promoter as described by Hoffman & Winston (1990) Genetics 124, 807-816.
  • Suitable promoters include AOX1 and AOX2.
  • Gleeson et al. (1986) J. Gen. Microbiol. 132, 3459-3465 include information on Hansenula vectors and transformation, suitable promoters being MOX1 and FMD1 ; whilst EP 361 991 , Fleer et al. (1991 ) and other publications from Rhone-Poulenc Rorer teach how to express foreign proteins in Kluyveromyces spp., a suitable promoter being PGKI.
  • the transcription termination signal is preferably the 3' flanking sequence of a eukaryotic gene which contains proper signals for transcription termination and polyadenylation.
  • Suitable 3' flanking sequences may, for example, be those of the gene naturally linked to the expression control sequence used, i.e. may correspond to the promoter. Alternatively, they may be different in which case the termination signal of the S. cerevisiae ADHI gene is preferred.
  • the desired heteromultimer protein is initially expressed with a secretion leader sequence, which may be any leader effective in the yeast chosen.
  • leader sequence useful in S. cerevisiae include that from the mating factor alpha polypeptide (MFa-1 ) and the hybrid leaders of EP-A- 387 319. Such leaders (or signals) are cleaved by the yeast before the mature albumin is released into the surrounding medium. Further such leaders include those of S. cerevisiae invertase (SUC2) disclosed in JP 62-096086 (granted as 91 1036516), acid phosphatase (PH05), the pre-sequence of MFa-1 , 0
  • MEL1 carlsbergensis a-galactosidase
  • K lactis killer toxin
  • Candida glucoarnylase
  • vectors containing polynucleotides encoding a heteromultimer construct described herein, host cells, and the production of the heteromultimer proteins by synthetic and recombinant techniques may be, for example, a phage, plasmid, viral, or retroviral vector. Retroviral vectors may be replication competent or replication defective. In the latter case, viral propagation generally will occur only in complementing host cells.
  • heteromultimer proteins described herein are joined to a vector containing a selectable marker for propagation in a host.
  • a plasmid vector is introduced in a precipitate, such as a calcium phosphate precipitate, or in a complex with a charged lipid. If the vector is a virus, it may be packaged in vitro using an appropriate packaging cell line and then transduced into host cells.
  • the polynucleotide insert is operatively linked to an appropriate promoter, such as the phage lambda PL promoter, the
  • E. coli lac, trp, phoA and rac promoters the SV40 early and late promoters and promoters of retroviral LTRs, to name a few.
  • Other suitable promoters will be known to the skilled artisan.
  • the expression constructs will further contain sites for transcription initiation, termination, and, in the transcribed region, a ribosome binding site for translation.
  • the coding portion of the transcripts expressed by the constructs will preferably include a translation initiating codon at the beginning and a termination codon (UAA, UGA or UAG) appropriately positioned at the end of the polypeptide to be translated.
  • the expression vectors will preferably include at least one selectable marker.
  • markers include dihydrofolate reductase, G418, glutamine synthase, or neomycin resistance for eukaryotic cell culture, and tetracycline, kanamycin or ampicillin resistance genes for culturing in E. coli and other bacteria.
  • Representative examples of appropriate hosts include, but are not limited to, bacterial cells, such as E. coli, Streptomyces and Salmonella
  • WSGF typhimurium cells
  • fungal cells such as yeast cells
  • insect cells such as Drosophila S2 and Spodoptera Sf9 cells
  • animal cells such as CHO, COS, NSO, 293, and Bowes melanoma cells
  • plant cells Appropriate culture mediums and conditions for the above-described host cells are known in the art.
  • vectors preferred for use in bacteria include pQE70, pQE60 and pQE-9, available from QIAGEN, Inc.; pBluescript vectors, Phagescript vectors, pNH8A, pNH16a, pNH18A; pNH46A, available from Stratagene Cloning Systems, Inc.; and ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 available from Pharmacia Biotech, Inc.
  • preferred eukaryotic vectors are pWLNEO, pSV2CAT, pOG44, pXT1 and pSG available from Stratagene; and pSVK3, pBPV, pMSG and pSVL available from Pharmacia.
  • Preferred expression vectors for use in yeast systems include, but are not limited to pYES2, pYD1 , pTEF1/Zeo, pYES2/GS, pPICZ, pGAPZ, pGAPZalph, pPIC9, pPIC3.5, pHIL-D2, pHIL-S1 , pPIC3.5K, pPIC9K, and PAO815 (all available from Invitrogen, Carlbad, CA).
  • Other suitable vectors will be readily apparent to the skilled artisan.
  • polynucleotides encoding a multispecific heteromultimer construct described herein are fused to signal sequences that will direct the localization of a protein of the invention to particular
  • a prokaryotic or eukaryotic cell For example, in E. coli, one may wish to direct the expression of the protein to the periplasmic space.
  • signal sequences or proteins (or fragments thereof) to which the heteromultimeric proteins are fused in order to direct the expression of the polypeptide to the periplasmic space of bacteria include, but are not limited to, the pelB signal sequence, the maltose binding protein (MBP) signal sequence, MBP, the ompA signal sequence, the signal sequence of the periplasmic E. coli heat-labile enterotoxin B-subunit, and the signal sequence of alkaline phosphatase.
  • MBP maltose binding protein
  • ompA signal sequence the signal sequence of the periplasmic E. coli heat-labile enterotoxin B-sub
  • polynucleotides albumin fusion proteins of the invention may be fused to the pelB pectate lyase signal sequence to increase the efficiency of expression and purification of such polypeptides in Gram-negative bacteria. See, U.S. Pat. Nos. 5,576,195 and 5,846,818, the contents of which are herein incorporated by reference in their entireties.
  • Examples of signal peptides that are fused to a heteromultimeric protein in order to direct its secretion in mammalian cells include, but are not limited to, the MPIF-1 signal sequence (e.g., amino acids 1 -21 of GenBank
  • a suitable signal sequence that may be used in conjunction with baculoviral expression systems is the gp67 signal sequence (e.g., amino acids 1 -19 of GenBank Accession Number AAA72759).
  • Vectors which use glutamine synthase (GS) or DHFR as the selectable markers can be amplified in the presence of the drugs methionine sulphoximine or methotrexate, respectively.
  • An advantage of glutamine synthase based vectors are the availably of cell lines (e.g., the murine myeloma cell line, NSO) which are glutamine synthase negative.
  • Glutamine synthase expression systems can also function in glutamine synthase expressing cells (e.g., Chinese Hamster Ovary (CHO) cells) by providing additional inhibitor to prevent the functioning of the endogenous gene.
  • a glutamine synthase expression system and components thereof are detailed in
  • glutamine synthase expression vectors can be obtained from Lonza Biologies, Inc. (Portsmouth, N.H.). Expression and production of monoclonal antibodies using a GS expression system in murine myeloma cells is described in Bebbington et al., Bio/technology 10:169(1992)
  • host cells containing vector constructs described herein and additionally host cells containing nucleotide sequences that are operably associated with one or more heterologous control regions (e.g., promoter and/or enhancer) using techniques known of in the art.
  • the host cell can be a higher eukaryotic cell, such as a mammalian cell (e.g., a human derived cell), or a lower eukaryotic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell.
  • a host strain may be chosen which modulates the expression of the inserted gene sequences, or modifies and processes the gene product in the specific fashion desired.
  • Expression from certain promoters can be elevated in the presence of certain inducers; thus expression of the genetically engineered polypeptide may be controlled.
  • different host cells have characteristics and specific mechanisms for the translational and post-translational processing and modification (e.g., phosphorylation, cleavage) of proteins. Appropriate cell lines can be chosen to ensure the desired modifications and processing of the foreign protein expressed.
  • nucleic acids and nucleic acid constructs of the invention into the host cell can be effected by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection, or other methods. Such methods are described in many standard laboratory manuals, such as Davis et al., Basic Methods In Molecular Biology (1986). It is specifically contemplated that the polypeptides of the present invention may in fact be expressed by a host cell lacking a recombinant vector.
  • the invention also encompasses primary, secondary, and immortalized host cells of vertebrate origin, particularly mammalian origin, that have been engineered to delete or replace endogenous genetic material (e.g., the coding sequence corresponding to a Cargo
  • Cargo polypeptide and/or to include genetic material.
  • the genetic material operably associated with the endogenous polynucleotide may activate, alter, and/or amplify endogenous polynucleotides.
  • heterologous polynucleotides e.g., polynucleotides encoding an albumin protein, or a fragment or variant thereof
  • heterologous control regions e.g., promoter and/or enhancer
  • endogenous polynucleotide sequences encoding a Therapeutic protein via homologous recombination (see, e.g., U.S. Pat. No. 5,641 ,670, issued Jun. 24, 1997; International
  • Heteromultimer proteins described herein can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography such as with protein A, hydroxylapatite chromatography, hydrophobic charge interaction
  • HPLC high performance liquid chromatography
  • heteromultimer proteins of the invention are purified using Anion Exchange Chromatography including, but not limited to, chromatography on Q-sepharose, DEAE sepharose, poros HQ, poros DEAF, Toyopearl Q, Toyopearl QAE, Toyopearl DEAE, Resource/Source Q and DEAE, Fractogel Q and DEAE columns.
  • Anion Exchange Chromatography including, but not limited to, chromatography on Q-sepharose, DEAE sepharose, poros HQ, poros DEAF, Toyopearl Q, Toyopearl QAE, Toyopearl DEAE, Resource/Source Q and DEAE, Fractogel Q and DEAE columns.
  • the proteins described herein are purified using Cation Exchange Chromatography including, but not limited to, SP- sepharose, CM sepharose, poros HS, poros CM, Toyopearl SP, Toyopearl CM,
  • heteromultimer proteins described herein can be chemically synthesized using techniques known in the art (e.g., see Creighton, 1983, Proteins: Structures and Molecular Principles, W. H. Freeman & Co., N.Y and Hunkapiller et al., Nature, 310:105-1 1 1 (1984)). For example, a
  • polypeptide corresponding to a fragment of a polypeptide can be synthesized by use of a peptide synthesizer.
  • nonclassical amino acids or chemical amino acid analogs can be introduced as a substitution or addition into the polypeptide sequence.
  • Non-classical amino acids include, but are not limited to, to the D-isomers of the common amino acids,
  • 2,4diaminobutyric acid alpha-amino isobutyric acid, 4aminobutyric acid, Abu, 2-amino butyric acid, g-Abu, e-Ahx, 6amino hexanoic acid, Aib, 2-amino isobutyric acid, 3-amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, ⁇ -alanine, fluoro-amino acids, designer amino acids such as ⁇ -methyl amino acids, Ca-methyl amino acids, Na-methyl amino acids, and amino acid analogs in general.
  • the amino acid can be D (dextrorotary) or L (levorotary).
  • heteromultimer constructs described herein can be assayed for functional activity (e.g., biological activity) using or routinely modifying assays known in the art, as well as assays described herein.
  • immunoassays known in the art can be used, including but not limited to, competitive and non-competitive assay systems using techniques such as radioimmunoassays, ELISA (enzyme
  • antibody binding is detected by detecting a label on the primary antibody.
  • the primary antibody is detected by detecting binding of a secondary antibody or reagent to the primary antibody.
  • the secondary antibody is labeled. Many means are known in the art for detecting binding in an immunoassay and are within the scope of the present invention.
  • a binding partner e.g., a receptor or a ligand
  • heteromultimer described herein is assayed, e.g., by means well-known in the art, such as, for example, reducing and non-reducing gel chromatography, protein affinity chromatography, and affinity blotting. See generally, Phizicky et al., Microbiol. Rev. 59:94-123 (1995).
  • the ability of physiological correlates of a heteromultimeric protein to bind to a substrate(s) of antigen binding polypeptide constructs of the heteromultimers described herein can be routinely assayed using techniques known in the art.
  • heteromultimers described herein are directed to antibody-based therapies which involve administering heteromultimers described comprising cargo polypeptide(s) which is an antibody, a fragment or variant of an antibody, to a patient for treating one or more of the disclosed diseases, disorders, or conditions.
  • Therapeutic compounds described herein include, but are not limited to, heteromultimers described herein, nucleic acids encoding heteromultimers described herein.
  • a proliferative disease a minimal residual cancer
  • a tumorous disease a tumorous disease
  • an inflammatory disease an inflammatory disease
  • immunological disorder an autoimmune disease, an infectious disease, viral disease, allergic reactions, parasitic reactions, graft-versus-host diseases or host-versus-graft diseases or cell malignancies, said method comprising administering to a subject in need of such a prevention, treatment or
  • the cancer is a solid tumor.
  • the solid tumor is one or more of sarcoma, carcinoma, and lymphoma.
  • the cancer is a hematological cancer.
  • the cancer is one or more of B-cell lymphoma, non-Hodgkin's lymphoma, and leukemia.
  • a method of treating cancer cells comprising providing to said cell a composition comprising a heteromultimer construct described herein. In some embodiments, the method further comprising providing said heteromultimer in conjugation with another therapeutic agent.
  • a method of treating a cancer non-responsive to blinatumomab in a mammal in need thereof comprising administering to the mammal a composition comprising an effective amount of the pharmaceutical composition comprising a heteromultimer construct described herein.
  • Aamethod of treating a cancer cell regressive after treatment with blinatumomab comprising providing to said cancer cell a composition comprising an effective amount of the pharmaceutical composition comprising a heteromultimer construct described herein.
  • composition comprising an effective amount of the pharmaceutical composition comprising a
  • the disease is not responsive to treatment with at least one of an anti-CD19 antibody and an anti-CD20 antibody.
  • the disease is a cancer or autoimmune condition resistant to CD19 or CD20 lytic antibodies
  • a method of treating an autoimmune condition in a mammal in need thereof comprising administering to said mammal a composition comprising an effective amount of the pharmaceutical composition described herein.
  • the autoimmune condition is one or more of multiple sclerosis, rheumatoid arthritis, lupus erytematosus, psoriatic arthritis, psoriasis, vasculitis, uveitis, Crohn's disease, and type 1 diabetes.
  • a method of treating an inflammatory condition in a mammal in need thereof comprising administering to said mammal a composition comprising an effective amount of the pharmaceutical composition comprising an heteromultimer described herein.
  • heteromultimers described herein, comprising at least a fragment or variant of an antibody may be administered alone or in combination with other types of treatments (e.g., radiation therapy, chemotherapy, hormonal therapy, immunotherapy and anti-tumor agents).
  • treatments e.g., radiation therapy, chemotherapy, hormonal therapy, immunotherapy and anti-tumor agents.
  • administration of products of a species origin or species reactivity in the case of antibodies
  • human antibodies, fragments derivatives, analogs, or nucleic acids are administered to a human patient for therapy or prophylaxis.
  • Gene therapy refers to therapy performed by the administration to a subject of an expressed or expressible nucleic acid.
  • the nucleic acids produce their encoded protein that mediates a therapeutic effect. Any of the methods for gene therapy available in the art can be used.
  • bispecific single chain antibodies described in the art have great therapeutic potential for the treatment of malignant diseases, most of these bispecific molecules are limited in that they are species specific and recognize only human antigen, and— due to genetic similarity— likely the chimpanzee counterpart.
  • the advantage of the present invention is the provision of a bispecific single chain antibody comprising a binding domain exhibiting cross-species specificity to human and non-chimpanzee primate of the CD3 epsilon chain.
  • heteromultimers or pharmaceutical compositions described herein are tested in vitro, and then in vivo for the desired therapeutic or prophylactic activity, prior to use in humans.
  • in vitro assays which can be used to determine whether administration of a specific compound is indicated, include in vitro cell culture assays in which a patient tissue sample is grown in
  • the heteromultimer is substantially purified (e.g., substantially free from substances that limit its effect or produce undesired side-effects).
  • the subject is an animal, including but not limited to animals such as cows, pigs, horses, chickens, cats, dogs, etc., and in certain embodiments, a mammal, and most preferably human.
  • Various delivery systems are known and can be used to administer a heteromultimer formulation described herein, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the compound, receptor-mediated endocytosis (see, e.g., Wu and
  • nucleic acid as part of a retroviral or other vector, etc.
  • Methods of introduction include but are not limited to intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes.
  • the compounds or compositions may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local.
  • intraventricular injection may be facilitated by an intraventricular catheter, for example, attached to a reservoir, such as an Ommaya reservoir.
  • Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent.
  • heteromultimers, or compositions described herein locally to the area in need of treatment may be achieved by, for example, and not by way of limitation, local infusion during surgery, topical application, e.g., in conjunction with a wound dressing after surgery, by injection, by means of a catheter, by means of a suppository, or by means of an implant, said implant being of a porous, non- porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.
  • care must be taken to use materials to which the protein does not absorb.
  • the heteromultimers or composition can be delivered in a vesicle, in particular a liposome (see Langer, Science 249:1527- 1533 (1990); Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353-365 (1989); Lopez-Berestein, ibid., pp. 317-327; see generally ibid.)
  • the heteromultimers or composition can be delivered in a controlled release system.
  • a pump may be used (see Langer, supra; Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al., Surgery 88:507 (1980); Saudek et al., N. Engl. J. Med. 321 :574 (1989)).
  • polymeric materials can be used (see Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla.
  • a controlled release system can be placed in proximity of the therapeutic target, e.g., the brain, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 1 15-138 (1984)).
  • the nucleic acid in a specific embodiment comprising a nucleic acid encoding a heteromultimer decribed herein, can be administered in vivo to promote expression of its encoded protein, by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by use of a retroviral vector (see U.S. Pat. No.
  • a nucleic acid can be introduced intracellular ⁇ and incorporated within host cell DNA for expression, by homologous recombination.
  • compositions comprise a therapeutically effective amount of a compound, and a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier for example
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
  • Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
  • the composition if
  • 87 WSGF desired can also contain minor amounts of wetting or
  • compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like.
  • the composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
  • Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E. W. Martin.
  • Such compositions will contain a therapeutically effective amount of the compound, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient.
  • the formulation should suit the mode of administration.
  • composition comprising the
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection.
  • the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • compositions described herein are formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include those formed with anions such as those derived from hydrochloric, phosphoric,
  • compositions described herein which will be effective in the treatment, inhibition and prevention of a disease or disorder associated with aberrant expression and/or activity of a Therapeutic protein can be determined by standard clinical techniques.
  • in vitro assays may optionally be employed to help identify optimal dosage ranges.
  • the precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each patient's circumstances. Effective doses are extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • a heteromultimer construct described herein is suitably administered to the patient at one time or over a series of treatments.
  • about 1 ⁇ g/kg to 15 mg/kg (e.g. 0.1 mg/kg - 10 mg/kg) of T cell activating bispecific antigen binding molecule can be an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion.
  • One typical daily dosage might range from about 1 ⁇ g/kg to 100 mg/kg or more, depending on the factors mentioned above.
  • the treatment would generally be sustained until a desired
  • a dosage of the heteromultimer described herein would be in the range from about 0.005 mg/kg to about 10 mg/kg.
  • a dose may also comprise from about 1 microgram/kg body weight, about 5 microgram/kg body weight, about 10 microgram/kg body weight, about 50 microgram/kg body weight, about 100 microgram/kg body weight, about 200 microgram/kg body weight, about 350 microgram/kg body weight, about 500 microgram/kg body weight, about 1 milligram/kg body weight, about 5 milligram/kg body weight, about 10
  • milligram/kg body weight about 200 milligram/kg body weight, about 350 milligram/kg body weight, about 500 milligram/kg body weight, to about 1000 mg/kg body weight or more per administration, and any range derivable therein.
  • a range of about 5 mg/kg body weight to about 100 mg/kg body weight, about 5 microgram kg body weight to about 500 milligram kg body weight, etc. can be administered, based on the numbers described above.
  • Such doses may be administered intermittently, e.g. every week or every three weeks (e.g. such that the patient receives from about two to about twenty, or e.g. about six doses of the T cell activating bispecific antigen binding molecule).
  • An initial higher loading dose, followed by one or more lower doses may be administered.
  • heteromultimers described herein are generally used in an amount effective to achieve the intended purpose.
  • a heteromultimer described herein, or pharmaceutical compositions thereof are administered or applied in a therapeutically effective amount. Determination of a therapeutically effective amount is well within the capabilities of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • a therapeutically effective dose can be estimated initially from in vitro assays, such as cell culture assays.
  • a dose can then be formulated in animal models to achieve a circulating concentration range that includes the IC 50 as determined in cell culture. Such information can be used to more accurately determine useful doses in humans.
  • Initial dosages can also be estimated from in vivo data, e.g., animal models, using techniques that are well known in the art.
  • in vivo data e.g., animal models
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the heteromultimer described herein which are sufficient to maintain therapeutic effect.
  • Usual patient dosages for administration by injection range from about 0.1 to 50 mg/kg/day, typically from about 0.5 to 1 mg/kg/day.
  • Therapeutically effective plasma levels may be achieved by administering multiple doses each day. Levels in plasma may be measured, for example, by HPLC.
  • the effective local concentration of the heteromultimer described herein may not be related to plasma concentration.
  • One having skill in the art will be able to optimize therapeutically effective local dosages without undue experimentation.
  • a therapeutically effective dose of the heteromultimer constructs described herein will generally provide therapeutic benefit without causing substantial toxicity.
  • Toxicity and therapeutic efficacy of a heteromultimer described herein can be determined by standard pharmaceutical procedures in cell culture or experimental animals. Cell culture assays and animal studies can be used to determine the LD 50 (the dose lethal to 50% of a population) and the
  • ED 50 the dose therapeutically effective in 50% of a population.
  • the dose ratio between toxic and therapeutic effects is the therapeutic index, which can be expressed as the ratio LD50/ED50.
  • T cell activating bispecific antigen binding molecules that exhibit large therapeutic indices are preferred.
  • the heteromultimer construct described herein according to the present invention exhibits a high therapeutic index.
  • the data obtained from cell culture assays and animal studies can be used in formulating a range of dosages suitable for use in humans.
  • the dosage lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity.
  • the dosage may vary within this range depending upon a variety of factors, e.g., the dosage form employed, the route of administration utilized, the condition of the subject, and the like.
  • 91 WSGF can be chosen by the individual physician in view of tr
  • the attending physician would also know to adjust treatment to higher levels if the clinical response were not adequate (precluding toxicity).
  • the magnitude of an administered dose in the management of the disorder of interest will vary with the severity of the condition to be treated, with the route of administration, and the like. The severity of the condition may, for example, be evaluated, in part, by standard prognostic evaluation methods. Further, the dose and perhaps dose frequency will also vary according to the age, body weight, and response of the individual patient.
  • Also provided is a process for the production of a pharmaceutical composition comprising a eteromutlimer described herein, said process comprising: culturing a host cell under conditions allowing the expression of a heteromultimer; recovering the produced heteromultimer from the culture; and producing the pharmaceutical composition.
  • the heteromultimer constructs described herein are administered in combination with one or more other agents in therapy.
  • a heteromultimer described herein is co-administered with at least one additional therapeutic agent.
  • therapeutic agent encompasses any agent administered to treat a symptom or disease in an individual in need of such treatment.
  • additional therapeutic agent may comprise any active ingredients suitable for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other.
  • an additional therapeutic agent is an immunomodulatory agent, a cytostatic agent,
  • the additional therapeutic agent is an anti-cancer agent, for example a microtubule disruptor, an antimetabolite, a topoisomerase inhibitor, a DNA intercalator, an alkylating agent, a hormonal therapy, a kinase inhibitor, a receptor antagonist, an activator of tumor cell apoptosis, or an antiangio genie agent.
  • an anti-cancer agent for example a microtubule disruptor, an antimetabolite, a topoisomerase inhibitor, a DNA intercalator, an alkylating agent, a hormonal therapy, a kinase inhibitor, a receptor antagonist, an activator of tumor cell apoptosis, or an antiangio genie agent.
  • Such other agents are suitably present in combination in amounts that are effective for the purpose intended.
  • the effective amount of such other agents depends on the amount of T cell activating bispecific antigen binding molecule used, the type of disorder or treatment, and other factors discussed above.
  • the heteromultimers described herein are generally used in the same dosages and with administration routes as described herein, or about from 1 to 99% of the dosages described herein, or in any dosage and by any route that is empirically/clinically determined to be appropriate.
  • Such combination therapies noted above encompass combined administration (where two or more therapeutic agents are included in the same or separate compositions), and separate administration, in which case, administration of the heteromultimer described herein can occur prior to, simultaneously, and/or following, administration of the additional therapeutic agent and/or adjuvant.
  • Heteromultimer constructs described herein can also be used in combination with radiation therapy.
  • an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of the disorders described above is provided.
  • the article of manufacture comprises a container and a label or package insert on or associated with the container.
  • Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition which is by itself or
  • the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • At least one active agent in the composition is a T cell activating bispecific antigen binding molecule of the invention.
  • the label or package insert indicates that the composition is used for treating the condition of choice.
  • the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises a heteromultimer described herein; and (b) a second container with a composition contained therein, wherein the composition comprises a further cytotoxic or otherwise therapeutic agent.
  • the article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the compositions can be used to treat a particular condition.
  • the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically- acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate- buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • Example 1 Bispecific CD3-CD19 scFvs fused to an asymmetric lgG1 Fc.
  • V873 belongs to a novel family of CD3- based bispecific azymetric lgG1 antibodies that can be expressed and purified with significantly higher yields in mammalian CHO cells compared to
  • V873 demonstrates unexpected effectontarget cell binding, bridging and target cell killing.
  • V873 and bispecific CD3-based azymetric antibodies have utility in targeted T cell mediated killing of diseased cells and hence may be useful for treating cancers and autoimmune and inflammatory diseases.
  • V873 is a bispecific CD3-CD19 scFvs fused to an azymetric lgG1 Fc.
  • v873 represents a novel bispecific azymetric antibody class comprising one anti-CD3 warhead and a second warhead comprising a cell surface antigen of a target cell, and an antibody Fc heterodimer comprising heterodimer lgG1 Fc.
  • the fusion of the CD3 warhead to chain A or B of the Fc is important for its druggable properties.
  • V874 and V875 are two other Bispecific CD3-CD19 scFvs fused to an asymmetric lgG1 Fc but have different CD3 amino acid compositions.
  • V873 shows unexpected good mammalian CHO expression and purification yields compared to the Amgen/Micromet blinatumomab CD3-CD19 BiTE tandem scFvs. V873 bridges T and B cells and results in potent killing of cultured human Burkitt lymphoma cells (Raji B-cell lymphoma line) using resting and IL-2 activated human PBMCs.
  • V873 and its related bispecific CD3-Azymetric antibodies differ from
  • V873 can bind FcRn and this heterodimeric Fc class shows typical antibody
  • V873 and related bispecific CD3-based constructs address known stability issues with tandem scFvs and generally recognized poor
  • V873 addresses the short PK of blinatumomab and CNS adverse effects owing to its wildtype FcRn binding affinity and MWT which restricts its distribution to the peripheral compartment.
  • the Azymetric heterodimer Fc confers additional tailored FcgR effector ADCC, CDC, and ADCP activities and hence efficacy to drug resistant tumors.
  • v873 to mediate PBMC (T cell)- B cell killing is highly unexpected.
  • the properties of blinatumomab bscCD19xCD3 and related BiTEs is reported to rely on the use of flexibly linked single-chain variable fragments (scFv) that are arranged in tandem joined by a short linker will allow for a much closer approximation of opposing cells than is possible with larger bispecific formats such as quadroma antibodies.
  • the flexible linkage is expected to enable free rotation and kinking of the 2 scFv arms, thereby facilitating the simultaneous recognition of 2 epitopes present on 2 opposing cell membranes and the formation of a cytolytic immunologic synapse.
  • bispecific CD3-CD19 construct as described herein with significantly different structural presentations of CD3 and CD19 scFv warheads on the heterodimer Fc can bind, bridge T and B cells, and mediate PBMC (T cell)-B cell killing.
  • Other applications include depletion of B cells in B cell driven autoimmune and inflammatory diseases such as RA, lupus, MS, IBD.
  • V873 related bispecific CD3-Based azymetric antibodies may be useful for diagnostic purposes.
  • Example 2 Design, expression and purification of heteromultimer constructs with a heterodimeric Fc.
  • FIG. 1A An exemplary schematic representation of an anti-CD3/anti-CD19 antibody is shown in Figure 1A.
  • v873, v874, v875 exemplify bispecific anti-CD3/ anti-CD19 heterodimeric Fc constructs and were prepared and tested as described below.
  • the description includes a reference to BiTE, it refers to the antibody construct having an identical amino acid sequence to either the VH or VL of the anti-CD3 anti-CD19 BiTE molecule with or without modifications to variable heavy and light chain orientation (e.g. VH-VL) as indicated below.
  • v873 has a anti-CD19 BiTE (VL-VH) scFv on chain A and a CD3 BiTETM (VH-VL) scFv on chain B of the heterodimer Fc with the following mutations L351Y_F405A_Y407V on chain A and T366L_K392M_T394W on chain B.
  • VL-VH anti-CD19 BiTE
  • VH-VL CD3 BiTETM
  • V874 has a anti-CD19 BiTETM (VL-VH) scFv on chain A and the CD3 BiTETM (VLVH) scFv on chain B of the heterodimer Fc with the following mutations L351Y_F405A_Y407V on chain A and T366L_K392M_T394W on chain B.
  • VL-VH anti-CD19 BiTETM
  • VLVH CD3 BiTETM
  • V875 has a anti-CD19 BiTETM (VL-VH) scFv on chain A and the CD3 OKT3 (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations L351Y_F405A_Y407V on chain A and T366L_K392M_T394W on chain B.
  • VL-VH anti-CD19 BiTETM
  • VL-VH CD3 OKT3
  • v1379 has a anti-CD19 BiTETM (VL-VH) scFv on chain A and a anti- CD3 (VH-VL) BiTE on chain B of the heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and
  • v865, v866, v867, v868 exemplify monospecific anti-CD3 or CD19 bivalent scFv-Fc constructs and were prepared and tested as described below:
  • v865 has a anti-CD19 BiTE (VL-VH) scFv
  • v866 has a anti-CD3 BiTETM (VH-VL) on chain A and a anti-CD3 BiTETM (VH-VL) scFv on chain with a WT Fc (105kDa).
  • VH-VL anti-CD3 BiTETM
  • VH-VL anti-CD3 BiTETM
  • WT Fc 105kDa
  • v867 has a anti-CD3 BiTETM (VL-VH) scFv on chain A and a anti- CD3 BiTETM (VL-VH) scFv on chain B with a WT Fc (105kDa).
  • VL-VH anti-CD3 BiTETM
  • VL-VH anti- CD3 BiTETM
  • WT Fc 105kDa
  • v868 has a anti-CD3 OKT3 (VL-VH) scFv on chain A and a anti- CD3 OKT3 VL-VH scFv on chain with a WT Fc (105 kDa).
  • VL-VH anti-CD3 OKT3
  • WT Fc 105 kDa
  • v869 has a anti-CD19 BiTETM scFv (VL-VH) on chain A and a Fc on chain B of the heterodimer Fc with the following mutations
  • v870 has a anti- CD3 BiTETM (VH-VL) scFv on chain B and a Fc on chain A of the heterodimer Fc with the following mutations
  • v871 has a anti-CD3 BiTETM (VL-VH) scFv on chain B and a Fc on chain A of the heterodimer Fc with the following mutations
  • v872 has a anti-CD3 OKT3 (VL-VH) scFv on chain B and a Fc on chain A of the heterodimer Fc with the following mutations
  • v891 has the identical sequence blinatumc
  • BiTE scFv and anti-CD19 BiTE scFv 50 kDa [Polypeptide sequence corresponds to SEQ ID No: 90
  • v1380 has a anti-CD19 BiTETM (VL-VH) scFv on chain A and a anti-CD3 VHVL BiTETM on chain B of the heterodimer Fc with the following mutations L234A_L235A_T350V_L351Y_F405A_Y407V on chain A and L234A_L235A_T350V_T366L_K392L_T394W on chain B (corresponding to polypeptide sequences SEQ ID NOs:93 and 94).
  • v1381 has a anti-CD19 BiTETM (VI-VH) scFv on chain A and a anti-
  • N297A_T350V_T366L_K392L_T394W on chain B (corresponding to polypeptide sequences SEQ ID NOs: 93 and 98)
  • the following variants contain mutations to the anti-CD3 scFv that include either changes to the linker length, VH-VL orientations, or point mutations to improve stability and yeild.
  • v1653 has a anti-CD19 BiTETM (VL-VH) scFv on chain A and a anti- CD3 OKT3 (VL-VH) (with C to S mutation at position 100A of the VH CDR3) on chain B of the heterodimer Fc with the following mutations
  • v1654 has a anti-CD19 BiTETM scFv on chain A and a anti-CD3
  • OKT3 OKT3 (VH-VL) with 18 amino acid linker chain B of the heterodimer Fc with the following mutations T350V_L351 Y_F405A_Y407V on chain A and
  • v1655 has a anti-CD19 BiTE ' scFv on cr
  • OKT3 OKT3 (VH-VL) with 10 amino acid linker chain B of the heterodimer Fc with the following mutations T350V_L351 Y_F405A_Y407V on chain A and T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 109 and 1 10)
  • v1656 has a anti-CD3 BiTETM VHVL scFv on chain A and a anti- CD19 BiTETM (VL-VH) on chain B of the heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and
  • v1657 has a anti-CD3 OKT3 (VL-VH) scFv (with C to S mutation at position 100A of the VH CDR3) on chain A and a anti-CD19 BiTETM (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations
  • v1658 has a anti-CD3 OKT3 (VH-VL) scFv with 18 amino acid linker on chain A and a anti-CD19 BiTETM (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 121 and 122)
  • v1659 has a anti-CD3 OKT3 (VH-VL) scFv with 10 amino acid linker on chain A and a anti-CD19 BiTETM (VL_VH) scFv on chain B of the heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 125 and 126)
  • v1660 has a anti-CD3 OKT3 (VH-VL) scFv with 19 amino acid linker on chain A and a anti-CD19 BiTETM (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 129 and 130)
  • the following variants are Fc knoock outs that contain mutations to the anti-CD3 scFv that include either changes to the linker length, VH-VL orientations, or point mutations to improve stability and yeild.
  • v1661 has a anti-CD19 BiTETM (VL-VH) scFv on chain A and a anti- CD3 OKT3 (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations D265S_L234A_L235A_T350V_L351Y_F405A_Y407V on chain A and D265S_L234A_L235A_T350V_T366L_K392L_T394W on chain B.
  • v1662 has a anti-CD19 BiTETM (VL-VH) scFv on chain A and a anti- CD3 OKT3 (VL-VH) scFv (with C to S mutation at position 100A of the VH CDR3) on chain B of the heterodimer Fc with the following mutations
  • v1663 has a anti-CD19 BiTETM (VL-VH) scFv on chain A and a anti- CD3 OKT3 (VL-VH) scFv with 18 amino acid linker on chain B of the
  • v1664 has a anti-CD19 BiTETM (VL-VH) scFv on chain A and a anti- CD3 OKT3 (VH-VL) scFv with 10 amino acid linker on chain B of the
  • v1665 has a anti-CD3 BiTETM (VH-VL) scFv on chain A and a anti-CD3 BiTETM (VH-VL) scFv on chain A and a anti-CD3 BiTETM (VH-VL) scFv on chain A and a anti-CD3 BiTETM (VH-VL) scFv on chain A and a anti-CD3 BiTETM (VH-VL) scFv on chain A and a anti-CD3 BiTETM (VH-VL) scFv on chain A and a anti-CD3 BiTETM (VH-VL) scFv on chain A and a anti-CD3 BiTETM (VH-VL) scFv on chain A and a anti-CD3 BiTETM (VH-VL) scFv on chain A and a anti-CD3 BiTETM (VH-VL) scFv on chain A
  • v1666 has a anti-CD3 OKT3 (VH-VL) scFv with a 19 amino acid linker on chain A and a anti-CD19 BiTETM (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations
  • v1667 has a anti-CD19 BiTETM (VL-VH) scFv on chain A and a anti- CD3 OKT3 (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations N297A_T350V_L351Y_F405A_Y407V on chain A and
  • v1668 has a anti-CD19 BiTETM (VL-VH) scFv on chain A and a anti- CD3 OKT3 (VL-VH) scFv (with C to S mutation at position 100A of the VH CDR3) on chain B of the heterodimer Fc with the following mutations
  • v1669 has a anti-CD19 BiTETM (VL-VH) scFv on chain A and a anti- CD3 OKT3 (VH-VL) scFv with 18 amino acid linker on chain B of the
  • v1670 has a anti-CD19 BiTETM (VL-VH) scFv on chain A and a anti-
  • v1671 has a anti-CD3 BiTE I M (VH-VL) sc
  • v1672 has a anti-CD3 OKT3 (VH-VL) scFv with a 19 amino acid linker on chain A and a anti-CD19 BiTETM (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations
  • the following variants contain mutations to the anti-CD3 scFv that include either changes to the linker length, VH-VL orientations, or point mutations to improve stability and yeild.
  • v1673 has a anti-CD3 OKT3 (VL-VH) scFv (with C to S mutation at position 100A of the VH CDR3) on chain B and a Fc on chain A of the heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 169 and 170)
  • v1674 has a anti-CD3 OKT3 (VH-VL) scFv with 18 amino acid linker on chain B and a Fc on chain A of the heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and
  • v1798 has a anti-CD3 OKT3 (VH-VL) scFv with 10 amino acid linker on chain B and a Fc on chain A of the heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and
  • v1799 has a anti-CD3 OKT3 (VH-VL) scF
  • the following variants contain point mutations for disulfide stabilization at position 100 in the variable light and position 44 in the variable heavy chain (denoted 44-100SS).
  • v1800 has a anti-CD19 BiTETM (VL-VH) scFv on chain A and anti- CD3 OKT3 (VL-VH) 44-100SS on chain B of the heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and
  • v1801 has a anti-CD19 BiTETM (VL-VH) scFv on chain A and anti- CD3 OKT3 (VL-VH) (with C to S mutation at position 100A of the VH CDR3) 44-100SS on chain B of the heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 189 and 190)
  • v1802 has a anti-CD3 BiTETM (VH-VL) 44-100SS scFv on chain A and anti-CD19 BiTETM (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and
  • v4541 has a anti-CD3 BiTETM (VH-VL) 44-100SS scFv on chain A and anti-CD19 BiTETM (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations D265S_L234A_L235A_T350V_L351Y_F405A_Y407V on chain A and D265S_L234A_L235A_T350V_T366L_K392L_T394W on chain B. [Polypeptide sequences correspond to SEQ ID No: 50 and 52]
  • v4542 has a cyno/human cross-reactive anti-CD3 BiTETM 12C (VH- VL) scFv on chain A and cyno/human cross-reactive anti-CD19 MOR208 (VH- VL) scFv on chain B of the heterodimer Fc with the following mutations
  • v4543 has a cyno/human cross-reactive anti-CD3 BiTETM 12C (VH- VL) 44-100SS scFv on chain A and cyno/human cross -reactive anti-CD19 MOR208 (VH-VL) 44-100SS scFv on chain B of the heterodimer Fc with the following mutations D265S_L234A_L235A_T350V_L351Y_F405A_Y407V on chain A and D265S_L234A_L235A_T350V_T366L_K392L_T394W on chain B.
  • Polypeptide sequences correspond to SEQ ID No: 58 and 60]
  • v4544 has a cyno/human cross-reactive anti-CD3 BiTETM 12C (VH-VL) scFv on chain A and cyno/human cross-reactive anti-CD19 MOR208 (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations D265S_L234A_L235A_T350V_L351Y_F405A_Y407V on chain A and
  • v4545 has a cyno/human cross-reactive anti-CD3 BiTETM 12C (VH- VL) 44-100SS scFv on chain A and cyno/human cross -reactive anti-CD19 MOR208 (VL-VH) 44-100SS scFv on chain B of the heterodimer Fc with the following mutations D265S_L234A_L235A_T350V_L351Y_F405A_Y407V on chain A and D265S_L234A_L235A_T350V_T366L_K392L_T394W on chain B.
  • Polypeptide sequences correspond to SEQ ID No: 66 and 68]
  • v4546 has a cyno/human cross-reactive anti-CD3 BiTETM 12C (VH- VL) scFv on chain A and cyno/human cross-reactive anti-CD19 MDX-1342 (VH-VL) scFv on chain B of the heterodimer Fc with the following mutations D265S L234A L235A T350V L351Y F405A Y407V on chain A and
  • v4547 has a cyno/human cross-reactive anti-CD3 BiTETM 12C (VH- VL) 44-100SS scFv on chain A and cyno/human cross-reactive anti-CD19 MDX-1342 (VH-VL) 44-100SS scFv on chain B of the heterodimer Fc with the following mutations D265S_L234A_L235A_T350V_L351Y_F405A_Y407V on chain A and D265S_L234A_L235A_T350V_T366L_K392L_T394W on chain B. [Polypeptide sequences correspond to SEQ ID No: 74 and 76].
  • v4548 has a cyno/human cross-reactive anti-CD3 BiTETM 12C (VH- VL) scFv on chain A and cyno/human cross-reactive anti-CD19 MDX-1342 (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations D265S_L234A_L235A_T350V_L351Y_F405A_Y407V on chain A and
  • v4549 has a cyno/human cross-reactive anti-CD3 BiTETM 12C (VH- VL) 44-100SS scFv on chain A and cyno/human cross -reactive anti-CD19 MDX-1342 (VL-VH) 44-100SS scFv on chain B of the heterodimer Fc with the following mutations D265S_L234A_L235A_T350V_L351Y_F405A_Y407V on chain A and D265S_L234A_L235A_T350V_T366L_K392L_T394W on chain B.
  • Polypeptide sequences correspond to SEQ ID No: 82 and 84]
  • the antibodies and antibody controls were cloned and expressed as follows.
  • the genes encoding the antibody heavy and light chains were constructed via gene synthesis using codons optimized for human/mammalian expression.
  • the Fab sequences were generated from a known Her2/neu binding Ab (Carter P. et al. (1992) Humanization of an anti P185 Her2 antibody for human cancer therapy. Proc Natl Acad Sci 89, 4285.) and the Fc was an lgG1 isotype.
  • the scFv-Fc and OAA sequences, were generated from a known anti-CD3 and CD19 scFv Bite antibodies (Kipriyanov et. al., 1998, Int.
  • the CHO cells were transfected in exponential growth phase (1 .5 to 2 million cells/mL) with aqueous 1 mg/ml_ 25kDa polyethylenimine (PEI, Polysciences) at a PEhDNA ratio of 2.5:1 .(Raymond C. et al. A simplified polyethylenimine-mediated transfection process for large-scale and high- throughput applications. Methods. 55(1 ):44-51 (201 1 )).
  • the DNA was transfected in optimal DNA ratios of the heavy chain A (HC-A), light chain (LC), and heavy chain B that allow for heterodimer formation (e.g.
  • HC-A/HC-B/ ratios 50:50% (OAAs; HC/Fc), 50:50%.
  • Transfected cells were harvested after 5-6 days with the culture medium collected after centrifugation at 4000rpm and clarified using a 0.45 ⁇ filter.
  • the clarified culture medium was loaded onto a MabSelect SuRe (GE Healthcare) protein-A column and washed with 10 column volumes of PBS buffer at pH 7.2.
  • the antibody was eluted with 10 column volumes of citrate buffer at pH 3.6 with the pooled fractions containing the antibody neutralized with TRIS at pH 1 1.
  • the protein was finally desalted using an Econo-Pac 10DG column (Bio-Rad).
  • the protein was futher purified by protein L chromatography by the method as follows. Capto L resin PBS was equilibrated with PBS and protein A purified v875, neutralized with 1 M Tris, was added to resin and incubated at RT for 30 min. Resin washed with PBS and flow through collected, bound protein was eluted with 0.5 ml 0.1 M Glycine, pH 3.
  • the protein was further purified by gel filtration,
  • the SDS-PAGE in Figure 7 shows that all examplary heteromultimer can be expressed transiently in CH03E7 cells with a cell viability of > 80 %.
  • Example 3 Heteromultimer v873 is able to bridge Jurkat CD3 T cells and Raji CD19 B cells.
  • Pellets were resuspended in 2 ml of L10 + GS1 + NaN3 to a final concentration 5x x106 cells/ml.
  • each cell line was mixed together at the desired ratio, at a final concentration of 1x10 6 cells/ml.
  • T:T bridging was assessed with Jurkat-violet + Jurkat-FarRed
  • B:B was assessed with RAJI-violet + RAJI-FarRed
  • T:B bridging was assessed with Jurkat-violet + RAJI-FarRed.
  • Antibodies were diluted to 2x in L10+GS1 +NaN3 at room
  • Bridging % was calculated as the percentage of events that are simultaneously labeled violet and Far-red.
  • Figure 1 B shows the ability of v873 and blinatumomab CD19-CD3 BiTE (v891 , MT-103) to bridge Jurkat CD3 T cells (Top left quadrant) with Raji CD19 B cells (bottom right quadrant) by FACS. Bridged T-B cells appear in top right quadrant. This result demonstrates that at 300 nM, heteromultimer v873 is able to specifically bridge Jurkat T cells and Raji B cells to a similar extent (23% of total cells) as BiTE (21 % of total cells).
  • Example 4 Heteromultimers bind selectively to CD3- and CD19- expressing cells.
  • Figure 2 also demonstrates that the one-armed anti-CD3 antibody specifically binds to Jurkat T cells and does not cross-react to CD19 expressing B cells, and that the one- armed anti-CD19 antibody specifically binds to Raji B cells and does not cross- react to Jurkat T cells.
  • FIG. 9 shows that the FACS assay shows that v873 binds selectively to Jurkat T-cells and to Raji B-cells (Figure 9 B).
  • Figure 9A shows that human IgG (hlgG) does not bind to Jurkat T-cells and has low level binding to Raji B-cells, as expected due to the interactions between the hlgG Fc and CD32B on the Raji B-cells.
  • Figure 9A also shows that the anti-CD19 OAA binding selectively to the Raji B-cells and does not cross-react to Jurkat T cells.
  • the FACS assay was also carried out to confirm that v873 does not bind to control cell lines that do not express CD3 or CD19.
  • Figure 10 shows that v873 does not bind to the K562 cell line, which does not express CD19 or CD3.
  • Figure 1 1 shows that v873 also does not bind to mouse lymphoid cells which does not express CD19 or CD3.
  • Example 5 Heteromultimers mediate PBMC killing of target Raji B cells
  • v873 to mediate T cell cytoxicity against target Raji B-cells was measured using IL-2-stimulated PBMCs as follows.
  • CD4+ and CD8+ cells were then labeled and analyzed by cytometry to evaluate the contents of CD69+ cells in the preparations.
  • the PBMCs and the enriched fractions from the second day were used in the assay as is without IL- 2 activation or in resting state.
  • CFSE was used and tested as a differential label between Raji and autologous B cells.
  • Raji target cells were pre-labeled with minimal amounts of CFSE before the incubation with effectors, with or without test items.
  • the cell pellets were resuspended in various antibody cocktails for flow cytometry analysis.
  • a Guava 8HT flow cytometer was used for analysis of cell
  • Each condition tested included appropriate controls; wells with all effector and target cell types separately, for all donors, incubated with all test items at all concentrations used in the potency assay.
  • markers used for B and T cell staining were anti- CD20 and anti-CD7, respectively for the study shown in Figure 23. This assay was performed with resting and IL2-activated PBMCs, purified CD4+ and CD8+ T cells.
  • the averaged culture medium background signal (OD values) is subtracted from all wells before evaluating cytotoxic response.
  • OD values averaged culture medium background signal
  • the spontaneous release of effectors and target combined is obtained from wells without any test items, for each effector population tested: the PBMCs, CD4 and CD8 negatively selected populations.
  • the background LDH activity of the test system is better evaluated in wells containing the experimental mixture of effectors -target populations without any test item present.
  • Figure 3A depicts the ability of v873 to redirect IL-2 activated PBMC to kill target Raji B cells from 3 donors.
  • Figure 3B demonstrates that v873 is able to mediate higher redirected T-cell cytotoxicity than v891 in one of the donors.
  • IL-2 stimulation of PBMC was tested at 1000-3000 units per mL.
  • Example 6 Heteromultimers mediate redirected killing of target Raji B cells with resting and IL-2 activated CD4+ and CD8+ T cells
  • Figure 16A depicts the ability of v875, v1379 and v1380 to mediate antibody dependent B cell cytoxicity by redirected CD4+ and CD8+ T cell towards Raji B cells.
  • the top panel of Figure 16 illustrates the cytoxicity of resting CD4+ and CD8+ T cells towards Raji B cells. These results illustrate that v875, v1380 and v891 ellicit a concentration dependant cytotoxic response that is more prominent in the CD8+ T cells.
  • the lower panel of Figure 16 illustrates that the cytoxicity of IL-2 stimulated CD4+ and CD8+ T cells towards target Raji B cells. These results indicate that v875 and v1379 ellicit similar
  • Figure 16 B-E depict representations of the data in Figure 16A normalized to human IgG, for v875 ( Figures 16B and C), and v1379 and v1380
  • Figures 16D and E include % cytotoxicity indicated at each test antibody concentration.
  • Figure 16B shows the % cytotoxicity to target Raji B cells with v875 with IL-2 activated CD4+ and CD8+ effector T cells.
  • Figure 16C shows the % cytotoxicity to target Raji B cells with v875 with resting CD4+ and CD8+ effector T cells.
  • Figure 16D and E show direct comparisons of v1379 (WT Fc) and v1380 (L234A_L235A Fc knockout) in IL-2 activated ( Figure 16D), and resting (Figure 16E) CD4+ and CD8+ T cells. The most signicant impact of the
  • FIG. 20 shows the target Raji B cell cytotoxicty of v875 with IL-2 activated CD4+ and CD8+ T cells.
  • the percent target B cell killing elicited with IL-2 activated CD4+ cells and CD8+ T cells did not increase at v875 concentrations above the 0.06 nM.
  • target B cell killing is greater with IL-2 activated CD8+ T cells, compared to IL-2 activated CD4+ T cells.
  • Figure 20B shows the target Raji B cell cytotoxicty of v875 with resting CD4+ and CD8+ T cells. The percent target B cell killing elicited with resting CD8+ cells does not increase greatly at v875
  • Dose-dependent B cell killing is seen with v875 and v873 when CD4+ and CD8+ effector T cells are used. As expected, target B cell killing is greater with resting CD8+ T cells, compared to resting CD4+ T cells.
  • Example 7 The heterodimeric Fc contributes to target Raji B cell cytotoxicity
  • Figure 17 depicts v875 and v873 Azymetri
  • FIG. 17A illustrates that Fc blocking of IL-2 activated PBMC results in a minor (v875) or no (v873) reduction in the % cytotoxicity of target Raji B cells.
  • Figure 17B illustrates that Fc blocking of resting PBMC results in a reduction in the % cytotoxicity of target Raji B cells for v875 and v873.
  • Figure 18A illustrates that Fc blocking of IL-2 activated PBMC results in a reduction in the % cytotoxicity of target Raji B cells at all antibody
  • Figure 18B illustrates that Fc blocking of resting PBMC results in a reduction in the % cytotoxicity of target Raji B cells at all antibody concentrations tested for v875 and v873.
  • Figures 17 and 18 show that the Fc contributes to target Raji B cell cytotoxicity in the
  • Example 8 Heteromultimers mediate autologous B cell cytotoxicity
  • Figure 21 shows, relative to untreated media and human IgG controls, that v875 and v873 (300 nM) mediate autologous B cell killing in total resting PBMC (left panel) and total IL-2 activated PBMC .
  • Example 9 Heteromultimer v875 spares autologous T cell cytotoxicity compared to BiTE
  • Figure 22 shows, relative to untreated me ⁇
  • v875 has a more selective B cell killing by sparing more autologous T cells compared to v873 and v891.
  • Example 10 Design, expression and purification of heteromultimers with an albumin scaffold.
  • the sequences for the anti-CD19 and anti-CD3 scFvs were chosen from two molecules that are currently in clinical trials and are well documented and tested for stability and production.
  • the anti-CD19 and anti-CD3 scFv were directly adopted from the BiTE molecule blinatumomab.
  • the antiCD3 scFv was chosen in the VH-VL orientation, consistent with what used in BiTE.
  • the benchmark molecule was an scFv molecule based on BiTE (v891 ).
  • AlbuCORE_1 (ABH2) CD3/CD19 fusions were created by attaching the antiCD3 warhead to the natural N terminus of fragment 1 and the antiCD19 to the C terminus of fragment 2 (v1092, polypeptide sequences corresponding to SEQ ID NO:264 and 266).
  • the linkers used were identical to the ones used for the multivalent HER2 AlbuCORE experiments: GGGS at the N terminus of fragment 1 and (GGSG) 4 GG at the C terminus of fragment 2.
  • a second molecule was created where the warheads were reversed (i.e. anti-CD19 warhead at the natural N terminus of fragment 1 and the anti-CD3 at the C terminus of fragment 2, v1093.
  • v1094 was designed to accommodate two different fusions at the natural termini of the albumin polypeptide (polypeptide sequences corresponding to SEQ ID NO:268).
  • the scFv fusions were linked to the albumin molecule through a GGS linker at the N terminus and a GGSG linker at the C terminus.
  • the length of the linkers reflect the ones used in the MM-1 1 1 molecule, despite having a different sequence type.
  • V221 is the albumin-based heteromultimer used to construct v1092, but without the cargo molecules (polypeptide sequences corresponding to SEQ ID NO:269 and 270).
  • Example 1 1 Heteromultimers with an albumin scaffold bind specifically to CD3- or CD19-expressing cells
  • Example 12 Heteromultimers with heterodimeric Fc or albumin scaffolds show comparable B-cell targeting and T-cell bridging
  • Example 13 Exemplary heteromultimers have higher anti-CD3 KD and higher Bmax in binding to T and B cell as determined by FACS.

Abstract

Disclosed herein are isolated multispecific heteromultimer constructs comprising multispecific heteromultimer construct comprising: a first polypeptide construct comprising a first heavy chain polypeptide and a CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second heavy chain polypeptide which is different from said first heavy chain polypeptide, and an antigen binding polypeptide construct that binds to a target antigen on at least one B cell; wherein: said multispecific heteromultimer construct simultaneously engages said at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell; and said first and second heavy chain polypeptides form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc with stability at least comparable to a native homodimeric Fc, and with high purity. Also provided are isolated multispecific heteromultimer construct comprising: a first polypeptide construct comprising a first transporter polypeptide fused to at least one CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second transporter polypeptide which is different from said first transporter polypeptide, fused to at least one antigen binding polypeptide construct that binds to a target antigen on at least one B cell; wherein said first and second transporter polypeptides are derived from a protein by segmentation of said protein, each transporter polypeptide comprising an amino acid sequence with at least 90% identity to a segment of said protein, and wherein said transporter polypeptides self-assemble to form a quasi-native structure of said monomeric protein.

Description

BISPECIFIC ASYMMETRIC HETERODIMERS COMPRISING ANTI-CD3
CONSTRUCTS
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of U.S. Application Serial No. 61/671 ,640, filed July 13, 2012; and U.S. Application Serial No. 61/845,948, filed July 13, 2013, which are hereby incorporated by reference in their entirety.
FIELD OF THE INVENTION
[0002] The field of the invention is the rational design of multispecific scaffolds comprising a CD3 binding domain for custom development of biotherapeutics.
BACKGROUND OF THE INVENTION
[0003] In the realm of therapeutic proteins, antibodies with their multivalent target binding features are excellent scaffolds for the design of drug
candidates. Advancing these features further, designed bispecific antibodies and other fused multispecific therapeutics exhibit dual or multiple target specificities and an opportunity to create drugs with novel modes of action. The development of such multivalent and multispecific therapeutic proteins with favorable pharmacokinetics and functional activity has been a challenge.
[0004] The immune system of both humans and animals include two principal classes of lymphocytes: the thymus derived cells (T cells), and the bone marrow derived cells (B cells). T cells exhibit immunological specificity and are directly involved in cell-mediated immune responses (such as graft rejection). T cells act against or in response to a variety of foreign structures (antigens). In many instances these foreign antigens are expressed on host cells as a result of infection. However, foreign antigens can also come from the host having been altered by neoplasia or infection.
[0005] T cell activation is a complex phenomenon that depends on the participation of a variety of cell surface molecules expressed on the responding WSGF
T cell population. For example, the antigen-specific T
composed of a disulfide-linked heterodimer, containing two clonally distributed, integral membrane glycoprotein chains, alpha and beta (a and β), or gamma and delta (γ and δ), non-covalently associated with a complex of low molecular weight invariant proteins, commonly designated as CD3.
SUMMARY OF THE INVENTION
[0006] Provided herein are multispecific heteromultimers comprising a CD3 binding domain. In an embodiment is provided a bispecific asymmetric heterodimer comprising anti-CD3 constructs.
[0007] Provided herein are isolated multispecific heteromultimer constructs comprising: a first polypeptide construct comprising a first heavy chain polypeptide and a CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second heavy chain polypeptide which is different from said first heavy chain polypeptide, and an antigen binding polypeptide construct that binds to a target antigen on at least one B cell; wherein: at least one of said CD3 binding polypeptide construct and said antigen binding polypeptide construct comprises a single chain Fv region; wherein said multispecific heteromultimer construct simultaneously engages said at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell; and said first and second heavy chain polypeptides form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc with stability at least comparable to a native homodimeric Fc, and with purity such that when said multispecific heteromultimer construct is coexpressed from a stable mammalian cell in an expression product, said expression product comprises at least about 70% of said multispecific heteromultimer, and less than 10% monomers or homodimers of said first or second polypeptide constructs. In certain
embodiments, said stable mammalian cell is transfected at least a first DNA
1 WSGF sequence encoding said first polypeptide construct an
sequence encoding said second polypeptide construct in a pre-determined ratio of 1 : 1 . In certain embodiments, the first or second polypeptide construct is devoid of at least one of immunoglobulin light chain, and immunoglobulin first constant (CH1 ) region.
[0008] In certain embodiments are the isolated heteromultimer constructs described herein, wherein the heterodimer Fc region comprises a variant CH2 domain or hinge comprising amino acid modifications that prevents functionally effective binding to all the Fcgamma receptors. In some embodiments is provided the isolated multispecific heteromultimer described herein, wherein wherien said variant CH2 domain or hinge comprising amino acid modification also prevents functionally effective binding to complement proteins (C1 q complex). In an embodiment is the isolated multispecific heteromultimer described herein, wherein the heterodimer Fc region comprises a variant CH2 domain or hinge comprising amino acid modifications that enhance binding to the FcyRllb receptor.
[0009] In an embodiment is provided an isolated multispecific
heteromultimer construct comprising: a first polypeptide construct comprising a first heavy chain polypeptide and a CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second heavy chain polypeptide which is different from said first heavy chain polypeptide, and an antigen binding polypeptide construct that binds to a target antigen on at least one B cell;
wherein: at least one of said CD3 binding polypeptide construct and said antigen binding polypeptide construct optionally comprises a single chain Fv region; said first and second heavy chain polypeptides form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc, wherein: said heterodimeric Fc is formed with stability at least comparable to a
2 WSGF native homodimeric Fc, and said heterodimeric Fc is f
that when said multispecific heteromultimer construct is coexpressed from a mammalian cell in an expression product, said expression product comprises greater than 70% of said multispecific heteromultimer, and less than 10% monomers or homodimers of said first or second polypeptide constructs; and said multispecific heteromultimer construct binds said at least one B cell with a valency greater than one, and said multispecific heteromultimer simultaneously engages said at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell. In certain embodiments, said multispecific heteromultimer construct binds said at least one B cell with a valency of two.
[0010] Provided herein is an isolated multispecific heteromultimer construct comprising: a first polypeptide construct comprising a first heavy chain polypeptide and a CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second heavy chain polypeptide which is different from said first heavy chain polypeptide, and a steric modulator construct which exhibits negligible receptor binding; wherein: said multispecific heteromultimer construct simultaneously engages at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell; and said first and second heavy chain polypeptides form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc, wherein: said heterodimeric Fc is formed with stability at least comparable to a native homodimeric Fc, and said heterodimeric Fc is formed with purity such that when said multispecific heteromultimer construct is coexpressed from a stable mammalian cell in an expression product, said expression product comprises at least about 75% of said multispecific heteromultimer, and less than 10% monomers or homodimers of said first or second polypeptide constructs.
3 WSGF
[0011] Provided is an isolated multispecific heterc
comprising: a first polypeptide construct comprising a first heavy chain polypeptide and a CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second heavy chain polypeptide which is different from said first heavy chain polypeptide, and wherein said second polypeptide construct does not comprise an antigen binding polypeptide construct; wherein: said multispecific heteromultimer construct simultaneously engages at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell; and said first and second heavy chain polypeptides form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc, wherein: said heterodimeric
Fc is formed with stability at least comparable to a native homodimeric Fc, and said heterodimeric Fc is formed with purity such that when said multispecific heteromultimer construct is coexpressed from a stable mammalian cell in an expression product, said expression product comprises at least about 75% of said multispecific heteromultimer, and less than 10% monomers or
homodimers of said first or second polypeptide constructs.
[0012] In certain embodiments is the isolated multispecific heteromultimer described herein wherein the heterodimer Fc region comprises a variant CH2 domain comprising amino acid modifications to promote selective binding of a
Fcgamma receptor. In some embodiments, the variant CH2 domain selectively binds Fcgammallb receptor greater than to wild-type CH2 domain. In certain embodiments, the variant CH2 domain selectively binds atleast one of
Fcgammallla and Fcgammalla receptor greater than a wild-type CH2 domain.
[0013] In certain embodiments is an isolated multispecific heteromultimer comstruct described herein, wherein the variant CH3 domain has a melting temperature (Tm) of about 73°C or greater. In some embodiments, the heterodimer Fc region is formed with a purity greater than about 90%. In certain embodiments, the heterodimer Fc region is formed with a purity of about 95%
4 WSGF or greater and the Tm is at least about 75 C. In some
heterodimer Fc region is formed with a purity of at least about 90% and the Tm is about 75°C. In an embodiment,the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications L351Y, F405A, and Y407V, and the variant CH3 sequence of the second transporter polypeptide comprises the amino acid modifications T366L, K392M, and T394W. In another embodiment, the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications L351Y, F405A, and Y407V, and the variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications T366L, K392L, and T394W. In a further
embodiment,the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications T350V, L351Y, F405A, and Y407V, and the variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications T350V, T366L, K392M, and T394W. In some embodiments, the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications T350V, L351Y, F405A, and Y407V, and the variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications T350V, T366L, K392L, and T394W. In yet another embodiment, the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications T366L, N390R, K392R, and T394W, and the variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications L351 Y, S400E, F405A, and Y407V. In so,e embodiments,the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications T350V, T366L, N390R, K392R, and T394W, and the variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications T350V, L351Y, S400E, F405A, and Y407V.
[0014] In certain embodiments is provided an isolated multispecific heteromultimer described herein, wherein the heterodimer Fc is glycosylated.
In some embodiments is provided an isolated multispecific heteromultimer described herein, wherein the heterodimer Fc is afucosylated. In another
5 WSGF embodiment is provided an isolated multispecific hete
herein, wherein the heterodimer Fc is aglycosylated.
[0015] In some embodiments is the isolated multispecific heteromultimer described herein wherein the antigen binding polypeptide construct that binds to a target antigen on at least one B cell comprises at least one target antigen binding domain derived from an antibody, a fibronectin, an affibody, anticalin, cysteine knot protein, DARPin, avimer, Kunitz domain or variant or derivative thereof. In an embodiment is the isolated multispecific heteromultimer described herein, wherein said antibody is a heavy chain antibody devoid of light chains. In further embodiments is the isolated multispecific heteromultimer described herein, wherein said antigen binding polypeptide construct comprises at least one CD19 binding domain. In certain embodiments is the isolated multispecific heteromultimer described herein, wherein said antigen binding polypeptide construct comprises at least one CD20 binding domain.
[0016] Provided herein is an isolated multispecific heteromultimer construct comprising: a first polypeptide construct comprising a first transporter polypeptide fused to at least one CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second transporter polypeptide which is different from said first transporter polypeptide, fused to at least one antigen binding polypeptide construct that binds to a target antigen on at least one B cell;
wherein said first and second transporter polypeptides are derived from a protein by segmentation of said protein, each transporter polypeptide comprising an amino acid sequence with at least 90% identity to a segment of said protein, and wherein said transporter polypeptides self-assemble to form a quasi-native structure of said monomeric protein.
[0017] In certain embodiments, said transporter polypeptides are not derived from an antibody. In certain embodiment, each transporter polypeptide is an albumin derivative. In some embodiments, said albumin is human serum albumin. In seome embodiments, least one transporter polypeptide is an allo-
6 WSGF albumin derivative. In certain embodiments, each tran
derived from a different alloalbumin.
[0018] Provided herein are isolated multispecific heteromultimer constructs comprising: a first polypeptide construct comprising a first transporter polypeptide fused to at least one CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second transporter polypeptide which is different from said first transporter polypeptide, fused to at least one antigen binding polypeptide construct that binds to a target antigen on at least one B cell;
wherein said first and second transporter polypeptides are obtained by segmentation of albumin, and each transporter polypeptide comprising an amino acid sequence with at least 90% identity to a segment of albumin such that said transporter polypeptides self-assemble to form quasi-native albumin, and wherein said first cargo polypeptide does not have any binding domain present in said second cargo polypeptide.
[0019] In certain embodiments is provided a heteromultimer described herein, wherein said multispecific heteromultimer construct simultaneously engages said at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell. In some embodiments, said antigen binding polypeptide construct that binds to a target antigen on at least one B cell comprises at least one target antigen binding domain derived from an antibody, a fibronectin, an affibody, anticalin, cysteine knot protein, DARPin, avimer, Kunitz domain or variant or derivative thereof. In certain embodiments, the antigen binding polypeptide construct comprises at least one CD19 binding domain.
[0020] In certain embodiments is provided a multispecific heteromultimer described herein, wherein said antigen binding polypeptide construct comprises at least one CD20 binding domain.
[0021] In certain embodiments is provided a multispecific heteromultimer described herein, wherein the at least one CD3 binding polypeptide construct comprises at least one CD3 binding domain derived from a CD3 specific
7 WSGF antibody, a nanobody, fibronectin, affibody, anticalin, (
DARPin, avimer, Kunitz domain or variant or derivative thereof. In some embodiments, the at least one CD3 binding domain comprises at least one amino acid modification that reduces immunogenicity as compared to a corresponding CD3 binding domain not comprising said modification. In some embodiments is the isolated multispecific heteromultimer described herein, wherein said at least one CD3 binding domain comprises at least one amino acid modification that increases its stability as measured by Tm, as compared to a corresponding CD3 binding domain not comprising said modification. In some embodiments, the at least one CD3 binding polypeptide construct comprises at least one CD3 binding domain derived from a CD3 specific antibody is a heavy chain antibody devoid of light chains. In certain embodiments, the at least one CD3 binding polypeptide construct comprises at least one CD3 binding domain derived from a non-antibody protein scaffold domain.
[0022] In an embodiment is provided the isolated heteromutlimer construct described herein, wherein at least one of said first and second polypeptide constructs further comprises a single-chain Fv polypeptide. In certain embodiments, is provided the isolated heteromutlimer construct described herein, wherein at least one of said first and second polypeptide constructs further comprises a single-chain Fab polypeptide.
[0023] In some embodiments is the isolated heteromutlimer construct described herein, where in the CD3 expressing cell is a T-cell. In certain embodiments is the isolated heteromultimer described herein, wherein said heteromultimer binds to the T-cell with sufficient affinity and decorates the T cell at sufficient capacity that induces the T-cell to display B cell killing activity when the T cell and the B cell are bridged.
[0024] Provided is an isolated heteromutlimer construct described herein, where in the CD3 expressing cell is a human cell. In certain embodiments, the
CD3 expressing cell is a non-human, mammalian cell. In some embodiments, the mammalian cell is a primate cell. In certain embodiments, the primate is a monkey. In some embodiments, the at least one CD3 binding polypeptide
8 WSGF construct binds to CD3 constructs across multiple spe
embodiments the CD3 binding polypeptide binds to CD3 constructs across multiple species which include at least one or more of human, rat, mouse and monkey.
[0025] Provided is an isolated heteromutlimer construct described herein wherein the at least one B cell is associated with a disease. In certain embodiments, the disease is a cancer selected from a carcinoma, a sarcoma, leukaemia, lymphoma and glioma. In some embodiments, the cancer is at least one of squamous cell carcinoma, adenocarcinoma, transition cell carcinoma, osteosarcoma and soft tissue sarcoma. In an embodiment, the at least one B cell is an autoimmune reactive cell that is a lymphoid or myeloid cell.
[0026] Provided is an isolated heteromutlimer construct described herein wherein said heteromultimer further comprises at least one binding domain that binds at least one of: EpCAM, EGFR, IGFR, HER-2 neu, HER-3, HER-4, PSMA, CEA, MUC-1 (mucin), MUC2, MUC3, MUC4, MUC5, MUC7, CCR4, CCR5, CD19, CD20, CD33, CD30, ganglioside GD3, 9-0-Acetyl-GD3, GM2, Poly SA, GD2, Carboanhydrase IX (MN/CA IX), CD44v6, Sonic Hedgehog (Shh), Wue-1 , Plasma Cell Antigen, (membrane-bound), Melanoma
Chondroitin Sulfate Proteoglycan (MCSP), CCR8, TNF-alpha precursor, STEAP, mesothelin, A33 Antigen, Prostate Stem Cell Antigen (PSCA), Ly-6; desmoglein 4, E-cadherin neoepitope, Fetal Acetylcholine Receptor, CD25, CA19-9 marker, CA-125 marker and Muellerian Inhibitory Substance (MIS) Receptor type II, sTn (sialylated Tn antigen; TAG-72), FAP (fibroblast activation antigen), endosialin, LG, SAS, EPHA4 CD63, CD3 BsAb immunocytokines TNF, IFNy, IL-2, and TRAIL.
[0027] Provided is an isolated heteromutlimer construct described herein wherein said heteromultimer optionally comprises at least one linker. In some embodiments, said at least one linker is a polypeptide comprising from about 1 to about 100 amino acids.
9 WSGF
[0028] Provided is a set of expression vectors for
multispecific heteromultimer described herein, comprising at least a first DNA sequence encoding said first polypeptide construct and at least a second DNA sequence encoding said second polypeptide construct.
[0029] Provided is a method of producing an expression product containing a multispecific heteromultimer described herein, in stable
mammalian cells, the method comprising: transfecting at least one mammalian cell with: at least a first DNA sequence encoding said first polypeptide construct and at least a second DNA sequence encoding said second polypeptide construct, such that said at least one first DNA sequence, said at least one second DNA sequence are transfected in said at least one mammalian cell in a pre-determined ratio to generate stable mammalian cells; culturing said stable mammalian cells to produce said expression product comprising said multispecific heteromultimer. In some embodiments is the method of producing an expressin product containing a multispecific heteromultimer construct described herien, wherein said predetermined ratio of the at least one first DNA sequence: at least one second DNA sequence is about 1 :1. In some
embodiments, said mammalian cell is selected from the group consisting of a VERO, HeLa, HEK, NS0, Chinese Hamster Ovary (CHO), W138, BHK, COS-7, Caco-2 and MDCK cell, and subclasses and variants thereof.
[0030] Provided is a pharmaceutical composition comprising a
multispecific heteromultimer described herein, and a suitable excipient. Also provided is a process for the production of said pharmaceutical composition, said process comprising: culturing a host cell under conditions allowing the expression of a heteromultimer as defined herein; recovering the produced heteromultimer from the culture; and producing the pharmaceutical
composition.
[0031] Provided is a method for the prevention, treatment or amelioration of at least one of: a proliferative disease, a minimal residual cancer, a tumorous disease, an inflammatory disease, an immunological disorder, an autoimmune disease, an infectious disease, viral disease, allergic reactions, parasitic
10 WSGF reactions, graft-versus-host diseases or host-versus-g
malignancies, said method comprising administering to a subject in need of such a prevention, treatment or amelioration a pharmaceutical composition described herein. Provided is a method of treating cancer in a mammal in need thereof, comprising administering to the mammal a composition comprising an effective amount of the pharmaceutical composition described herein, optionally in combination with other pharmaceutically active molecules. In certain embodiments said cancer is a solid tumor. In some other embodiments, said solid tumor is one or more of sarcoma, carcinoma, and lymphoma. In certain other embodiments, the cancer is a hematological cancer. In a further embodiment, the cancer is one or more of B-cell lymphoma, non-Hodgkin's lymphoma, and leukemia.
[0032] Provided is a method of treating cancer cells comprising providing to said cell a composition comprising a heteromultimer described herein. In certain embodiments, said heteromultimer is provided in conjugation with another therapeutic agent.
[0033] Provided is a method of treating a cancer non-responsive to at least one of a CD19 lytic antibody, a CD20 lytic antibody and blinatumomab, in a mammal in need thereof, comprising administering to the mammal a
composition comprising an effective amount of the pharmaceutical composition described herein.
[0034] Provided is a method of treating a cancer cell regressive after treatment with blinatumomab, comprising providing to said cancer cell a composition comprising an effective amount of the pharmaceutical composition described herein.
[0035] Provided is a method of treating an individual suffering from a disease characterized by expression of B cells, said method comprising providing to said individual an effective amount of a composition comprising an effective amount of the pharmaceutical composition described herein. In certain embodiments, said disease is not responsive to treatment with at least one of an anti-CD19 antibody and an anti-CD20 antibody.
11 WSGF
[0036] Provided is a method of treating an autoirr
mammal in need thereof, comprising administering to said mammal a composition comprising an effective amount of the pharmaceutical composition provided herein. In certain embodiments, said autoimmune condition is one or more of multiple sclerosis, rheumatoid arthritis, lupus erytematosus, psoriatic arthritis, psoriasis, vasculitis, uveitis, Crohn's disease, and type 1 diabetes.
[0037] Provided is a method of treating an inflammatory condition in a mammal in need thereof, comprising administering to said mammal a composition comprising an effective amount of the pharmaceutical composition comprising an heteromultimer provided herein.
[0038] Provided is a kit comprising a heteromultimer as defined herein, and instructions for use thereof.
[0039] Provided herein is a heteromultimer construct comprising: a first monomer comprising a first transporter polypeptide fused to a first cargo polypeptide that comprises at least one HER2 binding domain; a second monomer comprising a second transporter polypeptide which is different from said first transporter polypeptide, fused to a second cargo polypeptide that comprises at least one HER3 binding domain; wherein said first cargo polypeptide does not have any binding domain present in said second cargo polypeptide; wherein said first and second transporter polypeptide form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimer with stability comparable to a native homodimeric Fc.
[0040] Provided herein is a heteromultimer construct comprising: a first monomer comprising a first transporter polypeptide fused to a first cargo polypeptide that comprises at least one HER2 binding domain; a second monomer comprising a second transporter polypeptide which is different from said first transporter polypeptide, fused to a second cargo polypeptide polypeptide that comprises at least one HER3 binding domain; wherein said first and second transporter polypeptides are obtained by segmentation of
12 WSGF albumin such that said transporter polypeptides self-a:
native albumin.
[0041] Provided herein is a pharmaceutical composition comprising an insolated multispecific heteromultimer as defined herein, and a suitable excipient. Also provided is a process for the production of such a
pharmaceutical composition, said process comprising: culturing a host cell under conditions allowing the expression of a heteromultimer as defined herein; recovering the produced heteromultimer from the culture; and producing the pharmaceutical composition.
[0042]
[0043] Provided herein are host cells comprising nucleic acid encoding a heteromultimer described herein. In certain embodiments, the nucleic acid encoding the first monomeric protein and the nucleic acid encoding the second monomeric protein are present in a single vector. In certain embodiments, the nucleic acid encoding the first monomeric protein and the nucleic acid encoding the second monomeric protein are present in separate vectors.
[0044] Also provided is a kit comprising a heteromultimer as defined herein, and instructions for use thereof.
[0045]
[0046] BRIEF DESCRIPTION OF THE FIGURES
[0047]
[0048] Fig. 1 A-B: Fig. 1 A depicts exemplary schematic representation of heteromultimer constructs provided herein. For instance the Immunoglobulin based Anti-CD3 x CD19 constructs demonstrate different aspects of the heteromultimers for instance the cartoon shows the first and second
polypeptide constructs wherein the first polypeptide construct comprises a CH3 binding construct (black) and the second polypeptide construct comprises an antigen binding construct (blue). In some embodiments, the antigen binding construct is absent or replaced by a steric modulating construct. Also shown is the Fc heteromultimer formed by the variant CH3 regions of the first and
13 WSGF second polypeptide constructs. Fig. 1 B shows the abil
construct described herein (v873) and a construct that does not comprise a heteromultimer Fc (blinatumomab CD19-CD3 BiTE (v891 , MT-103)) to bridge Jurkat CD3 T cells (Top left quadrant) with Raji CD19 B cells (bottom right quadrant) by FACS
[0049] Fig. 2 demonstrates that a heteromultimer described herein (v873) is able to selectively bind and bridge to CD3-expressing Jurkat T cells (lower right panel) and to CD19-expressing Raji B cells (upper right panel). Figure 2 also demonstrates that the one-armed anti-CD3 antibody specifically binds to Jurkat T cells (lower middle panels)and does not cross-react to CD19 expressing B cells, (upper middle panels)and that the one-armed anti-CD19 antibody specifically binds to Raji B cells (upper left panel) and does not cross- react to Jurkat T cells (lower left panel).
[0050] Fig. 3A-3B: Fig. 3A depict the ability of a heteromultimer described herein (v873) to redirect IL-2 activated PBMC to kill target Raji B cells from 3 donors. Fig. 3B demonstrates that a heteromultimer described herein is able to mediate higher redirected T-cell cytotoxicity than a construct lacking the heterodimeric Fc in one of the donors.
[0051] Fig. 4 shows that heteromultimers described herein are able to bind to CD3-expressing Jurkat T-cells and to CD19-expressing Raji B-cells.
[0052] Fig. 5A-5B: Fig. 5A indicates that at the concentration tested, v1093, which is a heteromultimer described herein, was able to bridge 31 % of total cells, and v873 another heteromultimer construct described herein was able to bridge 25% of total cells. Fig. 5B demonstrates that v1093 is able to bridge Jurkat T cells and Raji B cells to a greater extent than v221 and similar to v891 and v873.
[0053] Fig. 6 shows antibody therapeutics that can be provided along with a heteromultimer described herein for treatment of certain indications.
14 WSGF
[0054] Fig. 7 shows an SDS-PAGE demonstratin
heteromultimer constructs described herein are expressed transiently in
CH03E7 cells with a cell viability of > 80 %.
[0055] Fig. 8 shows that a heteromultimer described herein (v873) induces are higher % cytotoxicity to target B cells when compared to negative control human lgG1 (G1 ) when comparing across individual donors.
[0056] Fig. 9A-9B: Fig. 9A shows that human IgG (hlgG) does not bind to Jurkat T-cells and has low level binding to Raji B-cells. Figure 9A also shows that anti-CD19 one arm constructs bind selectively to the Raji B-cells and does not cross-react to Jurkat T cells. Fig. 9B demonstrates FACS assay shows that v873-a heteromultimer described herein, binds selectively to Jurkat T-cells and to Raji B-cells.
[0057] Fig. 10 shows that v873-a heteromultimer described herein does not bind to the K562 cell line, which does not express CD19 or CD3.
[0058] Fig. 11 shows that v873-a heteromultimer described herein does not bind to mouse lymphoid cells which does not express CD19 or CD3.
[0059] Fig 12.A-B: Fig. 12 A and Fig. 12B show FACS binding curves of heteromultimer constructs described herein (v873, v875) and the construct lacking a heterodimeric Fc (v891 ), to CD3 expressing HPB-ALL and CD3 expressing Jurkat T cells, and to CD19 expressing Raji B cells.
[0060] Fig. 13A-B: Fig. 13A illustrates the FACS binding curves for heteromultimer constructs v875, v1379, v1380, v1381 , and control v891 binding to CD19 expressing Raji cells tested in 0.1 to 300 nM range. Fig. 13B illustrates the FACS binding curve for heteromultimer constructs v875, v1379, v1380, binding to HBP-ALL T cells tested in 0.1 to 300 nM range.
[0061] Fig. 14 indicates that heteomultimer constructs described herein (v875 and v891 ) facilitate comparable bridging between Raji B-cells and Jurkat T-cells. Use of the control human IgG resulted in 2.5% bridging between Raji and Jurkat cells, while v875 facilitated bridging of 22.9% of total cells, and v891 facilitated bridging of 14.5% of total cells.
15 WSGF
[0062] Fig. 15A-15B: Fig. 15A shows the amounl
ratio of T-cells to B-cells, with heteromultimer concentrations ranging from 0.3 nM to 3 nM. Fig. 15B shows the amount of bridging using a 15:1 ratio of T-cells to B-cells, with heteromultimer concentrations ranging from 0.3 nM to 3 nM. Both E:T ratios (1 :1 and 15:1 ) tested with v875 resulted in similar total T cell-B cell bridging when expressed as fold over background.
[0063] Fig 16A-16E: Fig. 16A depicts the ability of v875, v1379 and v1380 to mediate antibody dependent B cell cytoxicity by redirected CD4+ and CD8+ T cell towards Raji B cells. Fig. 16 B-16E depict representations of the data in Fig. 16A normalized to human IgG, for v875 (Fig. 16B and Fig. 16C), and v1379 and v1380 (Fig. 16D and Fig. 16E), and include % cytotoxicity indicated at each test antibody concentration.
[0064] Fig. 17A-17B: Fig. 17A illustrates that Fc blocking of IL-2 activated PBMC results in a minor (v875) or no (v873) reduction in the % cytotoxicity of target Raji B cells. Figure 17B illustrates that Fc blocking of resting PBMC results in a reduction in the % cytotoxicity of target Raji B cells for v875 and v873.
[0065] Fig. 18A-18B: Fig. 18A illustrates that Fc blocking of IL-2 activated PBMC results in a reduction in the % cytotoxicity of target Raji B cells at all antibody concentrations tested for v875 and v873. Fig. 18B illustrates that Fc blocking of resting PBMC results in a reduction in the % cytotoxicity of target Raji B cells at all antibody concentrations tested for v875 and v873.
[0066] Fig. 19A-19B: Fig. 19A shows that v875 and v873 elicit >30% cytotoxicity to target Raji B cells with IL-2 activiated CD8+ T cells as effectors, and maximal target cell killing is seen at the 3 nM concentration. Fig. 19B shows that v875 and v873 elicit dose dependent (>20%) cytotoxicity to target Raji B cells with resting CD8+ T cells as effectors.
[0067] Fig. 20A-20B: Fig. 20A shows the target Raji B cell cytotoxicty of v875 with IL-2 activated CD4+ and CD8+ T cells. Fig. 20B shows the target Raji B cell cytotoxicty of v875 with resting CD4+ and CD8+ T cells.
16 WSGF
[0068] Fig. 21 shows, relative to untreated media
that v875 and v873 (300 nM) mediate autologous B cell killing in total resting
PBMC and total IL-2 activated PBMC.
[0069] Fig. 22 shows, relative to untreated media and human IgG controls, v875 has a more selective B cell killing by sparing more autologous T cells compared to v873 and v891.
[0070] Fig. 23A-23D: Fig. 23A shows the effects of v875 on the viability of CD20+, CD4+, CD8+ subsets in IL-2 activated cell cultures. Fig. 23B shows the effects of v875 on the viability of CD20+, CD4+, CD8+ subsets resting cell cultures. Fig. 23C shows the effects of v1379 and v1380 on the viability of CD20+, CD4+, CD8+ subsets in IL-2 activated cell cultures. Fig. 23D shows shows the effects of v1379 and v1380 on the viability of CD20+, CD4+, CD8+ subsets resting cell cultures.
[0071] Fig. 24A-24B: Results of antibody mediated LDH release in resting effector and Raji B cells shown in Fig. 24A. Results of antibody mediated LDH release in activiated effector are shown in Figure 24B.
[0072] Fig. 25A-25D: Fig 25A illustrates the mediation of ADCC by rituximab and by a heteromultimer described herein with a WT Fc (v875) (ca. 40% max cell lysis). Fig 25B shows that v1379 which is a heteromultimer described herein with a WT Fc can mediate ADCC while v1380, with a
L234A__L235A knock Fc mutation, is impaired in ADCC to target Daudi B cells. Fig. 25C-25D show the results of the CDC assay with v1380 and v1379 (Fig. 25C) and v875 (Fig. 25D) of target Daudi B cells with comparisons to positive control Rituximab.
[0073] Fig. 26 shows that at 0.3 nM, v875 and v1380 do not induce PBMC proliferation compared to human IgG. The lower panel of Fig. 26 shows the results of the 100 nM antibody concentrations, and shows that v875, v1380 and v891 induce higher cell proliferation relative to human IgG. Fig. 26 (lower panel) also shows that at 100 nM, v875 has a similar proliferative index compared to anti-CD3 OKT3 in all four PBMC populations.
17 WSGF
[0074] Fig. 27A-27E: show that v1380 (L234AJJ
induces less cytokine release of TNFa, INFy, IL-2, IL-4, and IL-10 when compared to v875 (WT Fc) and OKT3. The results from the cytokine release assay as shown in Fig. 27A-27E include summary plots of PBMC supernatant TNFa (Fig. 27A) INFy (Fig. 27B), IL-2 (Fig. 27C), IL-4 (Fig. 27D), and IL-10 (Fig. 27E) levels following incubation with test items at 0.3 nM concentrations for 4 days (graph y-axis represents log cytokine levels in pg per mL from 4 donors).
[0075] Fig. 28A-28B: show the results from the average stimulation index induced by v875 at 0.3nM (Fig. 28A) and 100 nM (Fig. 28B) concentrations on purified CD8+ T cells in the absence or presence of purified CD19+ B cells at 4 days incubation time-point.
[0076] Fig. 29A-29B: show the results from the average stimulation index induced by v1380 at 0.3nM (Fig. 29A) and 100nM (Fig. 29B) concentrations on purified CD8+ T cells in the absence or presence of purified CD19+ B cells at 4 days incubation time-point.
[0077] Fig. 30A-30C: show the results from the T:B cell bridging
microscopy comparing v875 and human IgG (3 nM) at 200X and 400X magnification. Fig. 30A shows a direct comparison of human IgG and v875 at 200X magnification and illustrates a higher amount of bridging visible between Raji B cell and Jurkat T cells compared to human IgG. Figure 30B and Figure 30C show two fields of view for v875 (Figure 30B) and human IgG (Figure 30C) at 400X magnification.
[0078] Fig. 31 A-31 B: Fig. 31 A shows the SDS-PAGE analysis and relative purity of v875, v1380, v1379 and v891 following protein A and SEC purification, and following 47 day storage at 4°C. Fig. 31 B shows the SDS-PAGE analysis and relative purity of additional exemplary hetermultimers including v875, v1653, v1654, v1655, v1656, v1660, v1800, and v1802 following protein A and SEC purification.
[0079] Fig. 32 shows the LC-MS results of the Max Ent. molecular weight profiles for v875.
18 WSGF
[0080] Fig. 33A-33C: show DSC results for heter
described herein, showing that v875 has an estimated CH3 Tm > 76°C (Figure 33A), v1380 has an estimated CH3 Tm > 82.3°C, and v1379 has an estimated CH3 Tm > 82.5°C (Figure 33C).
[0081] Fig. 34A-C: show ability of heteromultimers described herein to bridge Raji B and Jurkat T cells (B:T), as well RajkRaji B cell bridging (B:B) and JurkatJurkat T cell bridging (T:T) assessed by FACS. Fig. 34A shows the amount of T:B, B:B and T:T bridging of v875, v1379, v1380, v891 , v1381 , commercial OKT3 and human IgG over three experimental replicates. Fig. 34B shows the amount of T:B, B:B and T:T bridging of variants with engineered anti-CD3 warheads for stability enhancement (v1653, v1654, v1655, v1656, v1660, v1800, v1802) and v875, and human IgG. Fig. 34C shows the amount of T:B, B:B and T:T bridging of Fc knock-out variants that have either engineered anti-CD3 warheads for stability enhancement (v1666), or have human/cynomolgous monkey cross-reactive anti-CD3 and anti-CD19 scFvs (v4541 , v4543, v4545, v4548) commercial OKT3 anti-CD3 control, v2176 anti- CD19 control and human IgG negative control, and all variants mediate low T:T bridging.
[0082] Fig. 35 illustrates binding of heteromultimers described herein to human CD3 (top panel) and binding to the cynomologous CD3 receptor (bottom panel) as determined by ELISA.
[0083] Fig. 36A-36B: illustrates affinity on linear and log scales
respectively of HER2/HER3 het-Fc construct demonstrating both bi-specificity and avidity.
[0084] Fig. 37 depicts the binding of variant 1090 compared to the control 1087 in MALME-3M cells and indicates that v1090 has similar binding as v1087 to target MALME-3M cells.
19 WSGF
DETAILED DESCRIPTION OF THE IN1
[0085] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which the claimed subject matter belongs. In the event that there are a plurality of definitions for terms herein, those in this section prevail. Where reference is made to a URL or other such identifier or address, it is understood that such identifiers can change and particular information on the internet can come and go, but equivalent information can be found by searching the internet. Reference thereto evidences the availability and public dissemination of such information.
[0086] It is to be understood that the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of any subject matter claimed. In this application, the use of the singular includes the plural unless specifically stated otherwise.
[0087] Terms understood by those in the art of antibody technology are each given the meaning acquired in the art, unless expressly defined differently herein. Antibodies are known to have variable regions, a hinge region, and constant domains. Immunoglobulin structure and function are reviewed, for example, in Harlow et al, Eds., Antibodies: A Laboratory Manual, Chapter 14 (Cold Spring Harbor Laboratory, Cold Spring Harbor, 1988).
[0088] In the present description, any concentration range, percentage range, ratio range, or integer range is to be understood to include the value of any integer within the recited range and, when appropriate, fractions thereof (such as one tenth and one hundredth of an integer), unless otherwise indicated. As used herein, "about" means ± 10% of the indicated range, value, sequence, or structure, unless otherwise indicated. It should be understood that the terms "a" and "an" as used herein refer to "one or more" of the enumerated components unless otherwise indicated or dictated by its context. The use of the alternative (e.g., "or") should be understood to mean either one, both, or any combination thereof of the alternatives. As used herein, the terms "include"
20 WSGF and "comprise" are used synonymously. In addition, it
that the individual single chain polypeptides or immunoglobulin constructs derived from various combinations of the structures and substituents described herein are disclosed by the present application to the same extent as if each single chain polypeptide or heterodimer were set forth individually. Thus, selection of particular components to form individual single chain polypeptides or heterodimers is within the scope of the present disclosure
[0089] The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. All documents, or portions of documents, cited in the application including, but not limited to, patents, patent applications, articles, books, manuals, and treatises are hereby expressly incorporated by reference in their entirety for any purpose.
[0090] It is to be understood that the methods and compositions described herein are not limited to the particular methodology, protocols, cell lines, constructs, and reagents described herein and as such may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the methods and compositions described herein, which will be limited only by the appended claims.
[0091] All publications and patents mentioned herein are incorporated herein by reference in their entirety for the purpose of describing and
disclosing, for example, the constructs and methodologies that are described in the publications, which might be used in connection with the methods, compositions and compounds described herein. The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the inventors described herein are not entitled to antedate such disclosure by virtue of prior invention or for any other reason.
[0092] In the present application, amino acid names and atom names (e.g.
N, O, C, etc.) are used as defined by the Protein DataBank (PDB)
21 WSGF
(www.pdb.org), which is based on the lUPAC nomenc
Nomenclature and Symbolism for Amino Acids and Peptides (residue names, atom names etc.), Eur. J. Biochem., 138, 9-37 (1984) together with their corrections in Eur. J. Biochem., 152, 1 (1985).
[0093] The terms "polypeptide," "peptide" and "protein" are used interchangeably herein to refer to a polymer of amino acid residues. That is, a description directed to a polypeptide applies equally to a description of a peptide and a description of a protein, and vice versa. The terms apply to naturally occurring amino acid polymers as well as amino acid polymers in which one or more amino acid residues is a non-naturally encoded amino acid. As used herein, the terms encompass amino acid chains of any length, including full length proteins, wherein the amino acid residues are linked by covalent peptide bonds.
[0094] The term "nucleotide sequence" is intended to indicate a
consecutive stretch of two or more nucleotide molecules. The nucleotide sequence may be of genomic, cDNA, RNA, semisynthetic or synthetic origin, or any combination thereof.
[0095] The term "polymerase chain reaction" or "PCR" generally refers to a method for amplification of a desired nucleotide sequence in vitro, as described, for example, in U.S. Pat. No. 4,683,195. In general, the PCR method involves repeated cycles of primer extension synthesis, using oligonucleotide primers capable of hybridising preferentially to a template nucleic acid.
[0096] "Cell", "host cell", "cell line" and "cell culture" are used
interchangeably herein and all such terms should be understood to include progeny resulting from growth or culturing of a cell. "Transformation" and "transfection" are used interchangeably to refer to the process of introducing DNA into a cell.
[0097] The term "amino acid" refers to naturally occurring and non- naturally occurring amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino
22 WSGF acids. Naturally encoded amino acids are the 20 comr
arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, praline, serine, threonine, tryptophan, tyrosine, and valine) and pyrrolysine and selenocysteine. Amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an a carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, such as, homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (such as, norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid. Reference to an amino acid includes, for example, naturally occurring proteogenic L-amino acids; D-amino acids, chemically modified amino acids such as amino acid variants and derivatives; naturally occurring non-proteogenic amino acids such as β-alanine, ornithine, etc.; and chemically synthesized compounds having properties known in the art to be characteristic of amino acids. Examples of non-naturally occurring amino acids include, but are not limited to, a-methyl amino acids (e.g. a-methyl alanine), D- amino acids, histidine-like amino acids (e.g., 2-amino-histidine, β-hydroxy- histidine, homohistidine), amino acids having an extra methylene in the side chain ("homo" amino acids), and amino acids in which a carboxylic acid functional group in the side chain is replaced with a sulfonic acid group (e.g., cysteic acid). The incorporation of non-natural amino acids, including synthetic non-native amino acids, substituted amino acids, or one or more D-amino acids into the proteins of the present invention may be advantageous in a number of different ways. D-amino acid-containing peptides, etc., exhibit increased stability in vitro or in vivo compared to L-amino acid-containing counterparts. Thus, the construction of peptides, etc., incorporating D-amino acids can be particularly useful when greater intracellular stability is desired or required. More specifically, D-peptides, etc., are resistant to endogenous peptidases and proteases, thereby providing improved bioavailability of the molecule, and
23 WSGF prolonged lifetimes in vivo when such properties are d
peptides, etc., cannot be processed efficiently for major histocompatibility complex class ll-restricted presentation to T helper cells, and are therefore, less likely to induce humoral immune responses in the whole organism.
[0098] Amino acids may be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the lUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes.
[0099] "Conservatively modified variants" applies to both amino acid and nucleic acid sequences. With respect to particular nucleic acid sequences, "conservatively modified variants" refers to those nucleic acids which encode identical or essentially identical amino acid sequences, or where the nucleic acid does not encode an amino acid sequence, to essentially identical sequences. Because of the degeneracy of the genetic code, a large number of functionally identical nucleic acids encode any given protein. For instance, the codons GCA, GCC, GCG and GCU all encode the amino acid alanine. Thus, at every position where an alanine is specified by a codon, the codon can be altered to any of the corresponding codons described without altering the encoded polypeptide. Such nucleic acid variations are "silent variations," which are one species of conservatively modified variations. Every nucleic acid sequence herein which encodes a polypeptide also describes every possible silent variation of the nucleic acid. One of ordinary skill in the art will recognize that each codon in a nucleic acid (except AUG, which is ordinarily the only codon for methionine, and TGG, which is ordinarily the only codon for tryptophan) can be modified to yield a functionally identical molecule.
Accordingly, each silent variation of a nucleic acid which encodes a polypeptide is implicit in each described sequence.
[00100] As to amino acid sequences, one of ordinary skill in the art will recognize that individual substitutions, deletions or additions to a nucleic acid, peptide, polypeptide, or protein sequence which alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a
24 WSGF
"conservatively modified variant" where the alteration
an amino acid, addition of an amino acid, or substitution of an amino acid with a chemically similar amino acid. Conservative substitution tables providing functionally similar amino acids are known to those of ordinary skill in the art. Such conservatively modified variants are in addition to and do not exclude polymorphic variants, interspecies homologs, and alleles of the invention.
[00101] Conservative substitution tables providing functionally similar amino acids are known to those of ordinary skill in the art. The following eight groups each contain amino acids that are conservative substitutions for one another:
[00102] 1 ) Alanine (A), Glycine (G);
[00103] 2) Aspartic acid (D), Glutamic acid (E);
[00104] 3) Asparagine (N), Glutamine (Q);
[00105] 4) Arginine (R), Lysine (K);
[00106] 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V);
[00107] 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W);
[00108] 7) Serine (S), Threonine (T); and [0139] 8) Cysteine (C),
Methionine (M)
[00109] (see, e.g., Creighton, Proteins: Structures and Molecular Properties (W H Freeman & Co.; 2nd edition (December 1993)
[00110] The terms "identical" or percent "identity," in the context of two or more nucleic acids or polypeptide sequences, refer to two or more sequences or subsequences that are the same. Sequences are "substantially identical" if they have a percentage of amino acid residues or nucleotides that are the same (i.e., about 50% identity, about 55% identity, 60% identity, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, or about 95% identity over a specified region), when compared and aligned for maximum correspondence over a comparison window, or designated region as measured using one of the following sequence comparison algorithms (or other algorithms available to persons of ordinary skill in the art) or by manual alignment and visual inspection. This definition also refers to the complement of a test sequence. The identity can exist over a region that is at least about 50 amino
25 WSGF acids or nucleotides in length, or over a region that is
nucleotides in length, or, where not specified, across the entire sequence of a polynucleotide or polypeptide. A polynucleotide encoding a polypeptide of the present invention, including homologs from species other than human, may be obtained by a process comprising the steps of screening a library under stringent hybridization conditions with a labeled probe having a polynucleotide sequence of the invention or a fragment thereof, and isolating full-length cDNA and genomic clones containing said polynucleotide sequence. Such
hybridization techniques are well known to the skilled artisan.
[00111] A derivative, or a variant of a polypeptide is said to share
"homology" or be "homologous" with the peptide if the amino acid sequences of the derivative or variant has at least 50% identity with a 100 amino acid sequence from the original peptide. In certain embodiments, the derivative or variant is at least 75% the same as that of either the peptide or a fragment of the peptide having the same number of amino acid residues as the derivative. . In certain embodiments, the derivative or variant is at least 85% the same as that of either the peptide or a fragment of the peptide having the same number of amino acid residues as the derivative. In certain embodiments, the amino acid sequence of the derivative is at least 90% the same as the peptide or a fragment of the peptide having the same number of amino acid residues as the derivative. In some embodiments, the amino acid sequence of the derivative is at least 95% the same as the peptide or a fragment of the peptide having the same number of amino acid residues as the derivative. In certain embodiments, the derivative or variant is at least 99% the same as that of either the peptide or a fragment of the peptide having the same number of amino acid residues as the derivative.
[00112] The term "bispecific" is intended to include any agent, e.g., heteromultimer, monomer, protein, peptide, or protein or peptide complex, which has two different binding specificities. For example, in some
embodiments, the molecule may bind to, or interact with, (a) a cell surface target molecule and (b) an Fc receptor on the surface of an effector cell. In
26 WSGF certain embodiments of a heteromultimer described hi
monomer is bispecific formed by attaching to the same transporter polypeptide, two cargo molecules with different binding specificities. In certain embodiments of a heteromultimer described herein, the heteromultimer is itself bispecific formed by attaching to the transporter polypeptides, at least two cargo molecules with different specificities.
[00113] The term "multispecific" or "heterospecific " is intended to include any agent, e.g., a protein, peptide, or protein or peptide complex, which has more than two different binding specificities. For example, the molecule may bind to, or interact with, (a) a cell surface target molecule such as but not limited to cell surface antigens, (b) an Fc receptor on the surface of an effector cell, and optionally (c) at least one other component. Accordingly, embodiments of the heteromultimers described herein, are inclusive of, but not limited to, bispecific, trispecific, tetraspecific, and other multispecific molecules. In certain embodiments, these molecules are directed to cell surface antigens, such as CD30, and to other targets, such as Fc receptors on effector cells.
[00114] As used herein, "isolated" heteromultimer means a heteromultimer that has been identified and separated and/or recovered from a component of its natural cell culture environment. Contaminant components of its natural environment are materials that would interfere with diagnostic or therapeutic uses for the heteromultimer, and may include enzymes, hormones, and other proteinaceous or non-proteinaceous solutes.
[00115] The phrase "selectively (or specifically) hybridizes to" refers to the binding, duplexing, or hybridizing of a molecule only to a particular nucleotide sequence under stringent hybridization conditions when that sequence is present in a complex mixture (including but not limited to, total cellular or library DNA or RNA).
[00116] The phrase "stringent hybridization conditions" refers to
hybridization of sequences of DNA, RNA, or other nucleic acids, or
combinations thereof under conditions of low ionic strength and high
temperature as is known in the art. Typically, under stringent conditions a probe
27 WSGF will hybridize to its target subsequence in a complex n
(including but not limited to, total cellular or library DNA or RNA) but does not hybridize to other sequences in the complex mixture. Stringent conditions are sequence-dependent and will be different in different circumstances. Longer sequences hybridize specifically at higher temperatures. An extensive guide to the hybridization of nucleic acids is found in Tijssen, Laboratory Techniques in Biochemistry and Molecular Biology-Hybridization with Nucleic Probes, "Overview of principles of hybridization and the strategy of nucleic acid assays" (1993).
[00117] As used herein, an "antibody" or "immunoglobulin" refers to a polypeptide substantially encoded by an immunoglobulin gene or
immunoglobulin genes, or fragments thereof, which specifically bind and recognize an analyte (antigen). The recognized immunoglobulin genes include the kappa, lambda, alpha, gamma, delta, epsilon and mu constant region genes, as well as the myriad immunoglobulin variable region genes. Light chains are classified as either kappa or lambda. Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, which in turn define the immunoglobulin classes, IgG, IgM, IgA, IgD, and IgE, respectively.
[00118] An exemplary immunoglobulin (antibody) structural unit is composed of two pairs of polypeptide chains, each pair having one "light"
(about 25 kD) and one "heavy" chain (about 50-70 kD). The N-terminal domain of each chain defines a variable region of about 100 to 1 10 or more amino acids primarily responsible for antigen recognition. The terms variable light chain (VL) and variable heavy chain (VH) refer to these light and heavy chain domains respectively. The lgG1 heavy chain comprises of the VH, CH1 , CH2 and CH3 domains respectively from the N to C-terminus. The light chain comprises of the VL and CL domains from N to C terminus. The lgG1 heavy chain comprises a hinge between the CH1 and CH2 domains. In certain embodiments, the immunoglobulin constructs comprise at least one
immunoglobulin domain from IgG, IgM, IgA, IgD, or IgE connected to a therapeutic polypeptide. In some embodiments, the immunoglobulin domain
28 WSGF comprised in an immunoglobulin construct provided hi
immunoglobulin based construct such as a diabody, or a nanobody. In certain embodiments, the immunoglobulin constructs described herein comprise at least one immunoglobulin domain from a heavy chain antibody such as a camelid antibody. In certain embodiments, the immunoglobulin constructs provided herein comprise at least one immunoglobulin domain from a mammalian antibody such as a bovine antibody, a human antibody, a camelid antibody, a mouse antibody or any chimeric antibody.
[00119] As used herein, the term "antigenic determinant" is synonymous with "antigen" and "epitope," and refers to a site (e.g. a contiguous stretch of amino acids or a conformational configuration made up of different regions of non-contiguous amino acids) on a polypeptide macromolecule to which an antigen binding moiety binds, forming an antigen binding moiety-antigen complex. Useful antigenic determinants can be found, for example, on the surfaces of tumor cells, on the surfaces of virus-infected cells, on the surfaces of other diseased cells, on the surface of immune cells, free in blood serum, and/or in the extracellular matrix (ECM). The proteins referred to as antigens herein (e.g. MCSP, FAP, CEA, EGFR, CD33, CD3) can be any native form the proteins from any vertebrate source, including mammals such as primates (e.g. humans) and rodents (e.g. mice and rats), unless otherwise indicated. In a particular embodiment the antigen is a human protein. Where reference is made to a specific protein herein, the term encompasses the "full-length", unprocessed protein as well as any form of the protein that results from processing in the cell. The term also encompasses naturally occurring variants of the protein, e.g. splice variants or allelic variants. Exemplary human proteins useful as antigens include, but are not limited to: Melanoma-associated
Chondroitin Sulfate Proteoglycan (MCSP), also known as Chondroitin Sulfate
Proteoglycan 4 (UniProt no. Q6UVK1 (version 70), NCBI RefSeq no. NP
001888.2); Fibroblast Activation Protein (FAP), also known as Seprase (Uni
Prot nos. Q12884, Q86Z29, Q99998, NCBI Accession no. NP 004451 );
Carcinoembroynic antigen (CEA), also known as Carcinoembryonic antigen-
29 WSGF related cell adhesion molecule 5 (UniProt no. P06731
RefSeq no. NP 004354.2); CD33, also known as gp67 or Siglec-3 (UniProt no. P20138, NCBI Accession nos. NP 001076087, NP 001 171079); Epidermal Growth Factor Receptor (EGFR), also known as ErbB-1 or Herl (UniProt no. P0053, NCBI Accession nos. NP 958439, NP 958440), and CD3, particularly the epsilon subunit of CD3 (see UniProt no. P07766 (version 130), NCBI RefSeq no. NP 000724.1 , SEQ ID NO: 265 for the human sequence; or UniProt no. Q95LI5 (version 49), NCBI GenBank no. BAB71849.1 , SEQ ID NO: 266 for the cynomolgus [Macaca fascicularis] sequence). In certain embodiments the T cell activating bispecific antigen binding molecule of the invention binds to an epitope of an activating T cell antigen or a target cell antigen that is conserved among the activating T cell antigen or target antigen from different species.
[00120] By "specific binding" or "selective binding" is meant that the binding is selective for the antigen and can be discriminated from unwanted or nonspecific interactions. The ability of an antigen binding moiety to bind to a specific antigenic determinant can be measured either through an enzyme- linked immunosorbent assay (ELISA) or other techniques familiar to one of skill in the art, e.g. surface plasmon resonance (SPR) technique (analyzed on a BIAcore instrument) (Liljeblad et al, Glyco J 17, 323-329 (2000)), and traditional binding assays (Heeley, Endocr Res 28, 217-229 (2002)). In one embodiment, the extent of binding of an antigen binding moiety to an unrelated protein is less than about 10% of the binding of the antigen binding moiety to the antigen as measured, e.g., by SPR. In certain embodiments, an antigen binding moiety that binds to the antigen, or an antigen binding molecule comprising that antigen binding moiety, has a dissociation constant (KD) of < 1 μΜ, < 100 nM, < 10 nM, < 1 nM, < 0.1 nM, < 0.01 nM, or < 0.001 nM (e.g. 10~8 M or less, e.g. from 10~8 M to 10"13 M, e.g., from 10"9 M to 10"13 M).
[00121] "Affinity" refers to the strength of the sum total of non-covalent interactions between a single binding site of a molecule (e.g., a receptor) and its binding partner (e.g., a ligand). Unless indicated otherwise, as used herein,
"binding affinity" refers to intrinsic binding affinity which reflects a 1 : 1
30 WSGF interaction between members of a binding pair (e.g., s
and an antigen, or a receptor and its ligand). The affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (KD), which is the ratio of dissociation and association rate constants (k0ff and kon, respectively). Thus, equivalent affinities may comprise different rate constants, as long as the ratio of the rate constants remains the same. Affinity can be measured by well established methods known in the art, including those described herein. A particular method for measuring affinity is Surface Plasmon Resonance (SPR).
[00122] "Reduced binding", for example reduced binding to an Fc receptor, refers to a decrease in affinity for the respective interaction, as measured for example by SPR. For clarity the term includes also reduction of the affinity to zero (or below the detection limit of the analytic method), i.e. complete abolishment of the interaction. Conversely, "increased binding" refers to an increase in binding affinity for the respective interaction.
[00123] An "activating T cell antigen" as used herein refers to an antigenic determinant expressed on the surface of a T lymphocyte, particularly a cytotoxic T lymphocyte, which is capable of inducing T cell activation upon interaction with an antigen binding molecule. Specifically, interaction of an antigen binding molecule with an activating T cell antigen may induce T cell activation by triggering the signaling cascade of the T cell receptor complex. In a particular embodiment the activating T cell antigen is CD3.
[00124] "T cell activation" as used herein refers to one or more cellular response of a T lymphocyte, particularly a cytotoxic T lymphocyte, selected from: proliferation, differentiation, cytokine secretion, cytotoxic effector molecule release, cytotoxic activity, and expression of activation markers. The
T cell activating bispecific antigen binding molecules of the invention are capable of inducing T cell activation. Suitable assays to measure T cell activation are known in the art described herein.
[00125] A "target cell antigen" as used herein refers to an antigenic determinant presented on the surface of a target cell, for example a B cell in a
31 WSGF tumor such as a cancer cell or a cell of the tumor stror
terms "first" and "second" with respect to antigen binding moieties etc., are used for convenience of distinguishing when there is more than one of each type of moiety. Use of these terms is not intended to confer a specific order or orientation of the T cell activating bispecific antigen binding molecule unless explicitly so stated.
[00126] A "Fab molecule" refers to a protein consisting of the VH and CH1 domain of the heavy chain (the "Fab heavy chain") and the VL and CL domain of the light chain (the "Fab light chain") of an immunoglobulin.
[00127] By "fused" is meant that the components (e.g. a Fab molecule and an Fc domain subunit) are linked by peptide bonds, either directly or via one or more peptide linkers.
[00128] As used herein, the term "single-chain" refers to a molecule comprising amino acid monomers linearly linked by peptide bonds. In certain embodiments, one of the antigen binding moieties is a single-chain Fab molecule, i.e. a Fab molecule wherein the Fab light chain and the Fab heavy chain are connected by a peptide linker to form a single peptide chain. In a particular such embodiment, the C-terminus of the Fab light chain is connected to the N-terminus of the Fab heavy chain in the single-chain Fab molecule. In certain other embodiments, one of the antigen binding moieties is a single- chain Fv molecule
[00129] By a "crossover" Fab molecule (also termed "Crossfab") is meant a
Fab molecule wherein either the variable regions or the constant regions of the
Fab heavy and light chain are exchanged, i.e. the crossover Fab molecule comprises a peptide chain composed of the light chain variable region and the heavy chain constant region, and a peptide chain composed of the heavy chain variable region and the light chain constant region. For clarity, in a crossover
Fab molecule wherein the variable regions of the Fab light chain and the Fab heavy chain are exchanged, the peptide chain comprising the heavy chain constant region is referred to herein as the "heavy chain" of the crossover Fab molecule. Conversely, in a crossover Fab molecule wherein the constant
32 WSGF regions of the Fab light chain and the Fab heavy chair
peptide chain comprising the heavy chain variable region is referred to herein as the "heavy chain" of the crossover Fab molecule.
[00130] "Framework" or "FR" refers to variable domain residues other than hypervariable region (HVR) residues. The FR of a variable domain generally consists of four FR domains: FR1 , FR2, FR3, and FR4. Accordingly, the HVR and FR sequences generally appear in the following sequence in VH (or VL): FR1 -H1 (L1 )-FR2-H2(L2)-FR3-H3(L3)-FR4.
[00131] The "class" of an antibody or immunoglobulin refers to the type of constant domain or constant region possessed by its heavy chain. There are five major classes of antibodies: IgA, IgD, IgE,
[00132] IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgGi, lgG2, lgG3, lgG , IgAi, and lgA2. The heavy chain constant domains that correspond to the different classes of
immunoglobulins are called α, δ, ε, γ, and μ, respectively.
[00133] The term "Fc domain" or "Fc region" herein is used to define a C- terminal region of an immunoglobulin heavy chain that contains at least a portion of the constant region. The term includes native sequence Fc regions and variant Fc regions. Although the boundaries of the Fc region of an IgG heavy chain might vary slightly, the human IgG heavy chain Fc region is usually defined to extend from Cys226, or from Pro230, to the carboxyl-terminus of the heavy chain. However, the C-terminal lysine (Lys447) of the Fc region may or may not be present. Unless otherwise specified herein, numbering of amino acid residues in the Fc region or constant region is according to the EU numbering system, also called the EU index, as described in Kabat et al, Sequences of Proteins of Immunological Interest, 5th Ed. Public Health
Service, National Institutes of Health, Bethesda, MD, 1991 . A "subunit" of an Fc domain as used herein refers to one of the two polypeptides forming the dimeric Fc domain, i.e. a polypeptide comprising C-terminal constant regions of an immunoglobulin heavy chain, capable of stable self-association. For
33 WSGF example, a subunit of an IgG Fc domain comprises ar
CH3 constant domain.
[00134] A "modification promoting the association of the first and the second subunit of the Fc domain" is a manipulation of the peptide backbone or the post-translational modifications of an Fc domain subunit that reduces or prevents the association of a polypeptide comprising the Fc domain subunit with an identical polypeptide to form a homodimer. A modification promoting association as used herein particularly includes separate modifications made to each of the two Fc domain subunits desired to associate (i.e. the first and the second subunit of the Fc domain), wherein the promote association of the two Fc domain subunits and the formation of heterodimers. For example in certain embodiments,, a modification promoting association may alter the structure or charge of one or both of the Fc domain subunits so as to make their
association favorable.
[00135] The term "effector functions" refers to those biological activities attributable to the Fc region of an antibody, which vary with the antibody isotype. Examples of antibody effector functions include: Clq binding and complement dependent cytotoxicity (CDC), Fc receptor binding, antibody- dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (AD CP), cytokine secretion, immune complex-mediated antigen uptake by antigen presenting cells, down regulation of cell surface receptors (e.g. B cell receptor), and B cell activation.
[00136] As used herein, "albumin" refers collectively to albumin protein or amino acid sequence, or an albumin segment or variant, having one or more functional activities (e.g., biological activities) of albumin. In particular "albumin" refers to human albumin or segments thereof (see for example, EP 201 239, EP 322 094 WO 97/24445, WO95/23857) especially the mature form of human albumin, or albumin from other vertebrates, or segments thereof, or analogs or variants of these molecules or fragments thereof. Inc ertain embodiments, albumin refers to a truncated version of albumin.
34 WSGF
[00137] The term "quasi-albumin" refers to a heter
has structure and/or function similar to the whole albumin, and wherein said heteromultimer molecule is formed by the assembly of two or more monomeric polypeptides designed based on the sequence of the whole albumin. In certain embodiments, the monomeric polypeptides are "segments" that preferentially associate as heteromultimeric pairs to form a quasi-protein. In some
embodiments, the quasi-albumin has 90% of the activity of the whole albumin. In some embodiments, the quasi-albumin has 75% of the activity of whole- albumin. In an embodiment, the quasi-albumin has 50% of the activity of whole albumin. In some embodiments, the quasi-albumin has 50-75% of the activity of whole albumin. In an embodiment, quasi-albumin has 80% of the activity of whole albumin. In some embodiments, the quasi-albumin has 90% of the structure of whole albumin as determined by molecular modeling. In some embodiments, the quasi-albumin has 80% of the structure of whole albumin as determined by molecular modeling. In some embodiments, the quasi-albumin has 70% of the structure of whole albumin as determined by molecular modeling. In some embodiments, the quasi-albumin has 50% of the structure of whole albumin as determined by molecular modeling. In some embodiments, the quasi-albumin has 50%-75% of the structure of whole albumin as determined by molecular modeling.
[00138] The terms, human serum albumin (HSA) and human albumin (HA) are used interchangeably herein. The terms, "albumin and serum albumin" are broader, and encompass human serum albumin (and fragments and variants thereof) as well as albumin from other species (and fragments and variants thereof).
[00139] In certain embodiments, each albumin-based construct described herein is based on a variant of normal HA. The term "variants" includes insertions, deletions and substitutions, either conservative or non conservative, where such changes do not substantially alter one or more of the oncotic, useful ligand-binding and non-immunogenic properties of albumin, or the active
35 WSGF site, or active domain which confers the therapeutic a<
proteins.
[00140] In certain embodiments, the isolated heteromultimeric constructs described herein include naturally occurring polymorphic variants of human albumin and fragments of human albumin, for example those fragments disclosed in EP 322 094 (namely HA (Pn), where n is 369 to 419).
[00141] In certain embodiments, the albumin is derived from any vertebrate, especially any mammal that includes but is not limited to human, cow, sheep, rat, mouse, rabbit, horse, dog or pig. In certain embodiments, the albumin is derived from non-mammalian albumins including, but are not limited to hen and salmon.
[00142] The sequence of human albumin is as shown:SEQ ID NO: 1
[00143] MKWVTFISLLFLFSSAYSRGVFRRDAHKSEVAHRFKDLGEENFKA
LVLIAFAQYLQQCPFEDHVKLVNEVTEFAKTCVADESAENCDKSLHTLFGDKL
CTVATLRETYGEMADCCAKQEPERNECFLQHKDDNPNLPRLVRPEVDVMCT
AFHDNEETFLKKYLYEIARRHPYFYAPELLFFAKRYKAAFTECCQAADKAACL
LPKLDELRDEGKASSAKQRLKCASLQKFGERAFKAWAVARLSQRFPKAEFAE
VSKLVTDLTKVHTECCHGDLLECADDRADLAKYICENQDSISSKLKECCEKPL
LEKSHCIAEVENDEMPADLPSLAADFVESKDVCKNYAEAKDVFLGMFLYEYA
RRHPDYSVVLLLRLAKTYETTLEKCCAAADPHECYAKVFDEFKPLVEEPQNLI
KQNCELFEQLGEYKFQNALLVRYTKKVPQVSTPTLVEVSRNLGKVGSKCCKH
PEAKRMPCAEDYLSVVLNQLCVLHEKTPVSDRVTKCCTESLVNRRPCFSALE
VDETYVPKEFNAETFTFHADICTLSEKERQIKKQTALVELVKHKPKATKEQLKA
VMDDFAAFVEKCCKADDKETCFAEEGKKLVAASQAALGL
[00144] An "alloalbumin" is a genetic variant of albumin. In certain embodiments the alloalbumin is human alloalbumin (HAA). Alloalbumins that differ in electrophoretic mobility from albumin have been identified through population genetics surveys in the course of clinical electrophoresis, or in blood donor surveys. As markers of mutation and migration, alloalbumins are of interest to geneticists, biochemists, and anthropologists, but most of these
36 WSGF alloalbumin are not associated with disease (Minchioti
29(8), 1007-1016(2008)).
[00145] Table 1 : List of substitutions comprised by various alloalbumins as compared to HA of SEQ ID NO: 1. Thermostability, half-life information and other HAAs are provided in Krogh-hansen et al. Biochim Biophys Acta 1747, 81 -88(
Figure imgf000040_0001
37 WSGF
Figure imgf000041_0001
38 WSGF
Figure imgf000042_0001
[00146]
[00147]
[00148] The term "segmentation" refers to a precise internal splice of the original protein sequence which results in "segments" of the protein sequence that preferentially associate as heteromultimers to form a quasi-protein.
[00149] Quasi-native Structure:
39 WSGF
[00150] With reference to a native protein or its str
proteins and/or 'quasi-native structures' present the native protein like functional and structural characteristics. Proteins are naturally dynamics molecules and display an ensemble of structural configurations although we ascribe a native structure to it, such as the one obtained by X-ray
crystallography. The alternate structural configurations observed in the ensemble of geometries of that protein can be deemed to be quasi-native structures relative to each other or relative to the structure observed in the crystal. On a different front, homologous proteins sequences or proteins belonging to common structural families tend to fold into similar structural geometries. The member proteins belonging to this family can be deemed to achieve a quasi-native structure relative to each other. Some of the unique sequences in the protein family could also exhibit similar functional attributes and hence can be referred to as quasi-native proteins relative to each other. In the case of heteromultimers described here comprising of two or more protein constructs each of which have a transporter polypeptide component, the transporter polypeptides assemble to form a quasi-native structure. The reference native protein in this case is the protein from which the transporter polypeptide is derived and the reference native structure is the structure of the monomeric protein from which the transporter polypeptide is derived. We describe a case where two or more different polypeptides self-assemble to form a heteromultimeric structural and exhibit functional characteristics like a native protein which itself is a monomeric entity. In certain embodiments, are provided heteromultimer constructs comprising transporter polypeptides derived from albumin that self-assemble to form a heteromultimer that exhibits native albumin like functional characteristics such as FcRn binding and structural characteristics. These heteromultimers are referred to as being quasi-native.
[00151] "CD3 complex" as described herein is a complex of at least five membrane-bound polypeptides in mature T-lymphocytes that are non- covalently associated with one another and with the T-cell receptor. The CD3 complex includes the gamma, delta, epsilon, zeta, and eta chains (also referred
40 WSGF to as subunits). Non-human monoclonal antibodies he
against some of these chains, as exemplified by the murine antibodies OKT3, SP34, UCHT1 or 64.1. (See e.g., June, et al., J. Immunol. 136:3945-3952 (1986); Yang, et al., J. Immunol. 137:1097-1 100 (1986); and Hayward, et al., Immunol. 64:87-92 (1988)). The expression of certain CD antigens is highly restricted to specific lineage, lymphohematopoietic cells and over the past several years, antibodies directed against lymphoid-specific antigens have been used to develop treatments that were effective either in vitro or in animal models (5-13). In this respect CD19 has proved to be a very useful target. CD19 is expressed in the whole B lineage from the pro B cell to the mature B cell, it is not shed, is uniformly expressed on all lymphoma cells, and is absent from stem cells.
[00152] The term "effective amount" as used herein refers to that amount of multispecific heteromultimer construct being administered, which will relieve to some extent one or more of the symptoms of the disease, condition or disorder being treated. Compositions containing a multispecific heteromultimeric construct described herein can be administered for prophylactic, enhancing, and/or therapeutic treatments.
[00153] As used herein, the terms "engineer, engineered, engineering", are considered to include any manipulation of the peptide backbone or the post- translational modifications of a naturally occurring or recombinant polypeptide or fragment thereof. Engineering includes modifications of the amino acid sequence, of the glycosylation pattern, or of the side chain group of individual amino acids, as well as combinations of these approaches. The engineered proteins are expressed and produced by standard molecular biology
techniques.
[00154] By "isolated nucleic acid molecule or polynucleotide" is intended a nucleic acid molecule, DNA or RNA, which has been removed from its native environment. For example, a recombinant polynucleotide encoding a polypeptide contained in a vector is considered isolated. Further examples of an isolated polynucleotide include recombinant polynucleotides maintained in
41 WSGF heterologous host cells or purified (partially or substar
solution. An isolated polynucleotide includes a polynucleotide molecule contained in cells that ordinarily contain the polynucleotide molecule, but the polynucleotide molecule is present extrachromosomally or at a chromosomal location that is different from its natural chromosomal location. Isolated RNA molecules include in vivo or in vitro RNA transcripts, as well as positive and negative strand forms, and double-stranded forms. Isolated polynucleotides or nucleic acids described herein, further include such molecules produced synthetically. In addition, a polynucleotide or a nucleic acid, in certain embodiments, include a regulatory element such as a promoter, ribosome binding site, or a transcription terminator.
[00155] By a nucleic acid or polynucleotide having a nucleotide sequence at least, for example, 95% "identical" to a reference nucleotide sequence of the present invention, it is intended that the nucleotide sequence of the
polynucleotide is identical to the reference sequence except that the
polynucleotide sequence may include up to five point mutations per each 100 nucleotides of the reference nucleotide sequence. In other words, to obtain a polynucleotide having a nucleotide sequence at least 95% identical to a reference nucleotide sequence, up to 5% of the nucleotides in the reference sequence may be deleted or substituted with another nucleotide, or a number of nucleotides up to 5% of the total nucleotides in the reference sequence may be inserted into the reference sequence. These alterations of the reference sequence may occur at the 5' or 3' terminal positions of the reference nucleotide sequence or anywhere between those terminal positions,
interspersed either individually among residues in the reference sequence or in one or more contiguous groups within the reference sequence. As a practical matter, whether any particular polynucleotide sequence is at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to a nucleotide sequence of the present invention can be determined conventionally using known computer programs, such as the ones discussed above for polypeptides (e.g. ALIGN-2).
42 WSGF
[00156] The term "expression cassette" refers to a
generated recombinantly or synthetically, with a series of specified nucleic acid elements that permit transcription of a particular nucleic acid in a target cell.
The recombinant expression cassette can be incorporated into a plasmid, chromosome, mitochondrial DNA, plastid DNA, virus, or nucleic acid fragment.
Typically, the recombinant expression cassette portion of an expression vector includes, among other sequences, a nucleic acid sequence to be transcribed and a promoter. In certain embodiments, the expression cassette of the invention comprises polynucleotide sequences that encode bispecific antigen binding molecules of the invention or fragments thereof.
[00157] The term "vector" or "expression vector" is synonymous with
"expression construct" and refers to a DNA molecule that is used to introduce and direct the expression of a specific gene to which it is operably associated in a target cell. The term includes the vector as a self-replicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced. The expression vector of the present invention comprises an expression cassette. Expression vectors allow transcription of large amounts of stable mRNA. Once the expression vector is inside the target cell, the ribonucleic acid molecule or protein that is encoded by the gene is produced by the cellular transcription and/or translation machinery. In one embodiment, the expression vector of the invention comprises an expression cassette that comprises polynucleotide sequences that encode bispecific antigen binding molecules of the invention or fragments thereof.
[00158] The terms "host cell", "host cell line," and "host cell culture" are used interchangeably and refer to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells. Host cells include
"transformants" and "transformed cells," which include the primary transformed cell and progeny derived therefrom without regard to the number of passages.
In certain embodiments, progeny are not completely identical in nucleic acid content to a parent cell, but may contain mutations. Mutant progeny that have the same function or biological activity as screened or selected for in the
43 WSGF originally transformed cell are included herein. A host
system that can be used to generate the bispecific antigen binding molecules of the present invention. Host cells include cultured cells, e.g. mammalian cultured cells, such as CHO cells, BHK cells, NSO cells, SP2/0 cells, YO myeloma cells, P3X63 mouse myeloma cells, PER cells, PER.C6 cells or hybridoma cells, yeast cells, insect cells, and plant cells, to name only a few, but also cells comprised within a transgenic animal, transgenic plant or cultured plant or animal tissue.
[00159] An "activating Fc receptor" is an Fc receptor that following engagement by an Fc domain of an antibody elicits signaling events that stimulate the receptor-bearing cell to perform effector functions. Human activating Fc receptors include FcyRllla (CD 16a), FcyRI (CD64), and FcyRlla (CD32).
[00160] Antibody-dependent cell-mediated cytotoxicity (ADCC) is an immune mechanism leading to the lysis of antibody-coated target cells by immune effector cells. The target cells are cells to which antibodies or derivatives thereof comprising an Fc region specifically bind, generally via the protein part that is N-terminal to the Fc region. As used herein, the term
"reduced ADCC" is defined as either a reduction in the number of target cells that are lysed in a given time, at a given concentration of antibody in the medium surrounding the target cells, by the mechanism of ADCC defined above, and/or an increase in the concentration of antibody in the medium surrounding the target cells, required to achieve the lysis of a given number of target cells in a given time, by the mechanism of ADCC. The reduction in
ADCC is relative to the ADCC mediated by the same antibody produced by the same type of host cells, using the same standard production, purification, formulation and storage methods (which are known to those skilled in the art), but that has not been engineered. For example the reduction in ADCC mediated by an antibody comprising in its Fc domain an amino acid substitution that reduces ADCC, is relative to the ADCC mediated by the same antibody without this amino acid substitution in the Fc domain.
44 WSGF
[00161] An "effective amount" of an agent such as
heteromultimer described herein, refers to the amount that is necessary to result in a physiological change in the cell or tissue to which it is administered.
[00162] A "therapeutically effective amount" of an agent, e.g. a
pharmaceutical composition comprising a multispecific heteromultimer described herein, refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result. A therapeutically effective amount of an agent for example eliminates, decreases, delays, minimizes or prevents adverse effects of a disease.
[00163] An "individual" or "subject" is a mammal. Mammals include, but are not limited to, domesticated animals (e.g. cows, sheep, cats, dogs, and horses), primates (e.g. humans and non-human primates such as monkeys), rabbits, and rodents (e.g. mice and rats). Particularly, the individual or subject is a human.
[00164] The term "pharmaceutical composition" refers to a preparation which is in such form as to permit the biological activity of a multispecific heteromultimer construct contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
[00165] A "pharmaceutically acceptable carrier" refers to an ingredient in a pharmaceutical composition, other than an active ingredient, which is nontoxic to a subject. A pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
[00166] As used herein, "treatment" (and grammatical variations thereof such as "treat" or "treating") refers to clinical intervention in an attempt to alter the natural course of a disease in the individual being treated, and can be performed either for prophylaxis or during the course of clinical pathology.
Desirable effects of treatment include, but are not limited to, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or
45 WSGF palliation of the disease state, and remission or impro1
embodiments, multispecific heteromultimer constructs described herein are used to delay development of a disease or to slow the progression of a disease. The term "instructions" is used to refer to instructions customarily included in commercial packages of therapeutic products that contain information about the indications, usage, dosage, administration, combination therapy, contraindications and/or warnings concerning the use of such therapeutic products.
[00167] The term "cross-species binding" or "interspecies binding" as used herein means binding of a binding domain described herein to the same target molecule in humans and other organisims for instance, but not restricted to non-chimpanzee primates. Thus, "cross-species binding" or "interspecies binding" is to be understood as an interspecies reactivity to the same molecule "X" (i.e. the homolog) expressed in different species, but not to a molecule other than "X". Cross-species specificity of a monoclonal antibody recognizing e.g. human CD3 epsilon, to a non-chimpanzee primate CD3 epsilon, e.g.
macaque CD3 epsilon, can be determined, for instance, by FACS analysis. The FACS analysis is carried out in a way that the respective monoclonal antibody is tested for binding to human and non-chimpanzee primate cells, e.g.
macaque cells, expressing said human and non-chimpanzee primate CD3 epsilon antigens, respectively. An appropriate assay is shown in the following examples. The above-mentioned subject matter applies mutatis mutandis for the PSCA, CD19, C-MET, Endosialin, EpCAM, IGF-1 R and FAPa antigen: Cross-species specificity of a monoclonal antibody recognizing e.g. human PSCA, CD19, C-MET, Endosialin, EpCAM, IGF-1 R or FAPa, to a non- chimpanzee primate PSCA, CD19, C-MET, Endosialin, EpCAM, IGF-1 R or FAPa, e.g. macaque PSCA, CD19, C-MET, Endosialin, EpCAM, IGF-1 R or FAPa, can be determined, for instance, by FACS analysis. The FACS analysis is carried out in a way that the respective monoclonal antibody is tested for binding to human and non-chimpanzee primate cells, e.g. macaque cells,
46 WSGF expressing said human and non-chimpanzee primate
Endosialin, EpCAM, IGF-1 R or FAPa antigens, respectively.
[00168]
[00169]
[00170] Detailed Description of Certain Embodiments
[00171] Immunoglobulin based multispecific heteromultimer constructs:
[00172] Provided herein are isolated multispecific heteromultimer constructs comprising a first polypeptide construct comprising a first heavy chain polypeptide and a CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; and a second polypeptide construct comprising a second heavy chain polypeptide which is different from said first heavy chain polypeptide, and an antigen binding polypeptide construct that binds to a target antigen on at least one B cell, wherein:at least one of said
CD3 binding polypeptide construct and said antigen binding polypeptide construct comprises a single chain Fv region; said multispecific heteromultimer construct simultaneously engages said at least one B cell and said at least one
CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell; and said first and second heavy chain polypeptides form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc which is formed with stability at least comparable to a native homodimeric Fc, and with purity such that when said multispecific heteromultimer construct is expressed from a mammalian cell in an expression product, said expression product comprises at least about 70% of said multispecific heteromultimer, and less than 10% monomers or homodimers of said first or second polypeptide constructs. In some embodiments, the expression product comprises at least about 75% of said multispecific heteromultimer, and less than 15% monomers or homodimers of said first or second polypeptide constructs. In certain embodiments, the expression product comprises at least about 80% of said multispecific heteromultimer, and less than 10% monomers or homodimers of said first or second polypeptide
47 WSGF constructs. In additional embodiments, the expression
least about 85% of said multispecific heteromultimer, and less than 10% monomers or homodimers of said first or second polypeptide constructs. In a further embodiment expression product comprises at least about 90% of said multispecific heteromultimer, and less than 10% monomers or homodimers of said first or second polypeptide constructs.
[00173] In certain embodiments is the isolated multispecific heteromultimer construct, wherein said first or second polypeptide construct is devoid of at least one of immunoglobulin light chain, and immunoglobulin first constant (CH1 ) region.
[00174] In certain embodiments is provided an isolated multispecific heteromultimer construct comprising: a first polypeptide construct comprising a first heavy chain polypeptide and a CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second heavy chain polypeptide which is different from said first heavy chain polypeptide, and wherein said second polypeptide construct does not comprise an antigen binding polypeptide construct that binds to a target antigen on a B cell; wherein: said multispecific heteromultimer construct simultaneously engages at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell; and said first and second heavy chain polypeptides form a heterodimeric Fc region comprising a variant
immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc, wherein: said heterodimeric
Fc is formed with stability at least comparable to a native homodimeric Fc, and said heterodimeric Fc is formed with purity such that when said multispecific heteromultimer construct is expressed from a stable mammalian cell in an expression product, said expression product comprises at least about 75% of said multispecific heteromultimer, and less than 10% monomers or
homodimers of said first or second polypeptide constructs. In some
embodiments, the heterodimeric Fc interacts with cell surface receptors such
48 WSGF as FcgRllb on the B-cell. In certain embodiment, the
engineered to interact preferentially with the FcgRllb receptor relative to the normal antibody.
[00175] Provided herein is an isolated multispecific heteromultimer construct comprising: a first polypeptide construct comprising a first heavy chain polypeptide and a CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second heavy chain polypeptide which is different from said first heavy chain polypeptide, and a steric modulator construct which exhibits negligible receptor binding; wherein: said multispecific heteromultimer construct simultaneously engages at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell; and said first and second heavy chain polypeptides form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc, wherein: said heterodimeric Fc is formed with stability at least comparable to a native homodimeric Fc, and said heterodimeric Fc is formed with purity such that when said multispecific heteromultimer construct is coexpressed from a stable mammalian cell in an expression product, said expression product comprises at least about 75% of said multispecific heteromultimer, and less than 10% monomers or homodimers of said first or second polypeptide constructs. In certain embodiment the steric modulator construct is actually incapable of binding any known target tissue or cell surface and thus functions as a dummy polypeptide arm that only plays a steric modulation role in the interactions of the multispecific heteromultimer construct.
In some embodiments, the steric modulator construct is a polypeptide sequence that helps modulate sterical features of the multispecific
heteromultimer as the multimer binds to T and/or B cells. In certain
embodiments the steric modulator construct comprises a polypeptide domain that is designed de-novo. In certain embodiments, the steric modulator construct comprises polypeptide domains obtained by engineering a known
49 WSGF polypeptide domain to remove its binding properties. F
embodiments, the steric modulator construct comprises an engineered Fab region or fragment thereof which is engineered to remove binding properties.
[00176] Provided is an isolated multispecific heteromultimer construct comprising: a first polypeptide construct comprising a first heavy chain polypeptide and a CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second heavy chain polypeptide which is different from said first heavy chain polypeptide, and wherein said second polypeptide construct does not comprise an antigen binding polypeptide construct; wherein: said multispecific heteromultimer construct simultaneously engages at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell; and said first and second heavy chain polypeptides form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc, wherein: said heterodimeric
Fc is formed with stability at least comparable to a native homodimeric Fc, and said heterodimeric Fc is formed with purity such that when said multispecific heteromultimer construct is coexpressed from a stable mammalian cell in an expression product, said expression product comprises at least about 75% of said multispecific heteromultimer, and less than 10% monomers or
homodimers of said first or second polypeptide constructs.
[00177] In some embodiments is the isolated multispecific heteromultimer construct described herein wherein the heterodimer Fc region comprises a variant CH2 domain or hinge comprising amino acid modifications that prevents functionally effective binding to all the Fcgamma receptors.
[00178] Provided are isolated multispecific heteromultimer constructs that bind at least one B cell with a valency greater than one, and simultaneously engage said at least one B cell and at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell. In certain embodiments, the multispecific heteromultimer comsprises: a first
50 WSGF polypeptide construct comprising a first heavy chain p
binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; and a second polypeptide construct comprising a second heavy chain polypeptide which is different from said first heavy chain polypeptide, and an antigen binding polypeptide construct that binds to a target antigen on at least one B cell; wherein: at least one of said CD3 binding polypeptide construct and said antigen binding polypeptide construct optionally comprises a single chain Fv region; said first and second heavy chain polypeptides form a heterodimeric Fc region comprising a variant
immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc with stability at least comparable to a native homodimeric Fc, and with purity such that when said multispecific heteromultimer construct is expressed from a mammalian cell in an expression product, said expression product comprises greater than 75% of said multispecific heteromultimer, and less than 10% monomers or
homodimers of said first or second polypeptide constructs. The said
multispecific heteromultimer construct is capable of interacting with the B-cell via the said antigen binding polypeptide construct on the second heavy chain as well as interaction via the said heterodimeric Fc with FcgRllb receptors on the B-cell to show valency greater than one during B-cell engagement.
[00179] In certain embodiments is the isolated multispecific heteromultimer described herein, wherein the heterodimer Fc region comprises a variant CH2 domain comprising amino acid modifications to promote selective binding of a Fcgamma receptor.
[00180] In some embodiuments is the heteromultimer wherein the variant CH2 domain selectively binds at least one of Fcgammallla and Fcgammallb receptor as compared to wild-type CH2 domain.
[00181] In certain embodiments is an isolated multispecific heteromultimer construct described herein wherein the heterodimer Fc is glycosylated.
[00182] In some embodiments is an isolated multispecific heteromultimer described herein, wherein the heterodimer Fc is afucosylated.
51 WSGF
[00183] In certain embodiments is an isolated mull
construct described herein wherein the heterodimer Fc is aglycosylated.
[00184]
[00185] Fc region modifications promoting heterodimerization:
[00186] Provded herein are multispecific heteromultimer comstructs that comprise different antigen binding moieties, fused to one or the other of the two subunits of the Fc domain, thus the two subunits of the Fc domain are typically comprised of two non-identical polypeptide chains. To improve the yield and purity of the heteromultimers described herein, the Fc region of the
polypeptides is modified to promote the association of the desired polypeptides.
[00187] In some embodiments, the first and second heavy chain
polypeptides of the heteromultimer constructs described herein form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc which is formed with stability at least comparable to a native homodimeric Fc, and with purity such that when said multispecific heteromultimer construct is coexpressed from a mammalian cell as an expression product, said expression product comprises at least about 75% of said multispecific heteromultimer, and less than 10% monomers or
homodimers of said first or second polypeptide constructs.
[00188] In some embodiments, the first and second heavy chain
polypeptides of the heteromultimer constructs described herein form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc which is formed with stability at least comparable to a native homodimeric Fc, and with purity such that when said multispecific heteromultimer construct is co-expressed from a mammalian cell as an expression product, said expression product comprises at least about 90% of said multispecific heteromultimer, and less than 10% monomers or
homodimers of said first or second polypeptide constructs.
52 WSGF
[00189] In some embodiments, the first and secon
polypeptides of the heteromultimer constructs described herein form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc which is formed with stability at least comparable to a native homodimeric Fc, and with purity such that when said multispecific heteromultimer construct is co-expressed from a mammalian cell as an expression product, said expression product comprises at least about 95% of said multispecific heteromultimer, and less than 10% monomers or
homodimers of said first or second polypeptide constructs.
[00190] In some embodiments is the isolated multispecific heteromultimer provided herein, wherein the variant CH3 domain has a melting temperature (Tm) of about 73°C or greater.
[00191] In certain embodiments is the isolated multispecific heteromultimer described herein, wherein the heterodimer Fc region is formed with a purity greater than about 78%.
[00192] In am embodiment is the isolated multispecific heteromultimer described herein, wherein the heterodimer Fc region is formed with a purity of at least about 78% or greater and the Tm is at least about 75°C.
[00193] In some embodiments is the isolated multispecific heteromultimer described herein, wherein the heterodimer Fc region is formed with a purity of at least about 75% and the Tm is about 75°C or greater.
[00194] In certain embodiments are provided isolated multispecific heteromultimer constructs wherein: a) the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications L351 Y,
F405A, and Y407V, and the variant CH3 sequence of the second transporter polypeptide comprises the amino acid modifications T366L, K392M, and
T394W; b) the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications L351Y, F405A, and Y407V, and the variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications T366L, K392L, and T394W; c) the variant CH3
53 WSGF sequence of the first heavy chain polypeptide compris
modifications T350V, L351Y, F405A, and Y407V, and the variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications T350V, T366L, K392M, and T394W; d) the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications T350V, L351Y, F405A, and Y407V, and the variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications T350V, T366L, K392L, and T394W; e) the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications T366L, N390R, K392R, and T394W, and the variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications L351 Y, S400E, F405A, and Y407V; or f) the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications T350V, T366L, N390R, K392R, and T394W, and the variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications T350V, L351Y, S400E, F405A, and Y407V.
[00195]
[00196] Fc region modifications reducing Fc receptor binding and/or effector function:
[00197] In certain embodiments the Fc regions of the heteromultimer constructs described herein comprises a variant CH2 domain comprising amino acid modifications to promote selective binding of a Fcgamma receptor. In some embodiments, the isolated multispecific heteromultimer described herein comprise a variant CH2 domain that selectively binds a Fcgammallb receptor with an affinity greater than that of the wild-type CH2 domain. In some embodiments, the isolated multispecific heteromultimer described herein comprise a variant CH2 domain that selectively binds a FcgammallA and/or FcgammalllA receptor with an affinity greater than that of the wild-type CH2 domain.
[00198] In certain embodiments the Fc regions of the heteromultimer constructs described herein exhibit reduced binding affinity to an Fc receptor
54 WSGF and/or reduced effector function, as compared to a na
one such embodiment the Fc region exhibits less than 50%, alternatively less than 20%, alternatively less than 10% and in some embodiments, less than 5% of the binding affinity to an Fc receptor, as compared to a native lgG1 Fc region, and/or less than 50%, alternatively less than 20%, alternatively less than 10% and in some embodiments less than 5% of the effector function, as compared to a native lgG1 Fc region.
[00199] In one embodiment, the Fc region of a heteromutlimer construct described herein does not substantially bind to an Fc receptor or induce appreciable effector function. In a certain embodiment the Fc receptor is an Fey receptor. In one embodiment the Fc receptor is a mammalian Fc receptor. In certain embodiments, the mammalian Fc receptor is a human Fc receptor. In one embodiment the Fc receptor is an activating Fc receptor. In a specific embodiment the Fc receptor is an activating human Fey receptor, more specifically human FcyRllla, FcyRI or FcyRlla, most specifically human
FcyRllla. In one embodiment the effector function is one or more function selected from the group consisting of CDC, ADCC, ADCP, and cytokine secretion. In a particular embodiment the effector function is ADCC. In one embodiment the Fc region exhibits binding affinity to neonatal Fc receptor (FcRn). In certain embodiments, the FcRn binding affinity is substantially similar to that of a native lgG1 Fc. In some embodiments, substantially similar binding to FcRn is achieved when the Fc region of a heteromultimer construct described herein exhibits greater than about 70%, or in some embodiments greater than about 80%, and in some particular embodiments greater than about 90% of the binding affinity of a native lgG1 Fc domain to FcRn.
[00200] In certain embodiments the Fc region of a heteromultimer construct described herein is engineered to have reduced binding affinity to an Fc receptor and/or reduced effector function, as compared to a non-engineered Fc domain. In some embodiments, the engineered mutations are present in the lower hinge and CH2 domain. In particular embodiments, the Fc region of a
55 WSGF heteromultimer described herein comprises one or mc
that reduces the binding affinity of the Fc region to an Fc receptor and/or effector function. In some embodiments, the same one or more amino acid mutation is present in each of the two subunits of the Fc region. In some embodiments, different amino acid mutations are introduced in each of the two subunits of the Fc region. In one embodiment the amino acid mutation reduces the binding affinity of the Fc region to an Fc receptor. In one embodiment the amino acid mutation reduces the binding affinity of the Fc region to an Fc receptor by at least 2-fold, or in some embodiments at least 5-fold, or in an embodiment at least 10-fold. In certain embodiments where there is more than one amino acid mutation that reduces the binding affinity of the Fc region to the Fc receptor, the combination of these amino acid mutations reduces the binding affinity of the Fc region to an Fc receptor by at least 10-fold, or in some embodiments at least 20-fold, or in certain embodiments at least 50-fold. In certain embodiment, the binding affinity of the Fc region for the Fc receptor is reduced to an extent where there is no longer any detectable binding for the mutant Fc for the Fc receptor in standard binding assay such as using the SPR instrument. In one embodiment the heteromultimer construct described herein comprising an engineered Fc domain exhibits less than 20%, and in certain embodiments less than 10%, and in select embodiments less than 5% of the binding affinity to an Fc receptor as compared to a corresponding construct comprising an Fc domain which is not engineered to reduce binding to an Fc receptor. In a particular embodiment the Fc receptor is an Fey receptor. In some embodiments the Fc receptor is a human Fc receptor. In some embodiments the Fc receptor is an activating Fc receptor. In a specific embodiment the Fc receptor is an activating human Fey receptor which in certain embodiments is one of human FcyRllla, FcyRI and FcyRlla. In some embodiments, binding to each of these receptors is reduced. In some embodiments binding affinity to a complement component, for instance, but not restricted to C1 q, is also reduced. In one embodiment binding affinity to
56 WSGF neonatal Fc receptor (FcRn) is not reduced. In certain
region of a heteromultimer described herein is engineered to have reduced effector function, as compared to a non-engineered Fc region. In certain embodiments, the reduced effector function can include, but is not limited to, one or more of the following: reduced complement dependent cytotoxicity (CDC), reduced antibody-dependent cell-mediated cytotoxicity (ADCC), reduced antibody-dependent cellular phagocytosis (ADCP), reduced cytokine secretion, reduced immune complex-mediated antigen uptake by antigen- presenting cells, reduced binding to NK cells, reduced binding to macrophages, reduced binding to monocytes, reduced binding to polymorphonuclear cells, reduced direct signaling inducing apoptosis, reduced crosslinking of target- bound antibodies, reduced dendritic cell maturation, or reduced T cell priming. In one embodiment the reduced effector function is one or more selected from the group of reduced CDC, reduced ADCC, reduced ADCP, and reduced cytokine secretion. In certain embodiments the reduced effector function is reduced ADCC. In one embodiment the reduced ADCC is less than 20% of the ADCC induced by a non-engineered Fc domain (or a T cell activating bispecific antigen binding molecule comprising a non-engineered Fc domain). In another embodiment the reduced ADCC is less than 50% of the ADCC induced by a non-engineered Fc domain (or a T cell activating bispecific antigen binding molecule comprising a non-engineered Fc domain). In a further embodiment the reduced ADCC is less than 10% of the ADCC induced by a non-engineered Fc domain (or a T cell activating bispecific antigen binding molecule comprising a non-engineered Fc domain).
[00201] In certain embodiments is the isolated multispecific heteromultimer described herein wherein the heterodimer Fc region comprises a variant CH2 domain comprising amino acid modifications to promote selective binding of a Fcgamma receptor as described herein.
[00202] In some embodiments is an isolated multispecific heteromultimer described herein, wherein the variant CH2 domain selectively binds at least
57 WSGF one of Fcgammallla and Fcgammallb receptor as con
domain.
[00203] In certain embodiments the multispecific heteromultimer construct comprises a variant CH2 region that binds at least one B cell such that the heteromultimer construct binds B cells with a valency greater than one.
[00204]
[00205] Albumin-based multispecific heteromultimer constructs:
[00206] Provided are isolated multispecific heteromultimer constructs comprising: a first polypeptide construct comprising a first transporter polypeptide fused to at least one CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second transporter polypeptide which is different from said first transporter polypeptide, fused to at least one antigen binding polypeptide construct that binds to a target antigen on at least one B cell;
wherein said first and second transporter polypeptides are derived from a protein by segmentation of said protein, each transporter polypeptide comprising an amino acid sequence with at least 90% identity to a segment of said protein, and wherein said transporter polypeptides self-assemble to form a quasi-native structure of said monomeric protein. In some embodiments, each transporter polypeptide comprises an amino acid sequence with at least 85% identity to a segment of the protein. In some embodiments, each transporter polypeptide comprises an amino acid sequence with at least 80% identity to a segment of the protein. In some embodiments, each transporter polypeptide comprises an amino acid sequence with at least 95% identity to a segment of the protein. In some other embodiments, each transporter polypeptide comprises an amino acid sequence with at least 99% identity to a segment of the protein.
[00207] In certain embodiments is the isolated multispecific heteromultimer described herein, wherein said transporter polypeptides are not derived from an antibody. In a further embodiment is an isolated multispecific heteromultimer described herein, wherein each transporter polypeptide is an albumin
58 WSGF derivative. In some embidments is an albumin based i
heteromultimer, wherein said albumin is human serum albumin. In some embodiments, at least one transporter polypeptide is an allo-albumin derivative. In certain embodiments is an isolated multispecific heteromultimer described herein wherein each transporter polypeptide is derived from a different alloalbumin. In some embodiments, each transporter polypeptide comprises an amino acid sequence with at least 75% identity to a segment of albumin. In some embodiments, each transporter polypeptide comprises an amino acid sequence with at least 80% identity to a segment of albumin. In some embodiments, each transporter polypeptide comprises an amino acid sequence with at least 90% identity to a segment of albumin. In some other embodiments, each transporter polypeptide comprises an amino acid sequence with at least 95% identity to a segment of albumin. In some other embodiments, each transporter polypeptide comprises an amino acid sequence with at least 99% identity to a segment of albumin.
[00208] Provided herein is an albumin based isolated multispecific heteromultimer construct comprising: a first monomer comprising a first transporter polypeptide fused to at least one CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell; a second polypeptide construct comprising a second transporter polypeptide which is different from said first transporter polypeptide, fused to at least one antigen binding polypeptide construct that binds to a target antigen on at least one B cell; wherein said first and second transporter polypeptides are obtained by segmentation of albumin, and each transporter polypeptide comprising an amino acid sequence with at least 90% identity to a segment of albumin such that said transporter polypeptides self-assemble to form quasi-native albumin, and wherein said first cargo polypeptide does not have any binding domain present in said second cargo polypeptide.
[00209] Provided herein are albumin based multispecific heteromultimer constructs as described above, wherein said first transporter polypeptide comprising at least one mutation selected from A194C, L198C, W214C,
59 WSGF
A217C, L331 C and A335C. In certain embodiments, tl
polypeptide comprises at least one mutation selected from L331 C, A335C,
V343C, L346C, A350C, V455C, and N458C.
[00210] Provided are isolated multispecific heteromultimer constructs described herein wherein said multispecific heteromultimer construct simultaneously engages said at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell.
[00211] In certain embodiments, are provided heteromultimer constructs comprising transporter polypeptides derived from albumin that self-assemble to form a heteromultimer that exhibits native albumin like functional characteristics such as FcRn binding and structural characteristics. In certain embodiments, the albumin based heteromultimer constructs described herein when
administered to a person in need, home to tumor cells. In some embodiments, the tumor cells are from a solid tumor. In some embodiments, the
heteromultimer constructs described herein home to tumor cells and
subsequently bind to said tumor cells. In certain embodiments, the
heteromultimer constructs described herein home to at least one tumor cell, bind simultaneously to said at least one tumor cell and and at least one T-cell in a manner that results in the lysis of said tumor cell. In certain embodiments, the heteromultimer constructs described herein home to at least one tumor cell, bind simultaneously to said at least one tumor cell and and at least one T-cell such that the binding to said tumor cell is with a higher valency than the binding to said T-cell, and causes the lysis of said tumor cell.
[00212] CD3 complex binding polypeptide constructs:
[00213] In certain embodiments of the immunoglobulin-based and albumin- based multispecific heteromultimer constructs provided herein, said
heteromultimer construct comprises at least one CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell. In some embodiments, the at least one CD3 binding polypeptide construct
60 WSGF comprises at least one CD3 binding domain from a Ct
nanobody, fibronectin, affibody, anticalin, cysteine knot protein, DARPin, avimer, Kunitz domain or variant or derivative thereof. In some embodiments, the at least one CD3 binding domain comprises at least one amino acid modification that reduces immunogenicity as compared to a corresponding CD3 binding domain not comprising said modification. In an embodiment, the at least one CD3 binding domain comprises at least one amino acid modification that increases its stability as measured by Tm, as compared to a corresponding CD3 binding domain not comprising said modification. In some embodiments, there is about a 3 degree increase in the Tm as compared to the native CD3 binding domain not comprising said at least one modification. In some embodiments, there is about a 5 degree increase in the Tm as compared to the native CD3 binding domain not comprising said at least one modification. In some embodiments, there is about a 8 degree increase in the Tm as compared to the native CD3 binding domain not comprising said at least one modification. In some embodiments, there is about a 10 degree increase in the Tm as compared to the native CD3 binding domain not comprising said at least one modification.
[00214] In some embodiments, the at least one CD3 binding polypeptide construct described herein comprises at least one CD3 binding domain from a CD3 specific antibody wherein said CD3 specific antibody is a heavy chain antibody devoid of light chains.
[00215] In certain other embodiments, the at least one CD3 binding polypeptide construct described herein comprises at least one CD3 binding domain derived from a non-antibody protein scaffold domain.
[00216] In certain embodiments, the CD3 binding polypeptide constructs are CD3 binding Fab constructs (i.e. antigen binding constructs comprising a heavy and a light chain, each comprising a variable and a constant region). In some embodiment said Fab construct is mammalian. In one embodiment said
Fab construct is human. In another embodiment said Fab construct is humanized. In yet another embodiment said Fab contruct comprises at least
61 WSGF one of human heavy and light chain constant regions,
said Fab construct is a single chain Fab (scFab).
[00217] In certain embodiments the CD3 binding polypeptide constructs comprise CD3 binding scFab constructs wherein the C-terminus of the Fab light chain is connected to the N-terminus of the Fab heavy chain by a peptide linker. The peptide linker allows arrangement of the Fab heavy and light chain to form a functional CD3 binding moiety. In certain embodiments, the peptide linkers suitable for connecting the Fab heavy and light chain include sequences comprising glycine-serine linkers for instance, but not limited to (GmS)n-GG, (SGn)m, (SEGn)m, wherein m and n are between 0-20. In certain embodiments, the scFab construct is a cross-over construct wherein the constant regions of the Fab light chain and the Fab heavy chain are exchanged. In another embodiment of a cross-over Fab, the variable regions of the Fab light chain and the Fab heavy chain are exchanged.
[00218] In certain embodiments, the CD3 binding polypeptide constructs comprise CD3 binding Fv constructs (i.e. antigen binding constructs comprising a heavy and a light chain, each comprising a variable region). In some embodiment said Fv construct is mammalian. In one embodiment said Fv construct is human. In another embodiment said Fv construct is humanized. In yet another embodiment said Fv contruct comprises at least one of human heavy and light chain variable regions. In a further embodiment said Fv construct is a single chain Fv (scFv).
[00219] In some embodiments, the CD3 binding polypeptide construct of a multispecific heteromultimer construct described herein bind to at least one component of the CD3 complex. In a specific embodiment, the CD3 binding polypeptide construct binds to at least one of CD3 epsilon, CD3 gamma, CD3 delta or CD3 zeta of the CD3 complex. In certain embodiments, the CD3 binding polypeptide construct binds the CD3epsilon domain. In certain embodiments, binding polypeptide construct binds a human CD3 complex. In certain embodiments, the CD3 binding polypeptide construct exhibits cross- species binding to a least one member of the CD3 complex.
62 WSGF
[00220] Provided herein are immunoglobulin-base
multispecific heteromultimer constructs comprising at least one CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell, where in the CD3 expressing cell is a T-cell. In certain embodiments, the CD3 expressing cell is a human cell. In some embodiments, the CD3 expressing cell is a non-human, mammalian cell. In some
embodiments, the T cell is a cytotoxic T cell. In some embodiments the T cell is a CD4+ or a CD8+ T cell.
[00221] In certain embodiments of the immunoglobulin-based and albumin- based multispecific heteromultimer constructs provided herein, the construct is capable of activating and redirecting cytotoxic activity of a T cell to a target cell such as a B cell. In a particular embodiment, said redirection is independent of MHC-mediated peptide antigen presentation by the target cell and and/or specificity of the T cell.
[00222] Provided herein are hetromultimer constructs that are capable of simultaneous binding to a B cell antigen for instance a tumor cell antigen, and an activating T cell antigen. In one embodiment, the heteromultimer construct is capable of crosslinking a T cell and a target B cell by simultaneous binding to a B cell antigen for instance CD19 or CD20 and an activating T cell antigen for instance CD3. In one embodiment, the simultaneous binding results in lysis of a target B cell, for instance a tumor cell. In one embodiment, such simultaneous binding results in activation of the T cell. In other embodiments, such simultaneous binding results in a cellular response of a T lymphocyte, for instance a cytotoxic T lymphocyte, selected from the group of: proliferation, differentiation, cytokine secretion, cytotoxic effector molecule release, cytotoxic activity, and expression of activation markers. In one embodiment, binding of the T cell activating bispecific antigen binding molecule to the activating T cell antigen without simultaneous binding to the target cell antigen does not result in T cell activation.
[00223]
[00224] B cell binding polypeptide constructs:
63 WSGF
[00225] Provided herein are isolated heteromultirrn
at least one antigen binding polypeptide construct that binds to a target antigen on at least one B cell. In certain embodiments, the antigen binding polypeptide construct binds at least one member of a B cell CD21 -CD19-CD81 complex. In some embodiments, the antigen binding polypeptide construct comprises at least one CD19 binding domain or fragment thereof. In an embodiment, the antigen binding polypeptide construct comprises at least one CD20 binding domain.
[00226] In some embodiments, the at least one antigen binding domain is a CD19 or CD20 binding domain which is obtained from a CD19 or CD20 specific antibody, a nanobody, fibronectin, affibody, anticalin, cysteine knot protein, DARPin, avimer, Kunitz domain or variant or derivative thereof. In some embodiments, the at least one antigen binding polypeptide construct described herein comprises at least one antigen binding domain which is a CD19 or CD20 binding domain from an antibody which is a heavy chain antibody devoid of light chains.
[00227] In some embodiments, the at least one antigen binding domain is a CD19 or CD20 binding domain that comprises at least one amino acid modification that reduces immunogenicity as compared to a corresponding antigen binding domain not comprising said modification. In an embodiment, the at least one antigen binding domain is a CD19 or CD20 binding domain comprising at least one amino acid modification that increases its stability as measured by Tm, as compared to a corresponding domain not comprising said modification.
[00228] In certain embodiments, the at least one antigen binding
polypeptide construct is a Fab construct that binds at least one of CD19 and
CD20 on a B cell. In some embodiment said Fab construct is mammalian. In one embodiment said Fab construct is human. In another embodiment said Fab construct is humanized. In yet another embodiment said Fab contruct comprises at least one of human heavy and light chain constant regions. In a further embodiment said Fab construct is a single chain Fab (scFab).
64 WSGF
[00229] In certain embodiments the CD19 and/or (
construct comprises a scFab construct wherein the C-terminus of the Fab light chain is connected to the N-terminus of the Fab heavy chain by a peptide linker. The peptide linker allows arrangement of the Fab heavy and light chain to form a functional CD19 and/or CD20 binding moiety. In certain
embodiments, the peptide linkers suitable for connecting the Fab heavy and light chain include sequences comprising glycine-serine linkers for instance, but not limited to (GmS)n-GG, (SGn)m, (SEGn)m, wherein m and n are between 0-20. In certain embodiments, the scFab construct is a cross-over construct wherein the constant regions of the Fab light chain and the Fab heavy chain are exchanged. In another embodiment of a cross-over Fab, the variable regions of the Fab light chain and the Fab heavy chain are exchanged.
[00230] In certain embodiments, the at least one antigen binding
polypeptide construct is a Fv construct that binds at least one of CD19 and CD20 on a B cell. In some embodiment said Fv construct is mammalian. In one embodiment said Fv construct is human. In another embodiment said Fv construct is humanized. In yet another embodiment said Fv contruct comprises at least one of human heavy and light chain variable regions. In a further embodiment said Fv construct is a single chain Fv (scFv).
[00231] In certain embodiments, the antigen binding polypeptide construct exhibits cross-species binding to a least one antigen expressed on the surface of a B cell. In some embodiments, the antigen binding polypeptide construct of a multispecific heteromultimer construct described herein bind to at least one of mammalian CD19 and CD20. In certain embodiments, binding polypeptide construct binds a human CD19 or CD20.
[00232] Provided herein are hetromultimer constructs that are capable of simultaneous binding to a B cell antigen for instance a tumor cell antigen, and an activating T cell antigen. In one embodiment, the heteromultimer construct is capable of crosslinking a T cell and a target B cell by simultaneous binding to a B cell antigen for instance CD19 or CD20 and an activating T cell antigen for instance CD3.
65 WSGF
[00233] In certain embodiments, a heteromultimer
comprises at least one antigen binding polypeptide construct that binds to a target antigen such as a CD19 or CD20 on at least one B cell associated with a disease. In some embodiments, the disease is a cancer selected from a carcinoma, a sarcoma, leukaemia, lymphoma and glioma. In an embodiment, the cancer is at least one of squamous cell carcinoma, adenocarcinoma, transition cell carcinoma, osteosarcoma and soft tissue sarcoma. In certain embodiments, the at least one B cell is an autoimmune reactive cell that is a lymphoid or myeloid cell.
[00234]
[00235] Additional Antigen binding constructs:
[00236] In certain embodiments, an albumin or immunoglobulin based multispecific heteromultimer construct described herein further comprises at least one binding domain that binds at least one of: EpCAM, EGFR, IGFR, HER-2 neu, HER-3, HER-4, PSMA, CEA, MUC-1 (mucin), MUC2, MUC3, MUC4, MUC5, MUC7, CCR4, CCR5, CD19, CD20, CD33, CD30, ganglioside GD3, 9-0-Acetyl-GD3, GM2, Poly SA, GD2, Carboanhydrase IX (MN/CA IX), CD44v6, Sonic Hedgehog (Shh), Wue-1 , Plasma Cell Antigen, (membrane- bound), Melanoma Chondroitin Sulfate Proteoglycan (MCSP), CCR8, TNF- alpha precursor, STEAP, mesothelin, A33 Antigen, Prostate Stem Cell Antigen (PSCA), Ly-6; desmoglein 4, E-cadherin neoepitope, Fetal Acetylcholine Receptor, CD25, CA19-9 marker, CA-125 marker and Muellerian Inhibitory Substance (MIS) Receptor type II, sTn (sialylated Tn antigen; TAG-72), FAP (fibroblast activation antigen), endosialin, LG, SAS, EPHA4 CD63, CD3 BsAb immunocytokines TNF which comprise a CD3 antibody attached to the cytokine, IFNy, IL-2, and TRAIL.
[00237]
[00238] Post translational modifications:
[00239] In certain embodiments are multispecific heteromultimer constructs described herein, which are differentially modified during or after translation. In some embodiments, the modification is at least one of: glycosylation,
66 WSGF acetylation, phosphorylation, amidation, derivatization
protecting/blocking groups, proteolytic cleavage and linkage to an antibody molecule or other cellular ligand. In some embodiments, the heteromultimer construct is chemically modified by known techniques, including but not limited, to specific chemical cleavage by cyanogen bromide, trypsin, chymotrypsin, papain, V8 protease, NaBH4 ; acetylation, formylation, oxidation, reduction; and metabolic synthesis in the presence of tunicamycin.
[00240] Additional post-translational modifications of heteromultimers described herein include, for example, N-linked or O-linked carbohydrate chains, processing of N-terminal or C-terminal ends), attachment of chemical moieties to the amino acid backbone, chemical modifications of N-linked or O- linked carbohydrate chains, and addition or deletion of an N-terminal methionine residue as a result of procaryotic host cell expression. The heteromultimer constructs described herein are modified with a detectable label, such as an enzymatic, fluorescent, isotopic or affinity label to allow for detection and isolation of the protein. In certain embodiments, examples of suitable enzyme labels include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol;
examples of bioluminescent materials include luciferase, luciferin, and aequorin; and examples of suitable radioactive material include iodine, carbon, sulfur, tritium, indium, technetium, thallium, gallium, palladium, molybdenum, xenon, fluorine.
[00241] In specific embodiments, heteromultimer constructs described herein are attached to macrocyclic chelators that associate with radiometal ions.
[00242] In some embodiments, the heteromultimer constructs described herein are modified by either natural processes, such as post-translational
67 WSGF processing, or by chemical modification techniques wl
art. In certain embodiments, the same type of modification may be present in the same or varying degrees at several sites in a given polypeptide. In certain embodiments, polypeptides from heteromultimers described herein are branched, for example, as a result of ubiquitination, and in some embodiments are cyclic, with or without branching. Cyclic, branched, and branched cyclic polypeptides are a result from posttranslation natural processes or made by synthetic methods. Modifications include acetylation, acylation, ADP- ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination. (See, for instance, PROTEINS-STRUCTURE AND MOLECULAR PROPERTIES, 2nd Ed., T. E. Creighton, W. H. Freeman and Company, New York (1993); POST- TRANSLATIONAL COVALENT MODIFICATION OF PROTEINS, B. C.
Johnson, Ed., Academic Press, New York, pgs. 1 -12 (1983); Seifter et al., Meth. Enzymol. 182:626-646 (1990); Rattan et al., Ann. N.Y. Acad. Sci. 663:48- 62 (1992)).
[00243] In certain embodiments, heteromultimeric constructs described herein are attached to solid supports, which are particularly useful for immunoassays or purification of polypeptides that are bound by, that bind to, or associate with albumin fusion proteins of the invention. Such solid supports include, but are not limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene.
[00244]
68 WSGF
[00245] Polynucleotides:
[00246] Provided herein are polynucleotide constructs encoding
multispecific heteromultimer constructs described herein. In certain
embodiments the polynucleotide or nucleic acid is DNA. In other embodiments, a polynucleotide described herein is RNA, for example, in the form of messenger RNA (mRNA). RNA of the present invention may be single stranded or double stranded.
[00247] In certain embodiments is a set of expression vectors for expressing a multispecific heteromultimer construct described herein which comprises a first and a second polypeptide construct, said set comprising at least a first DNA sequence encoding said first polypeptide construct and at least a second DNA sequence encoding said second polypeptide construct.
[00248] In certain embodiments are polynucleotide sequences encoding a heteromultimer construct described herein or a polypeptide construct thereof with sequence as provided herein. In certain embodiments is a polynucleotide comprising a sequence that is at least about 80%, 85%, 90%, 95%, 96%, 97%,
98%, or 99% identical to a nucleotide sequence shown in Figure . In certain embodiments are polynucleotide sequences encoding a heteromultimer construct described herein or a polypeptide construct thereof, wherein said polynucleotide comprises conservative mutations of the sequence as provided herein.
[00249]
[00250] Methods of Recombinant and Synthetic Production of Multispecific Heteromultimer Constructs:
[00251] Provided are methods of producing an expression product containing a multispecific heteromultimer construct as described herein, in stable mammalian cells, the method comprising: transfecting at least one mammalian cell with: at least a first DNA sequence encoding said first polypeptide construct and at least a second DNA sequence encoding said second polypeptide construct, such that said at least one first DNA sequence, said at least one second DNA sequence are transfected in said at least one
69 WSGF mammalian cell in a pre-determined ratio to generate
culturing said stable mammalian cells to produce said expression product comprising said multispecific heteromultimer. In certain embodiments, said predetermined ratio of the at least one first DNA sequence: at least one second DNA sequence is about 1 :1 . In certain other embodiments, said predetermined ratio of the at least one first DNA sequence: at least one second DNA sequence is skewed towards a larger amount of the one first DNA sequence such as about 2:1. In yet other embodiments, said predetermined ratio of the at least one first DNA sequence: at least one second DNA sequence is skewed towards a larger amount of the one first DNA sequence such as about 1 :2. In select embodiments, the mammalian cell is selected from the group consisting of a VERO, HeLa, HEK, NSO, Chinese Hamster Ovary (CHO), W138, BHK, COS-7, Caco-2 and MDCK cell, and subclasses and variants thereof.
[00252] In certain embodiments are heteromultimers produced as recombinant molecules by secretion from yeast, a microorganism such as a bacterium, or a human or animal cell line. In embodiments, the polypeptides are secreted from the host cells.
[00253] Embodiments include a cell, such as a yeast cell transformed to express a heteromultimer protein described herein. In addition to the
transformed host cells themselves, are provided culture of those cells, preferably a monoclonal (clonally homogeneous) culture, or a culture derived from a monoclonal culture, in a nutrient medium. If the polypeptide is secreted, the medium will contain the polypeptide, with the cells, or without the cells if they have been filtered or centrifuged away. Many expression systems are known and may be used, including bacteria (for example E. coli and Bacillus subtilis), yeasts (for example Saccharomyces cerevisiae, Kluyveromyces lactis and Pichia pastoris, filamentous fungi (for example Aspergillus), plant cells, animal cells and insect cells.
[00254] A heteromultimer described herein is produced in conventional ways, for example from a coding sequence inserted in the host chromosome or on a free plasmid. The yeasts are transformed with a coding sequence for the
70 WSGF desired protein in any of the usual ways, for example i
for transformation of yeast by electroporation are disclosed in Becker &
Guarente (1990) Methods Enzymol. 194, 182.
[00255] Successfully transformed cells, i.e., cells that contain a DNA construct of the present invention, can be identified by well known techniques. For example, cells resulting from the introduction of an expression construct can be grown to produce the desired polypeptide. Cells can be harvested and lysed and their DNA content examined for the presence of the DNA using a method such as that described by Southern (1975) J. Mol. Biol. 98, 503 or Berent et al. (1985) Biotech. 3, 208. Alternatively, the presence of the protein in the supernatant can be detected using antibodies.
[00256] Useful yeast plasmid vectors include pRS403-406 and pRS413-416 and are generally available from Stratagene Cloning Systems, La Jolla, Calif. 92037, USA. Plasmids pRS403, pRS404, pRS405 and pRS406 are Yeast Integrating plasmids (Yips) and incorporate the yeast selectable markers HIS3, 7RP1 , LEU2 and URA3. Plasmids pRS413-416 are Yeast Centromere plasmids (Ycps).
[00257] A variety of methods have been developed to operably link DNA to vectors via complementary cohesive termini. For instance, complementary homopolymer tracts can be added to the DNA segment to be inserted to the vector DNA. The vector and DNA segment are then joined by hydrogen bonding between the complementary honmopolymeric tails to form
recombinant DNA molecules.
[00258] Synthetic linkers containing one or more restriction sites provide an alternative method of joining the DNA segment to vectors. The DNA segment, generated by endonuclease restriction digestion, is treated with bacteriophage
T4 DNA polymerase or E. coli DNA polymerase 1 , enzymes that remove protruding, _single-stranded termini with their 3' 5'-exonucleolytic activities, and fill in recessed 3'-ends with their polymerizing activities.
[00259] The combination of these activities therefore generates blunt-ended
DNA segments. The blunt-ended segments are then incubated with a large
71 WSGF molar excess of linker molecules in the presence of ar
catalyze the ligation of blunt-ended DNA molecules, such as bacteriophage T4 DNA ligase. Thus, the products of the reaction are DNA segments carrying polymeric linker sequences at their ends. These DNA segments are then cleaved with the appropriate restriction enzyme and ligated to an expression vector that has been cleaved with an enzyme that produces termini compatible with those of the DNA segment.
[00260] Synthetic linkers containing a variety of restriction endonuclease sites are commercially available from a number of sources including
International Biotechnologies Inc, New Haven, Conn., USA.
[00261] Exemplary genera of yeast contemplated to be useful in the practice of the present invention as hosts for expressing the albumin, fusion proteins are Pichua (formerly classified as Hansenula), Saccharomyces, Kluyveromyces, Aspergillus, Candida, Torulopsis, Torulaspora,
Schizosaccharomyces, Citeromyces, Pachysolen, Zygosaccharomyces, Debaromyces, Trichoderma, Cephalosporium, Humicola, Mucor, Neurospora, Yarrowia, Metschunikowia, Rhodosporidium, Leucosporidium, Botryoascus, Sporidiobolus, Endomycopsis, and the like. Preferred genera are those selected from the group consisting of Saccharomyces, Schizosaccharomyces, Kluyveromyces, Pichia and Torulaspora. Examples of Saccharomyces spp. are S. cerevisiae, S. italicus and S. rouxii.
[00262] Examples of Kluyveromyces spp. are K. fragilis, K. lactis and K. marxianus. A suitable Torulaspora species is T. delbrueckii. Examples of Pichia (Hansenula) spp. are P. angusta (formerly H. polymorpha), P. anomala
(formerly H. anomala) and P. pastoris. Methods for the transformation of S. cerevisiae are taught generally in EP 251 744, EP 258 067 and WO 90/01063, all of which are incorporated herein by reference.
[00263] Exemplary species of Saccharomyces useful for the synthesis of heteromultimer constructs described herein include S. cerevisiae, S. italicus, S. diastaticus, and Zygosaccharomyces rouxii. Preferred exemplary species of
Kluyveromyces include K. fragilis and K. lactis. Preferred exemplary species of
72 WSGF
Hansenula include H. polymorpha (now Pichia angust
Pichia anomala), and Pichia capsulata. Additional preferred exemplary species of Pichia include P. pastoris. Preferred exemplary species of Aspergillusinclude
A. niger and A. nidulans. Preferred exemplary species of Yarrowia include Y. lipolytica. Many preferred yeast species are available from the ATCC. For example, the following preferred yeast species are available from the ATCC and are useful in the expression of albumin fusion proteins: Saccharomyces cerevisiae, Hansen, teleomorph strain BY4743 yap3 mutant (ATCC Accession
No. 4022731 ); Saccharomyces cerevisiae Hansen, teleomorph strain BY4743 hsp150 mutant (ATCC Accession No. 4021266); Saccharomyces cerevisiae
Hansen, teleomorph strain BY4743 pmtl mutant (ATCC Accession No.
4023792); Saccharomyces cerevisiae Hansen, teleomorph (ATCC Accession
Nos. 20626; 44773; 44774; and 62995); Saccharomyces diastaticus Andrews et Gilliland ex van der Walt, teleomorph (ATCC Accession No. 62987);
Kluyveromyces lactis (Dombrowski) van der Walt, teleomorph (ATCC
Accession No. 76492); Pichia angusta (Teunisson et al.) Kurtzman, teleomorph deposited as Hansenula polymorpha de Morais et Maia, teleomorph (ATCC
Accession No. 26012); Aspergillus niger van Tieghem, anamorph (ATCC
Accession No. 9029); Aspergillus niger van Tieghem, anamorph (ATCC
Accession No. 16404); Aspergillus nidulans (Eidam) Winter, anamorph (ATCC
Accession No. 48756); and Yarrowia lipolytica (Wickerham et al.) van der Walt et von Arx, teleomorph (ATCC Accession No. 201847).
[00264] Suitable promoters for S. cerevisiae include those associated with the PGKI gene, GAL1 or GALI O genes, CYCI, PH05, TRP1 , ADH1 , ADH2, the genes for glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, triose phosphate isomerase,
phosphoglucose isomerase, glucokinase, alpha-mating factor pheromone, [a mating factor pheromone], the PRBI promoter, the GUT2 promoter, the GPDI promoter, and hybrid promoters involving hybrids of parts of 5' regulatory regions with parts of 5' regulatory regions of other promoters or with upstream activation sites (e.g. the promoter of EP-A-258 067).
73 WSGF
[00265] Convenient regulatable promoters for use
pombe are the thiamine-repressible promoter from the nmt gene as described by Maundrell (1990) J. Biol. Chem. 265, 10857-10864 and the glucose repressible jbpl gene promoter as described by Hoffman & Winston (1990) Genetics 124, 807-816.
[00266] Methods of transforming Pichia for expression of foreign genes are taught in, for example, Cregg et al. (1993), and various Phillips patents (e.g. U.S. Pat. No. 4,857,467, incorporated herein by reference), and Pichia expression kits are commercially available from Invitrogen BV, Leek,
Netherlands, and Invitrogen Corp., San Diego, Calif. Suitable promoters include AOX1 and AOX2. Gleeson et al. (1986) J. Gen. Microbiol. 132, 3459-3465 include information on Hansenula vectors and transformation, suitable promoters being MOX1 and FMD1 ; whilst EP 361 991 , Fleer et al. (1991 ) and other publications from Rhone-Poulenc Rorer teach how to express foreign proteins in Kluyveromyces spp., a suitable promoter being PGKI.
[00267] The transcription termination signal is preferably the 3' flanking sequence of a eukaryotic gene which contains proper signals for transcription termination and polyadenylation. Suitable 3' flanking sequences may, for example, be those of the gene naturally linked to the expression control sequence used, i.e. may correspond to the promoter. Alternatively, they may be different in which case the termination signal of the S. cerevisiae ADHI gene is preferred.
[00268] In certain embodiments, the desired heteromultimer protein is initially expressed with a secretion leader sequence, which may be any leader effective in the yeast chosen. Leaders useful in S. cerevisiae include that from the mating factor alpha polypeptide (MFa-1 ) and the hybrid leaders of EP-A- 387 319. Such leaders (or signals) are cleaved by the yeast before the mature albumin is released into the surrounding medium. Further such leaders include those of S. cerevisiae invertase (SUC2) disclosed in JP 62-096086 (granted as 91 1036516), acid phosphatase (PH05), the pre-sequence of MFa-1 , 0
74 WSGF glucanase (BGL2) and killer toxin; S. diastaticus glucc
carlsbergensis a-galactosidase (MEL1 ); K. lactis killer toxin; and Candida glucoarnylase.
[00269] Provided are vectors containing polynucleotides encoding a heteromultimer construct described herein, host cells, and the production of the heteromultimer proteins by synthetic and recombinant techniques. The vector may be, for example, a phage, plasmid, viral, or retroviral vector. Retroviral vectors may be replication competent or replication defective. In the latter case, viral propagation generally will occur only in complementing host cells.
[00270] In certain embodiments, the polynucleotides encoding
heteromultimer proteins described herein are joined to a vector containing a selectable marker for propagation in a host. Generally, a plasmid vector is introduced in a precipitate, such as a calcium phosphate precipitate, or in a complex with a charged lipid. If the vector is a virus, it may be packaged in vitro using an appropriate packaging cell line and then transduced into host cells.
[00271] In certain embodiments, the polynucleotide insert is operatively linked to an appropriate promoter, such as the phage lambda PL promoter, the
E. coli lac, trp, phoA and rac promoters, the SV40 early and late promoters and promoters of retroviral LTRs, to name a few. Other suitable promoters will be known to the skilled artisan. The expression constructs will further contain sites for transcription initiation, termination, and, in the transcribed region, a ribosome binding site for translation. The coding portion of the transcripts expressed by the constructs will preferably include a translation initiating codon at the beginning and a termination codon (UAA, UGA or UAG) appropriately positioned at the end of the polypeptide to be translated.
[00272] As indicated, the expression vectors will preferably include at least one selectable marker. Such markers include dihydrofolate reductase, G418, glutamine synthase, or neomycin resistance for eukaryotic cell culture, and tetracycline, kanamycin or ampicillin resistance genes for culturing in E. coli and other bacteria. Representative examples of appropriate hosts include, but are not limited to, bacterial cells, such as E. coli, Streptomyces and Salmonella
75 WSGF typhimurium cells; fungal cells, such as yeast cells (e.|
cerevisiae or Pichia pastoris (ATCC Accession No. 201 178)); insect cells such as Drosophila S2 and Spodoptera Sf9 cells; animal cells such as CHO, COS, NSO, 293, and Bowes melanoma cells; and plant cells. Appropriate culture mediums and conditions for the above-described host cells are known in the art.
[00273] Among vectors preferred for use in bacteria include pQE70, pQE60 and pQE-9, available from QIAGEN, Inc.; pBluescript vectors, Phagescript vectors, pNH8A, pNH16a, pNH18A; pNH46A, available from Stratagene Cloning Systems, Inc.; and ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 available from Pharmacia Biotech, Inc. Among preferred eukaryotic vectors are pWLNEO, pSV2CAT, pOG44, pXT1 and pSG available from Stratagene; and pSVK3, pBPV, pMSG and pSVL available from Pharmacia. Preferred expression vectors for use in yeast systems include, but are not limited to pYES2, pYD1 , pTEF1/Zeo, pYES2/GS, pPICZ, pGAPZ, pGAPZalph, pPIC9, pPIC3.5, pHIL-D2, pHIL-S1 , pPIC3.5K, pPIC9K, and PAO815 (all available from Invitrogen, Carlbad, CA). Other suitable vectors will be readily apparent to the skilled artisan.
[00274] In one embodiment, polynucleotides encoding a multispecific heteromultimer construct described herein are fused to signal sequences that will direct the localization of a protein of the invention to particular
compartments of a prokaryotic or eukaryotic cell and/or direct the secretion of a protein of the invention from a prokaryotic or eukaryotic cell. For example, in E. coli, one may wish to direct the expression of the protein to the periplasmic space. Examples of signal sequences or proteins (or fragments thereof) to which the heteromultimeric proteins are fused in order to direct the expression of the polypeptide to the periplasmic space of bacteria include, but are not limited to, the pelB signal sequence, the maltose binding protein (MBP) signal sequence, MBP, the ompA signal sequence, the signal sequence of the periplasmic E. coli heat-labile enterotoxin B-subunit, and the signal sequence of alkaline phosphatase. Several vectors are commercially available for the
76 WSGF construction of fusion proteins which will direct the loc
such as the pMAL series of vectors (particularly the pMAL-.rho. series) available from New England Biolabs. In a specific embodiment, polynucleotides albumin fusion proteins of the invention may be fused to the pelB pectate lyase signal sequence to increase the efficiency of expression and purification of such polypeptides in Gram-negative bacteria. See, U.S. Pat. Nos. 5,576,195 and 5,846,818, the contents of which are herein incorporated by reference in their entireties.
[00275] Examples of signal peptides that are fused to a heteromultimeric protein in order to direct its secretion in mammalian cells include, but are not limited to, the MPIF-1 signal sequence (e.g., amino acids 1 -21 of GenBank
Accession number AAB51 134), the stanniocalcin signal sequence
(MLQNSAVLLLLVISASA), and a consensus signal sequence
(MPTWAWWLFLVLLLALWAPARG). A suitable signal sequence that may be used in conjunction with baculoviral expression systems is the gp67 signal sequence (e.g., amino acids 1 -19 of GenBank Accession Number AAA72759).
[00276] Vectors which use glutamine synthase (GS) or DHFR as the selectable markers can be amplified in the presence of the drugs methionine sulphoximine or methotrexate, respectively. An advantage of glutamine synthase based vectors are the availably of cell lines (e.g., the murine myeloma cell line, NSO) which are glutamine synthase negative. Glutamine synthase expression systems can also function in glutamine synthase expressing cells (e.g., Chinese Hamster Ovary (CHO) cells) by providing additional inhibitor to prevent the functioning of the endogenous gene. A glutamine synthase expression system and components thereof are detailed in
PCT publications: WO87/04462; WO86/05807; WO89/10036; WO89/10404; and WO91/06657, which are hereby incorporated in their entireties by reference herein. Additionally, glutamine synthase expression vectors can be obtained from Lonza Biologies, Inc. (Portsmouth, N.H.). Expression and production of monoclonal antibodies using a GS expression system in murine myeloma cells is described in Bebbington et al., Bio/technology 10:169(1992)
77 WSGF and in Biblia and Robinson Biotechnol. Prog. 1 1 :1 (19$
incorporated by reference.
[00277] Also provided are host cells containing vector constructs described herein, and additionally host cells containing nucleotide sequences that are operably associated with one or more heterologous control regions (e.g., promoter and/or enhancer) using techniques known of in the art. The host cell can be a higher eukaryotic cell, such as a mammalian cell (e.g., a human derived cell), or a lower eukaryotic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell. A host strain may be chosen which modulates the expression of the inserted gene sequences, or modifies and processes the gene product in the specific fashion desired. Expression from certain promoters can be elevated in the presence of certain inducers; thus expression of the genetically engineered polypeptide may be controlled. Furthermore, different host cells have characteristics and specific mechanisms for the translational and post-translational processing and modification (e.g., phosphorylation, cleavage) of proteins. Appropriate cell lines can be chosen to ensure the desired modifications and processing of the foreign protein expressed.
[00278] Introduction of the nucleic acids and nucleic acid constructs of the invention into the host cell can be effected by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection, or other methods. Such methods are described in many standard laboratory manuals, such as Davis et al., Basic Methods In Molecular Biology (1986). It is specifically contemplated that the polypeptides of the present invention may in fact be expressed by a host cell lacking a recombinant vector.
[00279] In addition to encompassing host cells containing the vector constructs discussed herein, the invention also encompasses primary, secondary, and immortalized host cells of vertebrate origin, particularly mammalian origin, that have been engineered to delete or replace endogenous genetic material (e.g., the coding sequence corresponding to a Cargo
78 WSGF polypeptide is replaced with a heteromultimer protein i
Cargo polypeptide), and/or to include genetic material. The genetic material operably associated with the endogenous polynucleotide may activate, alter, and/or amplify endogenous polynucleotides.
[00280] In addition, techniques known in the art may be used to operably associate heterologous polynucleotides (e.g., polynucleotides encoding an albumin protein, or a fragment or variant thereof) and/or heterologous control regions (e.g., promoter and/or enhancer) with endogenous polynucleotide sequences encoding a Therapeutic protein via homologous recombination (see, e.g., U.S. Pat. No. 5,641 ,670, issued Jun. 24, 1997; International
Publication Number WO 96/2941 1 ; International Publication Number WO 94/12650; Koller et al., Proc. Natl. Acad. Sci. USA 86:8932-8935 (1989); and Zijlstra et al., Nature 342:435-438 (1989), the disclosures of each of which are incorporated by reference in their entireties).
[00281] Heteromultimer proteins described herein can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography such as with protein A, hydroxylapatite chromatography, hydrophobic charge interaction
chromatography and lectin chromatography. Most preferably, high performance liquid chromatography ("HPLC") is employed for purification.
[00282] In certain embodiments the heteromultimer proteins of the invention are purified using Anion Exchange Chromatography including, but not limited to, chromatography on Q-sepharose, DEAE sepharose, poros HQ, poros DEAF, Toyopearl Q, Toyopearl QAE, Toyopearl DEAE, Resource/Source Q and DEAE, Fractogel Q and DEAE columns.
[00283] In specific embodiments the proteins described herein are purified using Cation Exchange Chromatography including, but not limited to, SP- sepharose, CM sepharose, poros HS, poros CM, Toyopearl SP, Toyopearl CM,
79 WSGF
Resource/Source S and CM, Fractogel S and CM coll
equivalents and comparables.
[00284] In addition, heteromultimer proteins described herein can be chemically synthesized using techniques known in the art (e.g., see Creighton, 1983, Proteins: Structures and Molecular Principles, W. H. Freeman & Co., N.Y and Hunkapiller et al., Nature, 310:105-1 1 1 (1984)). For example, a
polypeptide corresponding to a fragment of a polypeptide can be synthesized by use of a peptide synthesizer. Furthermore, if desired, nonclassical amino acids or chemical amino acid analogs can be introduced as a substitution or addition into the polypeptide sequence. Non-classical amino acids include, but are not limited to, to the D-isomers of the common amino acids,
2,4diaminobutyric acid, alpha-amino isobutyric acid, 4aminobutyric acid, Abu, 2-amino butyric acid, g-Abu, e-Ahx, 6amino hexanoic acid, Aib, 2-amino isobutyric acid, 3-amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, β-alanine, fluoro-amino acids, designer amino acids such as β-methyl amino acids, Ca-methyl amino acids, Na-methyl amino acids, and amino acid analogs in general. Furthermore, the amino acid can be D (dextrorotary) or L (levorotary).
[00285]
[00286] Assays:
[00287] The heteromultimer constructs described herein can be assayed for functional activity (e.g., biological activity) using or routinely modifying assays known in the art, as well as assays described herein.
[00288] For example, in one embodiment where one is assaying for the ability of a heteromultimer construct described herein to bind an antigen or to compete with another polypeptide for binding to an antigen, or bind to an Fc receptor and/or anti-albumin antibody, various immunoassays known in the art can be used, including but not limited to, competitive and non-competitive assay systems using techniques such as radioimmunoassays, ELISA (enzyme
80 WSGF linked immunosorbent assay), "sandwich" immunoass
assays, gel diffusion precipitation reactions, immunodiffusion assays, in situ immunoassays (using colloidal gold, enzyme or radioisotope labels, for example), western blots, precipitation reactions, agglutination assays (e.g., gel agglutination assays, hemagglutination assays), complement fixation assays, immunofluorescence assays, protein A assays, and Immunoelectrophoresis assays, etc. In one embodiment, antibody binding is detected by detecting a label on the primary antibody. In another embodiment, the primary antibody is detected by detecting binding of a secondary antibody or reagent to the primary antibody. In a further embodiment, the secondary antibody is labeled. Many means are known in the art for detecting binding in an immunoassay and are within the scope of the present invention.
[00289] In certain embodiments, where a binding partner (e.g., a receptor or a ligand) is identified for an antigen binding domain comprised by a
heteromultimer described herein, binding to that binding partner by a
heteromultimer described herein is assayed, e.g., by means well-known in the art, such as, for example, reducing and non-reducing gel chromatography, protein affinity chromatography, and affinity blotting. See generally, Phizicky et al., Microbiol. Rev. 59:94-123 (1995). In another embodiment, the ability of physiological correlates of a heteromultimeric protein to bind to a substrate(s) of antigen binding polypeptide constructs of the heteromultimers described herein can be routinely assayed using techniques known in the art.
[00290]
[00291] Therapeutic Uses:
[00292] In an aspect, heteromultimers described herein are directed to antibody-based therapies which involve administering heteromultimers described comprising cargo polypeptide(s) which is an antibody, a fragment or variant of an antibody, to a patient for treating one or more of the disclosed diseases, disorders, or conditions. Therapeutic compounds described herein include, but are not limited to, heteromultimers described herein, nucleic acids encoding heteromultimers described herein.
81 WSGF
[00293] In certain embodiments is provided a metf
treatment or amelioration of at least one of: a proliferative disease, a minimal residual cancer, a tumorous disease, an inflammatory disease, an
immunological disorder, an autoimmune disease, an infectious disease, viral disease, allergic reactions, parasitic reactions, graft-versus-host diseases or host-versus-graft diseases or cell malignancies, said method comprising administering to a subject in need of such a prevention, treatment or
amelioration a pharmaceutical composition comprising a heteromultimer construct described herein.
[00294] In certain embodiments is a method of treating cancer in a mammal in need thereof, comprising administering to the mammal a composition comprising an effective amount of the pharmaceutical composition described herein, optionally in combination with other pharmaceutically active molecules. Inc ertain embodiments, the cancer is a solid tumor. In some embodiments, the solid tumor is one or more of sarcoma, carcinoma, and lymphoma. In certain other embodiments, the cancer is a hematological cancer. In some
embodiments, the cancer is one or more of B-cell lymphoma, non-Hodgkin's lymphoma, and leukemia.
[00295] Provided is a method of treating cancer cells comprising providing to said cell a composition comprising a heteromultimer construct described herein. In some embodiments, the method further comprising providing said heteromultimer in conjugation with another therapeutic agent.
[00296] Provided is a method of treating a cancer non-responsive to blinatumomab in a mammal in need thereof, comprising administering to the mammal a composition comprising an effective amount of the pharmaceutical composition comprising a heteromultimer construct described herein.
[00297] In some embodiments is Aamethod of treating a cancer cell regressive after treatment with blinatumomab, comprising providing to said cancer cell a composition comprising an effective amount of the pharmaceutical composition comprising a heteromultimer construct described herein.
82 WSGF
[00298] In some embodiments is a method of treal
from a disease characterized by expression of B cells, said method comprising providing to said individual an effective amount of a composition comprising an effective amount of the pharmaceutical composition comprising a
heteromultimer construct described herein. In some embodiments the disease is not responsive to treatment with at least one of an anti-CD19 antibody and an anti-CD20 antibody. Inc ertain embodiments the disease is a cancer or autoimmune condition resistant to CD19 or CD20 lytic antibodies
[00299] Provided is a method of treating an autoimmune condition in a mammal in need thereof, comprising administering to said mammal a composition comprising an effective amount of the pharmaceutical composition described herein. In certain embodiments, the autoimmune condition is one or more of multiple sclerosis, rheumatoid arthritis, lupus erytematosus, psoriatic arthritis, psoriasis, vasculitis, uveitis, Crohn's disease, and type 1 diabetes.
[00300] Provided is a method of treating an inflammatory condition in a mammal in need thereof, comprising administering to said mammal a composition comprising an effective amount of the pharmaceutical composition comprising an heteromultimer described herein.
[00301] Armed with the teachings provided herein, one of ordinary skill in the art will know how to use the heteromultimers described herein for diagnostic, monitoring or therapeutic purposes without undue experimentation.
[00302] The heteromultimers described herein, comprising at least a fragment or variant of an antibody may be administered alone or in combination with other types of treatments (e.g., radiation therapy, chemotherapy, hormonal therapy, immunotherapy and anti-tumor agents). Generally, administration of products of a species origin or species reactivity (in the case of antibodies) that is the same species as that of the patient is preferred. Thus, in an embodiment, human antibodies, fragments derivatives, analogs, or nucleic acids, are administered to a human patient for therapy or prophylaxis.
[00303]
[00304] Gene Therapy:
83 WSGF
[00305] In a specific embodiment, nucleic acids cc
encoding heteromultimer proteins described herein are administered to treat, inhibit or prevent a disease or disorder associated with aberrant expression and/or activity of a protein, by way of gene therapy. Gene therapy refers to therapy performed by the administration to a subject of an expressed or expressible nucleic acid. In this embodiment of the invention, the nucleic acids produce their encoded protein that mediates a therapeutic effect. Any of the methods for gene therapy available in the art can be used.
[00306] Though T cell-engaging bispecific single chain antibodies described in the art have great therapeutic potential for the treatment of malignant diseases, most of these bispecific molecules are limited in that they are species specific and recognize only human antigen, and— due to genetic similarity— likely the chimpanzee counterpart. The advantage of the present invention is the provision of a bispecific single chain antibody comprising a binding domain exhibiting cross-species specificity to human and non-chimpanzee primate of the CD3 epsilon chain.
[00307] Demonstration of Therapeutic or Prophylactic Activity:
[00308] The heteromultimers or pharmaceutical compositions described herein are tested in vitro, and then in vivo for the desired therapeutic or prophylactic activity, prior to use in humans. For example, in vitro assays to demonstrate the therapeutic or prophylactic utility of a compound or
pharmaceutical composition include, the effect of a compound on a cell line or a patient tissue sample. The effect of the compound or composition on the cell line and/or tissue sample can be determined utilizing techniques known to those of skill in the art including, but not limited to, rosette formation assays and cell lysis assays. In accordance with the invention, in vitro assays which can be used to determine whether administration of a specific compound is indicated, include in vitro cell culture assays in which a patient tissue sample is grown in
84 WSGF culture, and exposed to or otherwise administered a h
effect of such heteromultimer upon the tissue sample is observed.
[00309]
[00310] Therapeutic/Prophylactic Administration and Composition:
[00311] Provided are methods of treatment, inhibition and prophylaxis by administration to a subject of an effective amount of a heteromultimer or pharmaceutical composition described herein. In an embodiment, the heteromultimer is substantially purified (e.g., substantially free from substances that limit its effect or produce undesired side-effects). In certain embodiments, the subject is an animal, including but not limited to animals such as cows, pigs, horses, chickens, cats, dogs, etc., and in certain embodiments, a mammal, and most preferably human.
[00312] Various delivery systems are known and can be used to administer a heteromultimer formulation described herein, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the compound, receptor-mediated endocytosis (see, e.g., Wu and
Wu, J. Biol. Chem. 262:4429-4432 (1987)), construction of a nucleic acid as part of a retroviral or other vector, etc. Methods of introduction include but are not limited to intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes. The compounds or compositions may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local. In addition, in certain embodiments, it is desirable to introduce the heteromultimer compositions described herein into the central nervous system by any suitable route, including intraventricular and intrathecal injection; intraventricular injection may be facilitated by an intraventricular catheter, for example, attached to a reservoir, such as an Ommaya reservoir.
Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent.
85 WSGF
[00313] In a specific embodiment, it is desirable to
heteromultimers, or compositions described herein locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion during surgery, topical application, e.g., in conjunction with a wound dressing after surgery, by injection, by means of a catheter, by means of a suppository, or by means of an implant, said implant being of a porous, non- porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers. Preferably, when administering a protein, including an antibody, of the invention, care must be taken to use materials to which the protein does not absorb.
[00314] In another embodiment, the heteromultimers or composition can be delivered in a vesicle, in particular a liposome (see Langer, Science 249:1527- 1533 (1990); Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353-365 (1989); Lopez-Berestein, ibid., pp. 317-327; see generally ibid.)
[00315] In yet another embodiment, the heteromultimers or composition can be delivered in a controlled release system. In one embodiment, a pump may be used (see Langer, supra; Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al., Surgery 88:507 (1980); Saudek et al., N. Engl. J. Med. 321 :574 (1989)). In another embodiment, polymeric materials can be used (see Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla. (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger and Peppas, J., Macromol. Sci. Rev. Macromol. Chem. 23:61 (1983); see also Levy et al., Science 228:190 (1985); During et al., Ann. Neurol. 25:351 (1989); Howard et al., J. Neurosurg. 71 :105 (1989)). In yet another
embodiment, a controlled release system can be placed in proximity of the therapeutic target, e.g., the brain, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 1 15-138 (1984)).
86 WSGF
[00316] Other controlled release systems are disci
Langer (Science 249: 1527-1533 (1990)).
[00317] In a specific embodiment comprising a nucleic acid encoding a heteromultimer decribed herein, the nucleic acid can be administered in vivo to promote expression of its encoded protein, by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by use of a retroviral vector (see U.S. Pat. No. 4,980,286), or by direct injection, or by use of microparticle bombardment (e.g., a gene gun; Biolistic, Dupont), or coating with lipids or cell-surface receptors or transfecting agents, or by administering it in linkage to a homeobox-like peptide which is known to enter the nucleus (see e.g., Joliot et al., Proc. Natl. Acad. Sci. USA 88:1864-1868 (1991 )), etc. Alternatively, a nucleic acid can be introduced intracellular^ and incorporated within host cell DNA for expression, by homologous recombination.
[00318] Also provided herein are pharmaceutical compositions. Such compositions comprise a therapeutically effective amount of a compound, and a pharmaceutically acceptable carrier. In a specific embodiment, the term
"pharmaceutically acceptable" means approved by a regulatory agency of the
Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans. The term "carrier" refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered. Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. The composition, if
87 WSGF desired, can also contain minor amounts of wetting or
pH buffering agents. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like. The composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides. Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E. W. Martin. Such compositions will contain a therapeutically effective amount of the compound, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient. The formulation should suit the mode of administration.
[00319] In certain embodiments, the composition comprising the
heteromultimer is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings. Typically, compositions for intravenous administration are solutions in sterile isotonic aqueous buffer. Where necessary, the composition may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection. Generally, the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
[00320] In certain embodiments, the compositions described herein are formulated as neutral or salt forms. Pharmaceutically acceptable salts include those formed with anions such as those derived from hydrochloric, phosphoric,
88 WSGF acetic, oxalic, tartaric acids, etc., and those formed wii
derived from sodium, potassium, ammonium, calcium, ferric hydroxide isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.
[00321] The amount of the composition described herein which will be effective in the treatment, inhibition and prevention of a disease or disorder associated with aberrant expression and/or activity of a Therapeutic protein can be determined by standard clinical techniques. In addition, in vitro assays may optionally be employed to help identify optimal dosage ranges. The precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each patient's circumstances. Effective doses are extrapolated from dose-response curves derived from in vitro or animal model test systems.
[00322] In certain embodiments, a heteromultimer construct described herein is suitably administered to the patient at one time or over a series of treatments. Depending on the type and severity of the disease, about 1 μg/kg to 15 mg/kg (e.g. 0.1 mg/kg - 10 mg/kg) of T cell activating bispecific antigen binding molecule can be an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion. One typical daily dosage might range from about 1 μg/kg to 100 mg/kg or more, depending on the factors mentioned above. For repeated administrations over several days or longer, depending on the condition, the treatment would generally be sustained until a desired
suppression of disease symptoms occurs. One exemplary dosage of the heteromultimer described herein would be in the range from about 0.005 mg/kg to about 10 mg/kg. In other non-limiting examples, a dose may also comprise from about 1 microgram/kg body weight, about 5 microgram/kg body weight, about 10 microgram/kg body weight, about 50 microgram/kg body weight, about 100 microgram/kg body weight, about 200 microgram/kg body weight, about 350 microgram/kg body weight, about 500 microgram/kg body weight, about 1 milligram/kg body weight, about 5 milligram/kg body weight, about 10
89 WSGF milligram/kg body weight, about 50 milligram/kg body
milligram/kg body weight, about 200 milligram/kg body weight, about 350 milligram/kg body weight, about 500 milligram/kg body weight, to about 1000 mg/kg body weight or more per administration, and any range derivable therein. In non-limiting examples of a derivable range from the numbers listed herein, a range of about 5 mg/kg body weight to about 100 mg/kg body weight, about 5 microgram kg body weight to about 500 milligram kg body weight, etc., can be administered, based on the numbers described above. Thus, one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 5.0 mg/kg or 10 mg/kg (or any
combination thereof) may be administered to the patient. Such doses may be administered intermittently, e.g. every week or every three weeks (e.g. such that the patient receives from about two to about twenty, or e.g. about six doses of the T cell activating bispecific antigen binding molecule). An initial higher loading dose, followed by one or more lower doses may be administered.
However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
[00323] The heteromultimers described herein are generally used in an amount effective to achieve the intended purpose. For use to treat or prevent a disease condition, a heteromultimer described herein, or pharmaceutical compositions thereof, are administered or applied in a therapeutically effective amount. Determination of a therapeutically effective amount is well within the capabilities of those skilled in the art, especially in light of the detailed disclosure provided herein.
[00324] For systemic administration, a therapeutically effective dose can be estimated initially from in vitro assays, such as cell culture assays. A dose can then be formulated in animal models to achieve a circulating concentration range that includes the IC50 as determined in cell culture. Such information can be used to more accurately determine useful doses in humans.
[00325] Initial dosages can also be estimated from in vivo data, e.g., animal models, using techniques that are well known in the art. One having ordinary
90 WSGF skill in the art could readily optimize administration to I
data.
[00326] Dosage amount and interval may be adjusted individually to provide plasma levels of the heteromultimer described herein which are sufficient to maintain therapeutic effect. Usual patient dosages for administration by injection range from about 0.1 to 50 mg/kg/day, typically from about 0.5 to 1 mg/kg/day. Therapeutically effective plasma levels may be achieved by administering multiple doses each day. Levels in plasma may be measured, for example, by HPLC.
[00327] In cases of local administration or selective uptake, the effective local concentration of the heteromultimer described herein may not be related to plasma concentration. One having skill in the art will be able to optimize therapeutically effective local dosages without undue experimentation.
[00328] A therapeutically effective dose of the heteromultimer constructs described herein will generally provide therapeutic benefit without causing substantial toxicity. Toxicity and therapeutic efficacy of a heteromultimer described herein can be determined by standard pharmaceutical procedures in cell culture or experimental animals. Cell culture assays and animal studies can be used to determine the LD50 (the dose lethal to 50% of a population) and the
ED50 (the dose therapeutically effective in 50% of a population). The dose ratio between toxic and therapeutic effects is the therapeutic index, which can be expressed as the ratio LD50/ED50. T cell activating bispecific antigen binding molecules that exhibit large therapeutic indices are preferred. In one
embodiment, the heteromultimer construct described herein according to the present invention exhibits a high therapeutic index. The data obtained from cell culture assays and animal studies can be used in formulating a range of dosages suitable for use in humans. The dosage lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon a variety of factors, e.g., the dosage form employed, the route of administration utilized, the condition of the subject, and the like. The exact formulation, route of administration and dosage
91 WSGF can be chosen by the individual physician in view of tr
(see, e.g., Fingl et al, 1975, in: The Pharmacological Basis of Therapeutics, Ch. 1 , p. 1 , incorporated herein by reference in its entirety).
[00329] The attending physician for patients treated with heteromultimer constructs described herein would know how and when to terminate, interrupt, or adjust administration due to toxicity, organ dysfunction, and the like.
Conversely, the attending physician would also know to adjust treatment to higher levels if the clinical response were not adequate (precluding toxicity). The magnitude of an administered dose in the management of the disorder of interest will vary with the severity of the condition to be treated, with the route of administration, and the like. The severity of the condition may, for example, be evaluated, in part, by standard prognostic evaluation methods. Further, the dose and perhaps dose frequency will also vary according to the age, body weight, and response of the individual patient.
[00330] Also provided is a process for the production of a pharmaceutical composition comprising a eteromutlimer described herein, said process comprising: culturing a host cell under conditions allowing the expression of a heteromultimer; recovering the produced heteromultimer from the culture; and producing the pharmaceutical composition.
[00331]
[00332] Other Agents and Treatments:
[00333] In certain embodiments, the heteromultimer constructs described herein are administered in combination with one or more other agents in therapy. For instance, in one embodiment, a heteromultimer described herein is co-administered with at least one additional therapeutic agent. The term
"therapeutic agent" encompasses any agent administered to treat a symptom or disease in an individual in need of such treatment. Such additional therapeutic agent may comprise any active ingredients suitable for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other. In certain embodiments, an additional therapeutic agent is an immunomodulatory agent, a cytostatic agent,
92 WSGF an inhibitor of cell adhesion, a cytotoxic agent, an acti'
an agent that increases the sensitivity of cells to apoptotic inducers. In a particular embodiment, the additional therapeutic agent is an anti-cancer agent, for example a microtubule disruptor, an antimetabolite, a topoisomerase inhibitor, a DNA intercalator, an alkylating agent, a hormonal therapy, a kinase inhibitor, a receptor antagonist, an activator of tumor cell apoptosis, or an antiangio genie agent.
[00334] Such other agents are suitably present in combination in amounts that are effective for the purpose intended. The effective amount of such other agents depends on the amount of T cell activating bispecific antigen binding molecule used, the type of disorder or treatment, and other factors discussed above. The heteromultimers described herein are generally used in the same dosages and with administration routes as described herein, or about from 1 to 99% of the dosages described herein, or in any dosage and by any route that is empirically/clinically determined to be appropriate.
[00335] Such combination therapies noted above encompass combined administration (where two or more therapeutic agents are included in the same or separate compositions), and separate administration, in which case, administration of the heteromultimer described herein can occur prior to, simultaneously, and/or following, administration of the additional therapeutic agent and/or adjuvant. Heteromultimer constructs described herein can also be used in combination with radiation therapy.
[00336]
[00337] Articles of Manufacture:
[00338] In another aspect of the invention, an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of the disorders described above is provided. The article of manufacture comprises a container and a label or package insert on or associated with the container.
Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc. The containers may be formed from a variety of materials such as glass or plastic. The container holds a composition which is by itself or
93 WSGF combined with another composition effective for treatii
diagnosing the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). At least one active agent in the composition is a T cell activating bispecific antigen binding molecule of the invention. The label or package insert indicates that the composition is used for treating the condition of choice. Moreover, the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises a heteromultimer described herein; and (b) a second container with a composition contained therein, wherein the composition comprises a further cytotoxic or otherwise therapeutic agent. The article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the compositions can be used to treat a particular condition. Alternatively, or additionally, the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically- acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate- buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
[00339]
[00340] EXAMPLES
[00341] The following specific and non-limiting examples are to be construed as merely illustrative, and do not limit the present disclosure in any way whatsoever. Without further elaboration, it is believed that one skilled in the art can, based on the description herein, utilize the present disclosure to its fullest extent. All publications cited herein are hereby incorporated by reference in their entirety. Where reference is made to a URL or other such identifier or address, it is understood that such identifiers can change and particular information on the internet can come and go, but equivalent information can be found by searching the internet. Reference thereto evidences the availability and public dissemination of such information.
94 WSGF
[00342]
[00343] Example 1 : Bispecific CD3-CD19 scFvs fused to an asymmetric lgG1 Fc.
[00344] Bispecific CD3-CD19 scFvs fused to an asymmetric lgG1 Fc heterodimer that exhibits stability comparable with native Fc homodimer, is a novel composition identified as v873. V873 belongs to a novel family of CD3- based bispecific azymetric lgG1 antibodies that can be expressed and purified with significantly higher yields in mammalian CHO cells compared to
Amgen/Micromet bscCD19xCD3 BiTE bispecific. V873 demonstrates unexpected effectontarget cell binding, bridging and target cell killing.
[00345] V873 and bispecific CD3-based azymetric antibodies have utility in targeted T cell mediated killing of diseased cells and hence may be useful for treating cancers and autoimmune and inflammatory diseases. V873 is a bispecific CD3-CD19 scFvs fused to an azymetric lgG1 Fc. v873 represents a novel bispecific azymetric antibody class comprising one anti-CD3 warhead and a second warhead comprising a cell surface antigen of a target cell, and an antibody Fc heterodimer comprising heterodimer lgG1 Fc. The fusion of the CD3 warhead to chain A or B of the Fc is important for its druggable properties. As additional examples, V874 and V875 are two other Bispecific CD3-CD19 scFvs fused to an asymmetric lgG1 Fc but have different CD3 amino acid compositions.
[00346] V873 shows unexpected good mammalian CHO expression and purification yields compared to the Amgen/Micromet blinatumomab CD3-CD19 BiTE tandem scFvs. V873 bridges T and B cells and results in potent killing of cultured human Burkitt lymphoma cells (Raji B-cell lymphoma line) using resting and IL-2 activated human PBMCs.
[00347] V873 and its related bispecific CD3-Azymetric antibodies differ from
Amgen/Micromet CD3-CD19 BiTE and possess an lgG1 heterodimeric Fc that can bind FcgRs to mediate ADCC, CDC, ADCP effector activities. V873 can bind FcRn and this heterodimeric Fc class shows typical antibody
manufacturability characteristics and long-half life of greater than 8 days in
95 WSGF cynomolgus monkey. In contrast, blinatumomab has ε
several hours in non-human monkey and in human patients.
[00348] V873 and related bispecific CD3-based constructs address known stability issues with tandem scFvs and generally recognized poor
manufacturability of blinatumomab. V873 addresses the short PK of blinatumomab and CNS adverse effects owing to its wildtype FcRn binding affinity and MWT which restricts its distribution to the peripheral compartment. Lastly, the Azymetric heterodimer Fc confers additional tailored FcgR effector ADCC, CDC, and ADCP activities and hence efficacy to drug resistant tumors.
[00349] The ability of v873 to mediate PBMC (T cell)- B cell killing is highly unexpected. The properties of blinatumomab bscCD19xCD3 and related BiTEs is reported to rely on the use of flexibly linked single-chain variable fragments (scFv) that are arranged in tandem joined by a short linker will allow for a much closer approximation of opposing cells than is possible with larger bispecific formats such as quadroma antibodies. The flexible linkage is expected to enable free rotation and kinking of the 2 scFv arms, thereby facilitating the simultaneous recognition of 2 epitopes present on 2 opposing cell membranes and the formation of a cytolytic immunologic synapse. Hence, based on what has been reported and generally accepted as unique to BiTE, it is unexpected that a bispecific CD3-CD19 construct as described herein with significantly different structural presentations of CD3 and CD19 scFv warheads on the heterodimer Fc can bind, bridge T and B cells, and mediate PBMC (T cell)-B cell killing. Other applications include depletion of B cells in B cell driven autoimmune and inflammatory diseases such as RA, lupus, MS, IBD. Further, V873 related bispecific CD3-Based azymetric antibodies may be useful for diagnostic purposes.
[00350] Additional details regarding the above are found in the following examples.
[00351]
[00352] Example 2. Design, expression and purification of heteromultimer constructs with a heterodimeric Fc.
96 WSGF
[00353] Exemplary bispecific anti-CD3 and anti-C[
antibodies
[00354]
[00355] An exemplary schematic representation of an anti-CD3/anti-CD19 antibody is shown in Figure 1A.
[00356] v873, v874, v875 exemplify bispecific anti-CD3/ anti-CD19 heterodimeric Fc constructs and were prepared and tested as described below. Where the description includes a reference to BiTE, it refers to the antibody construct having an identical amino acid sequence to either the VH or VL of the anti-CD3 anti-CD19 BiTE molecule with or without modifications to variable heavy and light chain orientation (e.g. VH-VL) as indicated below.
[00357] v873 has a anti-CD19 BiTE (VL-VH) scFv on chain A and a CD3 BiTE™ (VH-VL) scFv on chain B of the heterodimer Fc with the following mutations L351Y_F405A_Y407V on chain A and T366L_K392M_T394W on chain B. [Polypeptide sequences correspond to SEQ ID No: 26 and 28]
[00358] V874 has a anti-CD19 BiTE™ (VL-VH) scFv on chain A and the CD3 BiTE™ (VLVH) scFv on chain B of the heterodimer Fc with the following mutations L351Y_F405A_Y407V on chain A and T366L_K392M_T394W on chain B. [Polypeptide sequences correspond to SEQ ID No: 30 and 32]
[00359] V875 has a anti-CD19 BiTE™ (VL-VH) scFv on chain A and the CD3 OKT3 (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations L351Y_F405A_Y407V on chain A and T366L_K392M_T394W on chain B. [Polypeptide sequences correspond to SEQ ID No: 34 and 36]
[00360] v1379 has a anti-CD19 BiTE™ (VL-VH) scFv on chain A and a anti- CD3 (VH-VL) BiTE on chain B of the heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and
T350V_T366L_K392L_T394W on chain B. [Polypeptide sequences
correspond to SEQ ID No: 87 and 88]
[00361] Bivalent monospecific anti-CD3 and anti-CD19 antibodies
[00362] v865, v866, v867, v868 exemplify monospecific anti-CD3 or CD19 bivalent scFv-Fc constructs and were prepared and tested as described below:
97 WSGF
[00363] v865 has a anti-CD19 BiTE (VL-VH) scFv
CD19 BiTE scFv chain B with WT Fc (105kDa). [Polypeptide sequence correspond to SEQ ID No: 2]
[00364] v866 has a anti-CD3 BiTE™ (VH-VL) on chain A and a anti-CD3 BiTE™ (VH-VL) scFv on chain with a WT Fc (105kDa). [Polypeptide sequence correspond to SEQ ID No: 4]
[00365] v867 has a anti-CD3 BiTE™ (VL-VH) scFv on chain A and a anti- CD3 BiTE™ (VL-VH) scFv on chain B with a WT Fc (105kDa). [Polypeptide sequence correspond to SEQ ID No: 6]
[00366] v868 has a anti-CD3 OKT3 (VL-VH) scFv on chain A and a anti- CD3 OKT3 VL-VH scFv on chain with a WT Fc (105 kDa). [Polypeptide sequence correspond to SEQ ID No: 8].
[00367] Monovalent monospecific anti-CD3
[00368] v869 has a anti-CD19 BiTE™ scFv (VL-VH) on chain A and a Fc on chain B of the heterodimer Fc with the following mutations
L351Y_F405A_Y407V on chain A and T366L_K392M_T394W on chain B(80 kDa). [Polypeptide sequences correspond to SEQ ID No: 10 and 12].
[00369] v870 has a anti- CD3 BiTE™ (VH-VL) scFv on chain B and a Fc on chain A of the heterodimer Fc with the following mutations
L351Y_F405A_Y407V on chain A and T366L_K392M_T394W on chain B (80 kDa). [Polypeptide sequences correspond to SEQ ID No: 14 and 16].
[00370] v871 has a anti-CD3 BiTE™ (VL-VH) scFv on chain B and a Fc on chain A of the heterodimer Fc with the following mutations
L351Y_F405A_Y407V on chain A and T366L_K392M_T394W on chain B (80 kDa). [Polypeptide sequences correspond to SEQ ID No: 18 and 20].
[00371] v872 has a anti-CD3 OKT3 (VL-VH) scFv on chain B and a Fc on chain A of the heterodimer Fc with the following mutations
L351Y_F405A_Y407V on chain A and T366L_K392M_T394W on chain B (80 kDa). [Polypeptide sequences correspond to SEQ ID No: 22 and 24].
[00372] Benchmark control
98 WSGF
[00373] v891 has the identical sequence blinatumc
BiTE scFv and anti-CD19 BiTE scFv (50 kDa) [Polypeptide sequence corresponds to SEQ ID No: 90
[00374] Exemplary mono- or bispecific anti-CD3 and anti-CD19
heterodimeric antibodies with a knock-out Fc
[00375] v1380 has a anti-CD19 BiTE™ (VL-VH) scFv on chain A and a anti-CD3 VHVL BiTE™ on chain B of the heterodimer Fc with the following mutations L234A_L235A_T350V_L351Y_F405A_Y407V on chain A and L234A_L235A_T350V_T366L_K392L_T394W on chain B (corresponding to polypeptide sequences SEQ ID NOs:93 and 94).
[00376] v1381 has a anti-CD19 BiTE™ (VI-VH) scFv on chain A and a anti-
CD3 BiTE™ scFv (VH-VL) on chain B of the heterodimer Fc with the following mutations N297A_T350V_L351Y_F405A_Y407V on chain A and
N297A_T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 93 and 98)
[00377] Variants with engineered anti-CD3 warheads for stability
enhancement
[00378] The following variants contain mutations to the anti-CD3 scFv that include either changes to the linker length, VH-VL orientations, or point mutations to improve stability and yeild.
[00379] v1653 has a anti-CD19 BiTE™ (VL-VH) scFv on chain A and a anti- CD3 OKT3 (VL-VH) (with C to S mutation at position 100A of the VH CDR3) on chain B of the heterodimer Fc with the following mutations
T350V_L351Y_F405A_Y407V on chain A and T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 101 and 102)
[00380] v1654 has a anti-CD19 BiTE™ scFv on chain A and a anti-CD3
OKT3 OKT3 (VH-VL) with 18 amino acid linker chain B of the heterodimer Fc with the following mutations T350V_L351 Y_F405A_Y407V on chain A and
T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 105 and 106)
99 WSGF
[00381] v1655 has a anti-CD19 BiTE ' scFv on cr
OKT3 OKT3 (VH-VL) with 10 amino acid linker chain B of the heterodimer Fc with the following mutations T350V_L351 Y_F405A_Y407V on chain A and T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 109 and 1 10)
[00382] v1656 has a anti-CD3 BiTE™ VHVL scFv on chain A and a anti- CD19 BiTE™ (VL-VH) on chain B of the heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and
T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 113 and 1 14)
[00383] v1657 has a anti-CD3 OKT3 (VL-VH) scFv (with C to S mutation at position 100A of the VH CDR3) on chain A and a anti-CD19 BiTE™ (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations
T350V_L351Y_F405A_Y407V on chain A and T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 1 17 and 1 18)
[00384] v1658 has a anti-CD3 OKT3 (VH-VL) scFv with 18 amino acid linker on chain A and a anti-CD19 BiTE™ (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 121 and 122)
[00385] v1659 has a anti-CD3 OKT3 (VH-VL) scFv with 10 amino acid linker on chain A and a anti-CD19 BiTE™ (VL_VH) scFv on chain B of the heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 125 and 126)
[00386] v1660 has a anti-CD3 OKT3 (VH-VL) scFv with 19 amino acid linker on chain A and a anti-CD19 BiTE™ (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 129 and 130)
100 WSGF
[00387] Fc knock-out variants with engineered ant
stability enhancement
[00388] The following variants are Fc knoock outs that contain mutations to the anti-CD3 scFv that include either changes to the linker length, VH-VL orientations, or point mutations to improve stability and yeild.
[00389] v1661 has a anti-CD19 BiTE™ (VL-VH) scFv on chain A and a anti- CD3 OKT3 (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations D265S_L234A_L235A_T350V_L351Y_F405A_Y407V on chain A and D265S_L234A_L235A_T350V_T366L_K392L_T394W on chain B.
[Polypeptide sequences correspond to SEQ ID No: 38 and 40]
[00390] v1662 has a anti-CD19 BiTE™ (VL-VH) scFv on chain A and a anti- CD3 OKT3 (VL-VH) scFv (with C to S mutation at position 100A of the VH CDR3) on chain B of the heterodimer Fc with the following mutations
D265S_L234A_L235A_T350V_L351Y_F405A_Y407V on chain A and
D265S_L234A_L235A_T350V_T366L_K392L_T394W on chain B. [Polypeptide sequences correspond to SEQ ID No: 42 and 44]
[00391] v1663 has a anti-CD19 BiTE™ (VL-VH) scFv on chain A and a anti- CD3 OKT3 (VL-VH) scFv with 18 amino acid linker on chain B of the
heterodimer Fc with the following mutations
D265S_L234A_L235A_T350V_L351Y_F405A_Y407V on chain A and
D265S_L234A_L235A_T350V_T366L_K392L_T394W on chain B.
(corresponding to polypeptide sequences SEQ ID NOs: 133 and 134)
[00392] v1664 has a anti-CD19 BiTE™ (VL-VH) scFv on chain A and a anti- CD3 OKT3 (VH-VL) scFv with 10 amino acid linker on chain B of the
heterodimer Fc with the following mutations
D265S_L234A_L235A_T350V_L351Y_F405A_Y407V on chain A and
D265S_L234A_L235A_T350V_T366L_K392L_T394W on chain B.
(corresponding to polypeptide sequences SEQ ID NOs: 137 and 138)
[00393] v1665 has a anti-CD3 BiTE™ (VH-VL) scFv on chain A and a anti-
CD19 BiTE™ (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations D265S_L234A_L235A_T350V_L351Y_F405A_Y407V on chain A
101 WSGF and D265S_L234A_L235A_T350V_T366L_K392L_T
(corresponding to polypeptide sequences SEQ ID NOs: 141 and 142)
[00394] v1666 has a anti-CD3 OKT3 (VH-VL) scFv with a 19 amino acid linker on chain A and a anti-CD19 BiTE™ (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations
D265S_L234A_L235A_T350V_L351Y_F405A_Y407V on chain A and
D265S_L234A_L235A_T350V_T366L_K392L_T394W on chain B [Polypeptide sequences correspond to SEQ ID No: 46 and 48].
[00395] v1667 has a anti-CD19 BiTE™ (VL-VH) scFv on chain A and a anti- CD3 OKT3 (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations N297A_T350V_L351Y_F405A_Y407V on chain A and
N297A_T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 145 and 146)
[00396] v1668 has a anti-CD19 BiTE™ (VL-VH) scFv on chain A and a anti- CD3 OKT3 (VL-VH) scFv (with C to S mutation at position 100A of the VH CDR3) on chain B of the heterodimer Fc with the following mutations
N297A_T350V_L351Y_F405A_Y407V on chain A and
N297A_T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 149 and 150)
[00397] v1669 has a anti-CD19 BiTE™ (VL-VH) scFv on chain A and a anti- CD3 OKT3 (VH-VL) scFv with 18 amino acid linker on chain B of the
heterodimer Fc with the following mutations
N297A_T350V_L351Y_F405A_Y407V on chain A and
N297A_T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 153 and 154)
[00398] v1670 has a anti-CD19 BiTE™ (VL-VH) scFv on chain A and a anti-
CD3 OKT3 (VH-VL) scFv with 10 amino acid linker on chain B of the
heterodimer Fc with the following mutations
N297A_T350V_L351Y_F405A_Y407V on chain A and
N297A_T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 157 and 158)
102 WSGF
[00399] v1671 has a anti-CD3 BiTE I M (VH-VL) sc
CD19 BiTE™ (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations N297A_T350V_L351Y_F405A_Y407V on chain A and N297A_T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 161 and 162)
[00400] v1672 has a anti-CD3 OKT3 (VH-VL) scFv with a 19 amino acid linker on chain A and a anti-CD19 BiTE™ (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations
N297A_T350V_L351Y_F405A_Y407V on chain A and
N297A_T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 165 and 166)
[00401] Monovalent anti-CD3 antibodies
[00402] The following variants contain mutations to the anti-CD3 scFv that include either changes to the linker length, VH-VL orientations, or point mutations to improve stability and yeild.
[00403] v1673 has a anti-CD3 OKT3 (VL-VH) scFv (with C to S mutation at position 100A of the VH CDR3) on chain B and a Fc on chain A of the heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 169 and 170)
[00404] v1674 has a anti-CD3 OKT3 (VH-VL) scFv with 18 amino acid linker on chain B and a Fc on chain A of the heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and
T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 173 and 174)
[00405] v1798 has a anti-CD3 OKT3 (VH-VL) scFv with 10 amino acid linker on chain B and a Fc on chain A of the heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and
T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 177 and 178)
103 WSGF
[00406] v1799 has a anti-CD3 OKT3 (VH-VL) scF
linker on chain A and a Fc on chain B of the heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and
T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 181 and 182)
[00407] Bispecific disulfide 44-100 stabilization variants
[00408] The following variants contain point mutations for disulfide stabilization at position 100 in the variable light and position 44 in the variable heavy chain (denoted 44-100SS).
[00409] v1800 has a anti-CD19 BiTE™ (VL-VH) scFv on chain A and anti- CD3 OKT3 (VL-VH) 44-100SS on chain B of the heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and
T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 185 and 186)
[00410] v1801 has a anti-CD19 BiTE™ (VL-VH) scFv on chain A and anti- CD3 OKT3 (VL-VH) (with C to S mutation at position 100A of the VH CDR3) 44-100SS on chain B of the heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 189 and 190)
[00411] v1802 has a anti-CD3 BiTE™ (VH-VL) 44-100SS scFv on chain A and anti-CD19 BiTE™ (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and
T350V_T366L_K392L_T394W on chain B. (corresponding to polypeptide sequences SEQ ID NOs: 193 and 194)
[00412] v4541 has a anti-CD3 BiTE™ (VH-VL) 44-100SS scFv on chain A and anti-CD19 BiTE™ (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations D265S_L234A_L235A_T350V_L351Y_F405A_Y407V on chain A and D265S_L234A_L235A_T350V_T366L_K392L_T394W on chain B. [Polypeptide sequences correspond to SEQ ID No: 50 and 52]
[00413]
104 WSGF
[00414] Cyno/human cross-reactive anti-CD3 and
knock-out variants with or without disulfide 44-100 stabilization
[00415] v4542 has a cyno/human cross-reactive anti-CD3 BiTE™ 12C (VH- VL) scFv on chain A and cyno/human cross-reactive anti-CD19 MOR208 (VH- VL) scFv on chain B of the heterodimer Fc with the following mutations
D265S_L234A_L235A_T350V_L351Y_F405A_Y407V on chain A and
D265S_L234A_L235A_T350V_T366L_K392L_T394W on chain B. [Polypeptide sequences correspond to SEQ ID No: 54 and 56].
[00416] v4543 has a cyno/human cross-reactive anti-CD3 BiTE™ 12C (VH- VL) 44-100SS scFv on chain A and cyno/human cross -reactive anti-CD19 MOR208 (VH-VL) 44-100SS scFv on chain B of the heterodimer Fc with the following mutations D265S_L234A_L235A_T350V_L351Y_F405A_Y407V on chain A and D265S_L234A_L235A_T350V_T366L_K392L_T394W on chain B. [Polypeptide sequences correspond to SEQ ID No: 58 and 60]
[00417] v4544 has a cyno/human cross-reactive anti-CD3 BiTE™ 12C (VH-VL) scFv on chain A and cyno/human cross-reactive anti-CD19 MOR208 (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations D265S_L234A_L235A_T350V_L351Y_F405A_Y407V on chain A and
D265S_L234A_L235A_T350V_T366L_K392L_T394W on chain B. [Polypeptide sequences correspond to SEQ ID No: 62 and 64]
[00418] v4545 has a cyno/human cross-reactive anti-CD3 BiTE™ 12C (VH- VL) 44-100SS scFv on chain A and cyno/human cross -reactive anti-CD19 MOR208 (VL-VH) 44-100SS scFv on chain B of the heterodimer Fc with the following mutations D265S_L234A_L235A_T350V_L351Y_F405A_Y407V on chain A and D265S_L234A_L235A_T350V_T366L_K392L_T394W on chain B. [Polypeptide sequences correspond to SEQ ID No: 66 and 68]
[00419] v4546 has a cyno/human cross-reactive anti-CD3 BiTE™ 12C (VH- VL) scFv on chain A and cyno/human cross-reactive anti-CD19 MDX-1342 (VH-VL) scFv on chain B of the heterodimer Fc with the following mutations D265S L234A L235A T350V L351Y F405A Y407V on chain A and
105 WSGF
D265S_L234A_L235A_T350V T366L_K392L_T394V^
sequences correspond to SEQ ID No: 70 and 72]
[00420] v4547 has a cyno/human cross-reactive anti-CD3 BiTE™ 12C (VH- VL) 44-100SS scFv on chain A and cyno/human cross-reactive anti-CD19 MDX-1342 (VH-VL) 44-100SS scFv on chain B of the heterodimer Fc with the following mutations D265S_L234A_L235A_T350V_L351Y_F405A_Y407V on chain A and D265S_L234A_L235A_T350V_T366L_K392L_T394W on chain B. [Polypeptide sequences correspond to SEQ ID No: 74 and 76].
[00421] v4548 has a cyno/human cross-reactive anti-CD3 BiTE™ 12C (VH- VL) scFv on chain A and cyno/human cross-reactive anti-CD19 MDX-1342 (VL-VH) scFv on chain B of the heterodimer Fc with the following mutations D265S_L234A_L235A_T350V_L351Y_F405A_Y407V on chain A and
D265S_L234A_L235A_T350V_T366L_K392L_T394W on chain B. [Polypeptide sequences correspond to SEQ ID No: 78 and 80]
[00422] v4549 has a cyno/human cross-reactive anti-CD3 BiTE™ 12C (VH- VL) 44-100SS scFv on chain A and cyno/human cross -reactive anti-CD19 MDX-1342 (VL-VH) 44-100SS scFv on chain B of the heterodimer Fc with the following mutations D265S_L234A_L235A_T350V_L351Y_F405A_Y407V on chain A and D265S_L234A_L235A_T350V_T366L_K392L_T394W on chain B. [Polypeptide sequences correspond to SEQ ID No: 82 and 84]
[00423] The antibodies and antibody controls were cloned and expressed as follows. The genes encoding the antibody heavy and light chains were constructed via gene synthesis using codons optimized for human/mammalian expression. The Fab sequences were generated from a known Her2/neu binding Ab (Carter P. et al. (1992) Humanization of an anti P185 Her2 antibody for human cancer therapy. Proc Natl Acad Sci 89, 4285.) and the Fc was an lgG1 isotype. The scFv-Fc and OAA sequences, were generated from a known anti-CD3 and CD19 scFv Bite antibodies (Kipriyanov et. al., 1998, Int. J Cancer: 77,763-772), anti-CD3 BiTE antibodies (US201 1/0275787) anti-CD3 monoclonal antibody OKT3 (Drug Bank reference: DB00075), anti-CD19 anitbody MDX-1342 (WO2009054863; WO2007002223).
106 WSGF
[00424] The final gene products were sub-cloned i
expression vector pTT5 (NRC-BRI, Canada) and expressed in CHO cells (Durocher, Y., Perret, S. & Kamen, A. High-level and high-throughput recombinant protein production by transient transfection of suspension-growing CHO cells. Nucleic acids research 30, E9 (2002)).
[00425] The CHO cells were transfected in exponential growth phase (1 .5 to 2 million cells/mL) with aqueous 1 mg/ml_ 25kDa polyethylenimine (PEI, Polysciences) at a PEhDNA ratio of 2.5:1 .(Raymond C. et al. A simplified polyethylenimine-mediated transfection process for large-scale and high- throughput applications. Methods. 55(1 ):44-51 (201 1 )). In order to determine the optimal concentration range for forming heterodimers, the DNA was transfected in optimal DNA ratios of the heavy chain A (HC-A), light chain (LC), and heavy chain B that allow for heterodimer formation (e.g. HC-A/HC-B/ ratios = 50:50% (OAAs; HC/Fc), 50:50%. Transfected cells were harvested after 5-6 days with the culture medium collected after centrifugation at 4000rpm and clarified using a 0.45μηη filter.
[00426] The clarified culture medium was loaded onto a MabSelect SuRe (GE Healthcare) protein-A column and washed with 10 column volumes of PBS buffer at pH 7.2. The antibody was eluted with 10 column volumes of citrate buffer at pH 3.6 with the pooled fractions containing the antibody neutralized with TRIS at pH 1 1. The protein was finally desalted using an Econo-Pac 10DG column (Bio-Rad).
[00427] In some cases, the protein was futher purified by protein L chromatography by the method as follows. Capto L resin PBS was equilibrated with PBS and protein A purified v875, neutralized with 1 M Tris, was added to resin and incubated at RT for 30 min. Resin washed with PBS and flow through collected, bound protein was eluted with 0.5 ml 0.1 M Glycine, pH 3.
[00428] In some cases, the protein was further purified by gel filtration,
3.5mg of the antibody mixture was concentrated to 1.5ml_ and loaded onto a
Superdex 200 HiLoad 16/600 200pg column (GE Healthcare) via an AKTA
Express FPLC at a flow-rate of 1 mL/min. PBS buffer at pH 7.4 was used at a
107 WSGF flow-rate of 1 ml_/min. Fractions corresponding to the p
collected, concentrated to ~1 mg/ml_ and stored at -80°C.
[00429] The transient expression of exemplary v873, 874, 875 and other azymetric antibodies compared to the reference v891 is shown in Figure 7.
The SDS-PAGE in Figure 7 shows that all examplary heteromultimer can be expressed transiently in CH03E7 cells with a cell viability of > 80 %.
[00430]
[00431] Example 3: Heteromultimer v873 is able to bridge Jurkat CD3 T cells and Raji CD19 B cells.
[00432] The ability of v873 to bridge T cells and B cells was tested by FACS analysis as follows.
[00433] Whole Cell Bridging by FACS
[00434] 1 x 106 cells/ml suspended in RPMI were labeled with 0.3 μΜ of the appropriate CellTrace label and mixed and incubated at 37°C in a water bath for 25 minutes
[00435] Pellets were resuspended in 2 ml of L10 + GS1 + NaN3 to a final concentration 5x x106 cells/ml.
[00436] Cell suspensions were analyzed (1/5 dilution) by flow cytometry to verify the appropriate cell labeling and laser settings. Flow-check and flow-set Fluorospheres were used to verify instrument standardization, optical alignment and fluidics.
[00437] After flow cytometry verification, and prior to bridging, each cell line was mixed together at the desired ratio, at a final concentration of 1x106 cells/ml.
[00438] T:T bridging was assessed with Jurkat-violet + Jurkat-FarRed, B:B was assessed with RAJI-violet + RAJI-FarRed and T:B bridging was assessed with Jurkat-violet + RAJI-FarRed.
[00439] Antibodies were diluted to 2x in L10+GS1 +NaN3 at room
temperature then added to cells followed by gentle mixing and a 30 min incubation.
108 WSGF
[00440] Following the 30 min incubation 2 μΙ of pre
and slowly mixed and immediately analyze by flow cytometry.
[00441] Bridging % was calculated as the percentage of events that are simultaneously labeled violet and Far-red.
[00442]
[00443] Figure 1 B shows the ability of v873 and blinatumomab CD19-CD3 BiTE (v891 , MT-103) to bridge Jurkat CD3 T cells (Top left quadrant) with Raji CD19 B cells (bottom right quadrant) by FACS. Bridged T-B cells appear in top right quadrant. This result demonstrates that at 300 nM, heteromultimer v873 is able to specifically bridge Jurkat T cells and Raji B cells to a similar extent (23% of total cells) as BiTE (21 % of total cells).
[00444]
[00445] Example 4: Heteromultimers bind selectively to CD3- and CD19- expressing cells.
[00446] The ability of an exemplary heteromultimer, v873, to bind specifically to CD3 and CD19 was assessed by FACS. One-armed antibodies (OAAs) against CD3 and CD19 were also prepared as described in Example 2 and tested as controls in the whole cell FACS binding assay described below.
[00447] Whole Cell Binding by FACS Protocol:
[00448] 2x106 cells/ml cells (> 80% viability) were resuspended in L10+GS1 media, mixed with antibody dilutions, and incubated on ice for 1 h.
[00449] Cells were washed by adding 10ml of cold R-2 buffer, and centrifuging at 233g for 10 min at 4°C. The cell pellet was resuspended with 100 μΙ (1/100 dilution in L10+GS1 media) of fluorescently labeled anti-mouse or anti-human IgG and incubated for 1 h at RT.
[00450] Cell treatments were washed by adding 10ml of cold R-2 as previously described, and the cell pellet resuspended with 400 μΙ of cold L-2 and the sample was filtered through Nitex and added to a tube containing 4 μΙ of propidium iodide.
[00451] Samples were analyzed by flow cytometry.
109 WSGF
[00452] The results are shown in Figure 2 and der
able to selectively bind and bridge to CD3-expressing Jurkat T cells (lower panel) and to CD19-expressing Raji B cells (upper panel). Figure 2 also demonstrates that the one-armed anti-CD3 antibody specifically binds to Jurkat T cells and does not cross-react to CD19 expressing B cells, and that the one- armed anti-CD19 antibody specifically binds to Raji B cells and does not cross- react to Jurkat T cells.
[00453] This experiment was repeated and results are shown in Figure 9. As in the previous experiment, the FACS assay shows that v873 binds selectively to Jurkat T-cells and to Raji B-cells (Figure 9 B). Figure 9A shows that human IgG (hlgG) does not bind to Jurkat T-cells and has low level binding to Raji B-cells, as expected due to the interactions between the hlgG Fc and CD32B on the Raji B-cells. Figure 9A also shows that the anti-CD19 OAA binding selectively to the Raji B-cells and does not cross-react to Jurkat T cells.
[00454] The FACS assay was also carried out to confirm that v873 does not bind to control cell lines that do not express CD3 or CD19. Figure 10 shows that v873 does not bind to the K562 cell line, which does not express CD19 or CD3. Figure 1 1 shows that v873 also does not bind to mouse lymphoid cells which does not express CD19 or CD3.
[00455]
[00456] Example 5: Heteromultimers mediate PBMC killing of target Raji B cells
[00457] In preliminary experiments, the ability of an exemplary
heteromultimer, v873, to mediate T cell cytoxicity against target Raji B-cells was measured using IL-2-stimulated PBMCs as follows.
[00458] Human blood (120-140 ml_) for individual studies was collected on two subsequent days from selected donors. On both days, PBMC were freshly isolated from donors and where needed a portion of the PMBC were passed through EasySep (STEMCELL Technologies Inc.) columns for CD4+ and CD8+ enrichment. On the first day, the PBMCs and the enriched fractions were activated with 1000-3000 units per mL of IL-2 with an overnight incubation. The
110 WSGF culture was processed with a negative selection colurr
CD4+ and CD8+ cells. Cells were then labeled and analyzed by cytometry to evaluate the contents of CD69+ cells in the preparations. The PBMCs and the enriched fractions from the second day were used in the assay as is without IL- 2 activation or in resting state.
[00459] Resting and IL-2 activated PBMCs and purified CD4+ and CD8+ were used as effector cells and Raji human B cells as target cells in the cytotoxicity assay. An effectontarget ratio of 30:1 for PBMCs and 15:1 was tested for purified CD4+ and CD8+. The PBMC were also tested in presence of het-FC at 20 ug/mL and cell cytotoxicity was assayed at various concentrations of test items.
[00460] After incubating the cells with test articles for 20-26 hours, 50 uL of cell culture supernatant was collected for LDH analysis using a Promega LDH enzyme kit. In some studies, autologous T and B cells and/or allogenic B cells were assessed for their respective proportions in the culture and their 7AAD+ cell contents
[00461] CFSE was used and tested as a differential label between Raji and autologous B cells. Raji target cells were pre-labeled with minimal amounts of CFSE before the incubation with effectors, with or without test items. The cell pellets were resuspended in various antibody cocktails for flow cytometry analysis. A Guava 8HT flow cytometer was used for analysis of cell
subpopulations. All antibodies were obtained from BD biosciences unless specified.
[00462] Each condition tested included appropriate controls; wells with all effector and target cell types separately, for all donors, incubated with all test items at all concentrations used in the potency assay.
[00463] To avoid any hindrance between binding of the test items to their targets (CD3 and CD19), markers used for B and T cell staining were anti- CD20 and anti-CD7, respectively for the study shown in Figure 23. This assay was performed with resting and IL2-activated PBMCs, purified CD4+ and CD8+ T cells.
1 1 1 WSGF
[00464] Data analysis:
[00465] For LDH analysis, optical densities (OD) at 490 nm were determined for each well using a Molecular Devices Emax. Data analysis was performed using LibreOffice Calc software. The following analysis scheme is applied to calculate the cytotoxic response:
[00466] First, the averaged culture medium background signal (OD values) is subtracted from all wells before evaluating cytotoxic response. For detergent induced maximum release of pure target cells, a volume corrected specific background signal accounting for the presence of detergent is used and subtracted from maximum release OD values.
[00467] The spontaneous release of effectors and target combined is obtained from wells without any test items, for each effector population tested: the PBMCs, CD4 and CD8 negatively selected populations. The background LDH activity of the test system is better evaluated in wells containing the experimental mixture of effectors -target populations without any test item present.
[00468] Between all plates of all donors, spontaneous and maximum LDH release from Raji cells, media control wells, and LDH positive control wells were used to monitor inter plate variations.
[00469] Figure 3A depicts the ability of v873 to redirect IL-2 activated PBMC to kill target Raji B cells from 3 donors. Figure 3B demonstrates that v873 is able to mediate higher redirected T-cell cytotoxicity than v891 in one of the donors. These preliminary results indicate that heteromultimers as exemplified by v873 may be able to mediate higher T-cell cytotoxicity than v891.
[00470] The abililty of v873 to mediate PBMC killing of Raji B cells in 3 donors compared to human lgG1 control was assessed. The methods for this experiement follow that outlined in Example 5, with the following modifications:
IL-2 stimulation of PBMC was tested at 1000-3000 units per mL.
[00471] The results shown in Figure 8 indicated that v873 induces are higher % cytotoxicity to target B cells when compared to negative control
1 12 WSGF human lgG1 (G1 ) when comparing across individual c
stimulation with a higher concentration of IL-2 at 3000 units per ml_, resulted in higher % cytotoxicity to target B cells.
[00472] Example 6: Heteromultimers mediate redirected killing of target Raji B cells with resting and IL-2 activated CD4+ and CD8+ T cells
[00473] The ability of an exemplary heteromultimers, v875, v1379, v1380 to mediate CD4+ and CD8+ T cell cytoxicity against target Raji B-cells was measured as described in Example 5.
[00474] Figure 16A depicts the ability of v875, v1379 and v1380 to mediate antibody dependent B cell cytoxicity by redirected CD4+ and CD8+ T cell towards Raji B cells. The top panel of Figure 16 illustrates the cytoxicity of resting CD4+ and CD8+ T cells towards Raji B cells. These results illustrate that v875, v1380 and v891 ellicit a concentration dependant cytotoxic response that is more prominent in the CD8+ T cells. The lower panel of Figure 16 illustrates that the cytoxicity of IL-2 stimulated CD4+ and CD8+ T cells towards target Raji B cells. These results indicate that v875 and v1379 ellicit similar
Raji B-cells cytoxicity compared to v891 with IL-2 activated CD8+ effector T cells. In all cases, a greater cytotoxic response was elicted with the IL-2 activiated CD8+ T cells compared to the IL-2 activated CD4+ T cells. These results also indicate that the FcGr knock-out variant, v1380, is cytotoxic toward target Raji B cells, but is slightly less effective (ca. 40% max cytotoxicity) compared to v875 and v1379 (ca. 60% max) that have WT heterodimers Fc.
[00475] Figure 16 B-E depict representations of the data in Figure 16A normalized to human IgG, for v875 (Figures 16B and C), and v1379 and v1380
(Figures 16D and E), and include % cytotoxicity indicated at each test antibody concentration. Figure 16B shows the % cytotoxicity to target Raji B cells with v875 with IL-2 activated CD4+ and CD8+ effector T cells. Figure 16C shows the % cytotoxicity to target Raji B cells with v875 with resting CD4+ and CD8+ effector T cells. Figure 16D and E show direct comparisons of v1379 (WT Fc) and v1380 (L234A_L235A Fc knockout) in IL-2 activated (Figure 16D), and resting (Figure 16E) CD4+ and CD8+ T cells. The most signicant impact of the
113 WSGF
L234A_L235A Fc knockout (v1380) to target B cell cyl
2 activated CD8+ T cell population (Figure 16D, left panel), where v1380 is less cytotoxic compared to v1379.
[00476] The experiment was repeated as shown in Figures 19 with v875, v873 and human IgG, using resting (Figure 19A) and IL-2 activated CD8+ T cells (Figure 19B) as effectors cells targeting Raji B cells. Figure 19A shows that v875 and v873 elicit >30% cytotoxicity to target Raji B cells with IL-2 activiated CD8+ T cells as effectors, and maximal target cell killing is seen at the 3 nM concentration. Figure 19B shows that v875 and v873 elicit dose dependent (>20%) cytotoxicity to target Raji B cells with resting CD8+ T cells as effectors. Greater target Raji B cell killing is seen when IL-2 activated CD8+ T cells were used as the effector cells.
[00477] The experiment was repeated as shown in Figure 20 to compare the relative contributions of CD4+ and CD8+ T cell killing to target Raji B cells at v875 concentations ranging from 0.06-10.0 nM. Figure 20A shows the target Raji B cell cytotoxicty of v875 with IL-2 activated CD4+ and CD8+ T cells. The percent target B cell killing elicited with IL-2 activated CD4+ cells and CD8+ T cells, did not increase at v875 concentrations above the 0.06 nM. As expected, target B cell killing is greater with IL-2 activated CD8+ T cells, compared to IL-2 activated CD4+ T cells. Figure 20B shows the target Raji B cell cytotoxicty of v875 with resting CD4+ and CD8+ T cells. The percent target B cell killing elicited with resting CD8+ cells does not increase greatly at v875
concentrations above 0.1 nM. Dose-dependent B cell killing is seen with v875 and v873 when CD4+ and CD8+ effector T cells are used. As expected, target B cell killing is greater with resting CD8+ T cells, compared to resting CD4+ T cells.
[00478] Example 7: The heterodimeric Fc contributes to target Raji B cell cytotoxicity
[00479] The ability of exemplary heteromultimers, v875, and v873, to mediate target Raji B-cell cytotoxicity in the presence and absence of Fc was measured as described in Example 5.
114 WSGF
[00480] Figure 17 depicts v875 and v873 Azymetri
activated PBMC and target Raji B cell cytotoxicity in the presence or absence of Fc blocking as determined by LDH assay. Figure 17A illustrates that Fc blocking of IL-2 activated PBMC results in a minor (v875) or no (v873) reduction in the % cytotoxicity of target Raji B cells. Figure 17B illustrates that Fc blocking of resting PBMC results in a reduction in the % cytotoxicity of target Raji B cells for v875 and v873.
[00481] The experiment was repeated and the results shown in Figure 18. Figure 18A illustrates that Fc blocking of IL-2 activated PBMC results in a reduction in the % cytotoxicity of target Raji B cells at all antibody
concentrations tested for v875 and v873. Figure 18B illustrates that Fc blocking of resting PBMC results in a reduction in the % cytotoxicity of target Raji B cells at all antibody concentrations tested for v875 and v873. Figures 17 and 18 show that the Fc contributes to target Raji B cell cytotoxicity in the
heterodimeric v875 and v873 antibodies.
[00482] Example 8: Heteromultimers mediate autologous B cell cytotoxicity [00483] The ability of exemplary the heteromultimer v875 and v873 to kill autologous B-cells was measured in total and resting IL-2-stimulated PBMCs where the percent of CD19+ 7AAD+ cells following incubation with v875 and v873 (300 nM, n=3 donors) was determined by flow cytometry as described in Example 5.
[00484] Figure 21 shows, relative to untreated media and human IgG controls, that v875 and v873 (300 nM) mediate autologous B cell killing in total resting PBMC (left panel) and total IL-2 activated PBMC .
[00485] Example 9: Heteromultimer v875 spares autologous T cell cytotoxicity compared to BiTE
[00486] The effects of exemplary heteromultimer v875 and v873 treatment on the autologous T cell population was assesed in total and resting IL-2- stimulated PBMCs where the percent of CD3+ 7AAD+ cells following
incubation with v875 and v873 (300 nM, n=3 donors) was determined by flow cytometry as described in Example 5.
115 WSGF
[00487] Figure 22 shows, relative to untreated me<
controls, v875 has a more selective B cell killing by sparing more autologous T cells compared to v873 and v891.
[00488] Example 10: Design, expression and purification of heteromultimers with an albumin scaffold.
[00489] The following exemplary CD3-CD19 binding heteromultimers based on an albumin scaffold were designed and prepared as follows.
[00490] The sequences for the anti-CD19 and anti-CD3 scFvs were chosen from two molecules that are currently in clinical trials and are well documented and tested for stability and production. The anti-CD19 and anti-CD3 scFv were directly adopted from the BiTE molecule blinatumomab. The antiCD3 scFv was chosen in the VH-VL orientation, consistent with what used in BiTE. The benchmark molecule was an scFv molecule based on BiTE (v891 ).
AlbuCORE_1 (ABH2) CD3/CD19 fusions were created by attaching the antiCD3 warhead to the natural N terminus of fragment 1 and the antiCD19 to the C terminus of fragment 2 (v1092, polypeptide sequences corresponding to SEQ ID NO:264 and 266). The linkers used were identical to the ones used for the multivalent HER2 AlbuCORE experiments: GGGS at the N terminus of fragment 1 and (GGSG)4GG at the C terminus of fragment 2. A second molecule was created where the warheads were reversed (i.e. anti-CD19 warhead at the natural N terminus of fragment 1 and the anti-CD3 at the C terminus of fragment 2, v1093. v1094 was designed to accommodate two different fusions at the natural termini of the albumin polypeptide (polypeptide sequences corresponding to SEQ ID NO:268). The scFv fusions were linked to the albumin molecule through a GGS linker at the N terminus and a GGSG linker at the C terminus. The length of the linkers reflect the ones used in the MM-1 1 1 molecule, despite having a different sequence type.
[00491] V221 is the albumin-based heteromultimer used to construct v1092, but without the cargo molecules (polypeptide sequences corresponding to SEQ ID NO:269 and 270).
116 WSGF
[00492] Expression and purification were performe
described for the multivalent HER2.
[00493] Example 1 1 : Heteromultimers with an albumin scaffold bind specifically to CD3- or CD19-expressing cells
[00494] The ability of Anti-CD3 x CD19 loaded AlbuCORE-1 (v1092) to CD3+ and CD19+ cells was assessed using FACS and compared to WT-HSA loaded with the same anti-CD scFvs (v1094).
[00495] The results are shown in Figure 4 and demonstrate that both v1092 and v1094 are able to bind to CD3-expressing Jurkat T-cells and to CD19- expressing Raji B-cells.
[00496] Example 12: Heteromultimers with heterodimeric Fc or albumin scaffolds show comparable B-cell targeting and T-cell bridging
[00497] The ability of heteromultimers with different scaffolds to direct B-cell targeting and T-cell bridging was compared by FACS analysis, according to the method described in Example 3. The v1092 construct was additionally tested against v873 and the v891 BiTE control for B-cell and T-cell binding according to the method described in Example 4.
[00498] The results are shown in Figure 5A and indicate that at the concentration tested, v1093 was able to bridge 31 % of total cells, and v873 was able to bridge 25% of total cells (lower panels). The upper panels are the results using media as a control. As described in Example 3, the ability of v873 to bridge B-cells and T-cells is comparable to that of the v891 BiTE control. Figure 5A further indicates that the ability of v1093 to bridge B-cells and T-cells is comparable to that of v873.
[00499] In an additional experiment, the ability of v1092 to bridge T-cells and B-cells was directly compared to the v221 control and to v873 and the v891 control. The results are shown in Figure 5B which demonstrates that v1092 is able to bridge Jurkat T cells and Raji B cells to a greater extent than v221 and similar to v891 and v873.
[00500] Example 13: Exemplary heteromultimers have higher anti-CD3 KD and higher Bmax in binding to T and B cell as determined by FACS.
117 WSGF
[00501] The KD of exemplary heteromultimers, v87
assessed by FACS as described as described in Example 4 with data analysis and curve fitting performed in GraphPad Prism.
[00502] The results of this experiment are shown in Figure 12 and indicate that both v873 and v875 have a higher anti-CD3 affinity compared to v891. The KD for v873 and v875 binding to CD19 expressing Raji cells is similar across all antibodies and is comparable to v891. Figure 12 A and B show FACS binding curves of v873, v875 and v891 to CD3 expressing HPB-ALL and Jurkat T cells, and to CD19 expressing Raji B cells. Figure 12 B also illustrates the v875 has a higher Bmax for binding to Raji B cell and Jurkat T cells when compared to v891. Table 1 summarises the KD values for v875, v873, and v891 binding to HPB-ALL, Jurkat and Raji cells.
[00503] Table 1 : KD summary of T and B cell binding determined by FACS
Figure imgf000121_0001
[00520]
[00521] The KD of exemplary heteromultimers v875, v1379, v1380, v1381 , and v891 was assessed for binding to CD19 expressing Raji cells by FACS as described as described in Example 4 with data analysis and curve fitting performed in GraphPad Prism.
[00522] The results of this experiment are shown in Figure 13 A and Table 2 and indicate that heteromultimers v875, v1379, v1380, v1381 , and v891 have a similar KD for binding to CD19 expressing Raji cells. Figure 13A illustrates
1 18 WSGF the FACS binding curve obtained with the antibodies t
range. Table 2 summaries the derived KD in nM, Bmax and Hill slope obtained from the FACS binding experiment. Figure 13A also shows that all
heteromultimers tested have a higher Bmax for binding to CD19 expressing Raji cells compared to BiTE.
[00523] Table 2: Summary of binding properties of heteromultimers to Raji B cells
Figure imgf000122_0001
[00548]
[00549] The results of the FACS binding to H BP-ALL T cells are shown in Figure 13B and Table 3 and indicate that heteromultimers v875, v1379, v1380, have a lower KD compared to v891. Figure 13B illustrates the FACS binding curve obtained with the antibodies tested in 0.1 to 300 nM range. Table 3 summaries the derived KD in nM, Bmax and Hill slope obtained from the FACS binding experiment. Figure 13B also shows that all heteromultimers tested have a higher Bmax for binding to HBP-ALL compared to v891.
[00550] Table 3: Summary of binding properties of heteromultimers to HBP-ALL T cells
[00551]
Figure imgf000122_0002
119 WSGF
Figure imgf000123_0001
[ 5 ]
[00581] Example 14: Heteromultimer v875 is able to bridge Jurkat CD3 T cells and Raji CD19 B cells.
[00582] The ability of the heteromultimer, v875, to bridge T cells and B cells was tested by FACS analysis as described in Example 3.
[00583] The results of this experiment are shown in Figure 14 and indicate that both v875 and v891 facilitate comparable bridging between Raji B-cells and Jurkat T-cells. Use of the control human IgG resulted in 2.5% bridging between Raji and Jurkat cells, while v875 facilitated bridging of 22.9% of total cells, and v891 facilitated bridging of 14.5% of total cells. These results were also presented as fold bridging over background, where v875 mediated a 9.2- fold increase in bridging over background, while v891 mediated a 5.8-fold increase in bridging over background.
[00584] Example 15: Bridging of B and T cells by heteromultimers is robust at varying antibody concentrations or cell ratios.
[00585] The ability of v875 to bridge B and T cells at varying concentrations was assessed by FACS as described in Example 3 with modifications including changes to the effector to target (E:T) cell ratios (1 :1 or 15:1 ) at three different concentrations of v875.
[00586] The results are shown in Figures 15A and 15B. Figure 15A shows the amount of bridging using a 1 :1 ratio of T-cells to B-cells, with
heteromultimer concentrations ranging from 0.3 nM to 3 nM. Figure 15B shows the amount of bridging using a 15:1 ratio of T-cells to B-cells, with
heteromultimer concentrations ranging from 0.3 nM to 3 nM. Both E:T ratios
120 WSGF
(1 :1 and 15:1 ) tested with v875 resulted in similar tota
when expressed as fold over background.
[00587] Example 16: Bridging of B and T cells is robust across differently engineered heteromultimers constructs.
[00588] The ability of v875 to bridge Raji B and Jurkat T cells (B:T), as well RajkRaji B cell bridging (B:B) and Jurkat:Jurkat T cell bridging (T:T) was assessed by FACS as described in Example 3.
[00589] The results are shown in Figures 35A, 34B and 34C. Figure 34A shows the amount of T:B, B:B and T:T bridging of v875, v1379, v1380, v891 , v1381 , commercial OKT3 and human IgG over three experimental replicates. These results show that all heterodimeric antibodies mediate a high percentage of T:B bridging and that all variants have a % T:B bridging that is higher than v891. These results also show a low percentage of T:T and B:B bridging relative to the % T:B bridging for all variants.
[00590] Figure 34B shows the amount of T:B, B:B and T:T bridging of variants with engineered anti-CD3 warheads for stability enhancement (v1653, v1654, v1655, v1656, v1660, v1800, v1802) and v875, and human IgG. These results show that all heterodimeric antibodies (v875, v1653, v1654, v1655, v1656, v1660, v1800, v1802) mediate a higher percentage of T:B bridging compared to human IgG negative control, and all variants mediate low T:T bridging. These results also show that some variants (v1660, v1654 and v1655) mediate higher B:B bridging relative to T:B bridging.
[00591] Figure 34C shows the amount of T:B, B:B and T:T bridging of Fc knock-out variants that have either engineered anti-CD3 warheads for stability enhancement (v1666), or have human/cynomolgous monkey cross-reactive anti-CD3 and anti-CD19 scFvs (v4541 , v4543, v4545, v4548) commercial OKT3 anti-CD3 control, v2176 anti-CD19 control and human IgG negative control, and all variants mediate low T:T bridging. These results show that all heterodimeric antibodies (v1666, v4541 , v4543, v4545, v4548) mediate a higher percentage of T:B bridging compared to human IgG negative control.
121 WSGF
These results also show that some variants (v1666, v<
bridging relative to T:B bridging.
[00592] Example 17: Effects of v875, v1380, v1379 on IL-2 activated and resting CD20+, CD4+, CD8+ subsets
[00593] The effects of v875, v1379 and v1380 treatment on the viability of CD20+, CD4+, CD8+ subsets in IL-2 activated or resting T and B cell cultures, was examined by 7AAD+ staining and FACS and normalized to isotype control (n=4 donors), as described in Example 5.
[00594] The results are shown in Figure 23, and illustrate that the exemplary heteromultimers v875, v1379 and v1380, mediate CD20+ B cell cytotoxicity but not CD4+ or CD8+ T cell cytotoxocity relative to the human IgG control. Figure 23A shows the effects of v875 on the viability of CD20+, CD4+, CD8+ subsets in IL-2 activated cell cultures. Figure 23B shows the effects of v875 on the viability of CD20+, CD4+, CD8+ subsets resting cell cultures.
Figure 23C shows the effects of v1379 and v1380 on the viability of CD20+, CD4+, CD8+ subsets in IL-2 activated cell cultures. Figure 23D shows shows the effects of v1379 and v1380 on the viability of CD20+, CD4+, CD8+ subsets resting cell cultures. The contribution of the WT Fc to CD20+ B cell cytotoxicity is also show in Figures 23C and D, where v1379 mediates a greater to CD20+ B cell cytotoxicity in both IL-2 activated and resting CD4+ and CD8+ T cells.
[00595] Example 18: Exemplary heteromultimers v875 and v873 require the presence of both effector T cell and target B cells to mediate cytotoxic effects [00596] The impact of incubating v875 and v873 alone with either effector or target cells was assessed by LDH release as decribed in Examples 5 with the following modifications. A total of 300,000 resting and IL-2 activated PBMC, 150,000 CD8+ effector cells or 10,000 Raji target cells were incubated overnight with each of the antibodies at 300 nM, along with a control without test item for each condition. The data presented is the averaged results from the 3 donors
[00597] The results of antibody mediated LDH release in resting effector and Raji B cells shown in Figure 24A and the results of antibody mediated LDH
122 WSGF release in activiated effector are shown in Figure 24B.
v875 and v873 are not cytotoxic to CD8+ T cell populations relative to the untreated media and human IgG control. Figure 24A also shows that v875 and 873 increase the cytoxoicity in total PBMC populations, likely due to the redirected killing of effector T cells to target B cells. Additonally, Figure 24A shows that in the absence of effector T cells, v875 and v873 are not cytotoxic to Raji B cell populations relative to the media and human IgG control.
[00598] Figure 24B shows the results of v875 and v873 mediated LDH release in activated effector PBMC and CD8+ T cells. These results show an increase in cell death when v875 and v873 are incubated with activated PBMC like due to the redirected B cell killing mediated by the heterdimeric antibodies in the presence of effector T and target B cells. Figure 24B also shows that incubation of v875 with activated CD8+ T cells, results in a higher percentage of cell death relative to v873 and the media and human IgG controls.
[00599] Example 19: Exemplary heteromultimers can mediate ADCC or impaired ADCC to target Daudi B cells
[00600] Antibody-dependent cell-mediated cytotoxicity assays (ADCC) were performed with v875, v1379 and v1380 using Daudi cells as target B cells and FcRy3a immobilized NK92 cells as the effector cells (GS193761 ) by the following method.
[00601] Dose-response studies were performed with various concentrations of the samples with a pre-optimized Effector/Target (E/T) ratio (5:1 ). Triton X- 100 was added to cell controls without effector cells and antibody in a final concentration of 1 % to lyze the target cells and it served as the maximum lysis control; assay buffers were added in to cell controls without effector cells and antibody and it served as the minimum LDH release control. Target cells incubated with effector cells without the presence of antibodies were set as background control of non-specific LDH release when both cells were incubated together.
[00602] Test article was incubated with cells at 37°C/5%C02 for 5-6 hours and cell viability was assessed with a LDH kit. The absorbance was read at
123 WSGF
OD492nm and OD650nm. The percentages of cell lys
calculated according the formula below:
[00603] Cell lysis %=100*(1 -(ODSample data- ODtumor cells plus effector cells) / (ODMaximum release - ODMinimum release)). Half maximal effective concentration (EC50) values were analyzed withthe Sigmoidal dose-response non-linear regression fit by GraphPad Prism.
[00604] The results are shown in Figure 25A and B, and illustrate that heteromultimers with a WT Fc (v875 and v1379) can mediate ADCC (ca. 40% max cell lysis), whereas v1380, with a L234A_L235A knock Fc mutation, is impaired in ADCC to target Daudi B cells. In Figures 25 A and B, comparison to the internal positive control, Rituximab, are presented.
[00605] Example 20: Exemplary heteromultimers have impaired CDC- mediated lysis of Daudi B cells
[00606] Cell based complement dependent cytotoxicity assays (CDC) were performed with v875, v1379 and v1380 using Daudi cells as target B cells. Human serum from healthy donors (NHS) was used as the source of complement. 10 μΙ NHS (10% final concentration in 40 reaction volume) were added to each well to initiate the CDC cascade and incubated for 2 hours. Cell viability was measured with CellTiter-Glo® Luminescent Cell Viability Assay Kit.
[00607] The percentage of cell viability was calculated with the formula:
[00608] % Cell viability= 100 * ( (RLUsample) / (RLUcell+NHS)), in which NHS stands for normal human serum. Half maximal effective concentration (EC50) values were analyzed with the Sigmoidal dose-response non-linear regression fit by GraphPad Prism.
[00609] Figure 25C and D show the results of the CDC assay with v1380 and v1379 (Figure 25C) and v875 (Figure 25D) of target Daudi B cells with comparisons to positive control Rituximab. All heteromultimers show impaired CDC relative to Rituximab, and mediate a maximum target cell lysis of 15% or less.
124 WSGF
[00610] Example 21 : Cell proliferation and cytokin<
exemplary heteromultimers
[00611] The impact of incubating exemplary heteromultimers on cell proliferation and cytokine release was examined in PBMC and PBMC derived subpopulations. The PBMC derived subpopulations included PBMC, PBMC without B cells (PBMC - B), PBMC without NK cells (PBMC - NK), PBMC without NK and B cells (PBMC-NK-B), CD8+ T cells with B cells, and CD8+ without B cells. Briefly, Four (4) donors were tested for PBMC and PBMC derived subpopulations CD19-, CD56- or CD19/CD56-depleted PBMC populations) and two (2) donors were tested for CD8 and CD8 plus B cells during an incubation period of 4 and 6 days. The proliferation assay was performed with test items at 2 different concentrations (0.3 and 100 nM) each with thymidine incorporation as proliferation readout. The cytokines were measured using the CBA (Cytometric Bead Array) platform on a flow cytometer by the method described below.
[00612] A blood sample was collected from 6 normal donors (the CD8 counts of potential donors used in CD8 panel were determined previously), of whom two donors with high CD8 counts were selected for the CD8
experimental panel; one donor was also scheduled as back-up. On Day 1 , about 135 ml_ of blood was collected from each of the 4 donors (for the PBMC panel), whereas on Day 2 -165 ml of blood was collected from each of the 2 donors (for the CD8 panel). On both days, PBMC were freshly isolated and the PMBC were passed through EasySep columns (STEMCELL Technologies Inc.) for CD19 and/or CD56 depletion by positive selection (day 1 ) and CD8 (±CD19) enrichment by negative selection (day 2).
[00613] To verify the composition/purity/viability of the selected
subpopulations for both days, an antibody cocktail composed of CD8-
FITC/CD56-PE/7AAD/CD19-PECY7/CD20-APC was employed.
[00614] The test items were prepared at 2x concentration for a final (after dilution with the cells) of 0.3 and 100 nM; were added in 100 ul of volume. The
PBMCs were plated at 250,000 cells/well in 100 ul of suspension; the CD8
125 WSGF fractions were plated at 150,000 cells/well. The mixtur
and 5 days respectively, after which 50 ul/well of cell supernatant was transferred into low binding plates and freeze for later cytokine analysis. 50 ul of tritiated thymidine was added to the cell containing wells for a final of 0.5uCi thymidine/well; the plates were incubated for an additional 18 hours, after which the plates were frozen. Total incubation times were 4 and 6 days.
[00615] The plates were thawed two days later, filtered and counted (CPMs) using a β-counter.
[00616] The cell proliferation was assayed at 2 concentrations of test items (0.3 and 100 nM). From the averages, a Stimulation Index (SI) was calculated as follows and the data was tabulated: average CPM of test item/ average CPM of media only
[00617] The composition/viability (trypan blue, 7AAD) of PBMCs and CD8- enriched cell populations were assessed for their respective proportions after selection using an antibody cocktail including anti:
CD8/CD56/7AAD/CD20/CD56.
[00618] Once the proliferation results were analyzed, the supernatant from the replicates were pooled, excluding any identified outlier from the proliferation results. The pooled supernatant was used for cytokine measurements, in duplicates, using the CBA Human Th1/Th2 Cytokine Kit II from BD
Biosciences. This kit measures IL-2, IL-4, IL-6, IL-10, TNF and IFg.
[00619] The results from the cell proliferation 4-day incubation assay are shown in Figure 26 for the 0.3 nM (top panel) and the 100 nM (bottom panel) concentrations. The top panel of Figure 26 shows that at 0.3 nM, v875 and v1380 do not induce PBMC proliferation compared to human IgG. The lower panel of Figure 26 shows the results of the 100 nM antibody concentrations, and shows that v875, v1380 and v891 induce higher cell proliferation relative to human IgG. Figure 26 (lower panel) also shows that at 100 nM, v875 has a similar proliferative index compared to anti-CD3 OKT3 in all four PBMC populations. However, compared to v875, v1380 (L234A_L235A Fc knock out variant) and v891 mediate a reduced cell proliferation and show a trend toward
126 WSGF
B cell dependence for cell proliferation, where the cell
the PBMC-B and PBMC-B-NK subpopulations.
[00620]
[00621] Figure 28 show the results from the average stimulation index induced by v875 at 0.3
[00622] nM (Figure 28A) and 100 nM (Figure 28B) concentrations on purified CD8+ T cells in the absence or presence of purified CD19+ B cells at 4 days incubation time-point. Figure 28A shows that at 0.3 nM, v875 has a higher stimulation index compared to human IgG and lower compared to OKT3.
Figure 28A also shows that v875 has a higher stimulation index of CD8+ T cells in the presence of target B cells (CD8+B). Figure 28B shows that at 100 nM, v875 has a similar stimulation index compared to OKT3 and higher compared to human IgG with little to no influence of target B cell populations.
[00623]
[00624] Figure 29 shows the results from the average stimulation index induced by v1380 at 0.3nM (Figure 29A) and 100nM (Figure 29B)
concentrations on purified CD8+ T cells in the absence or presence of purified CD19+ B cells at 4 days incubation time-point. Figure 29A shows that at 0.3 nM, v1380 has a higher stimulation index compared to human IgG and lower compared to OKT3. Figure 28A also shows that v1380 has a higher stimulation index of CD8+ T cells in the absence of target B cells (CD8). Figure 29B shows that at 100 nM, v1380 has a similar stimulation index of CD8+ T cells compared to OKT3 and a higher stimulation index compared to human IgG. Similar to the 0.3 nM data, v1380 has a higher stimulation index of CD8+ T cells in the absence of target B cells (CD8). The L234A_L235A Fc knockout mutation in v1380 seems to reduced the B-cell dependent stimulation of CD8+ T cells that was apparent with the exemplary WT Fc variant v875.
[00625]
[00626] The results from the cytokine release assay are shown in Figure 27 and include summary plots of PBMC supernatant TNFa (Figure 27A) INFy
(Figure 27B), IL-2 (Figure 27C), IL-4 (Figure 27D), and IL-10 (Figure 27E)
127 WSGF levels following incubation with test items at 0.3 nM cc
(graph y-axis represents log cytokine levels in pg per ml_ from 4 donors).
Figure 27 shows that v1380 (L234A_L235A Fc knockout) induces less cytokine release of TNFa, INFy, IL-2, IL-4, and IL-10 when compared to v875 (WT Fc) and OKT3.
[00627]
[00628] Example 22: Exemplary heteromultimers can bridge two or more target B cells per effector T cell
[00629]
[00630] The ability and ratio of numbers of T cell bridged to B cells was examined with the exemplary heteromultimer v875 by microscopy using the method described in Example 3 with the following modifications.
[00631]
[00632] Labeled Raji B cells (red) and labeled Jurkat T cells (blue) were incubated for 30 min at RT with 3 nM of human IgG or v875. The cell suspension was concentrated by removing 180 μΙ of supernatant. Cell were resuspended in the remaining volume and imaged at 200x and 400X.
[00633]
[00634] Figure 30 shows the results from the T:B cell bridging microscopy comparing v875 and human IgG (3 nM) at 200X and 400X magnification; the phase image (top panel), fluorescence image (middle panel) and inverted fluorescence (bottom panel) are presented. Figure 30A shows a direct comparison of human IgG and v875 at 200X magnification and illustrates a higher amount of bridging visible between Raji B cell and Jurkat T cells compared to human IgG. Figure 30B and Figure 30C show two fields of view for v875 (Figure 30B) and human IgG (Figure 30C) at 400X magnification.
Figure 30B shows images of v875-mediated immune complex formation between Jurkat T cells (dark grey cells in fluorescence inverted image) and Raji
B cells (light grey cells in fluorescence inverted image), and that one Jurkat T cell can bridge 1 -3 Raji B cells. Figure 30C shows images following human
IgG incubation with Jurkat T cells and Raji B cells. Figures 30C shows an
128 WSGF absence of the Jurkat-Raji bridging following incubatic
compared to the v875-mediated JurkatRaji bridging that is visible in Figure
30B.
[00635] Example 23: Exemplary heteromultimer binding to Fey Receptors as assessed by surface plasmon resonance
[00636] The ability of the exemplary heteromultimeric antibodies to bind to FcyRs CD16a and CD32a/b was examined using Surface Plasmon Resonance (SPR).
[00637] Surface Plasmon Resonance Analysis: Affinity of FcyR receptors to antibody Fc was measured by SPR using a ProteOn XPR36 system from BIO- RAD. Purified anti-CD3/anti-CD19-based antibodies were indirectly captured when injected at 25Dl_/min for 240s (resulting in approx. 500RUs) following a buffer injection to establish a stable baseline.
[00638] FcyR concentrations (10,000, 3333, 1 1 1 1 , 370, 123nM) were injected at 60μΙ_/ηιίη for 120s with a 180s dissociation phase to obtain a set of binding sensograms. Resultant KD values were determined from binding isotherms were fit globally to a 1 : 1 Langmuir binding model with reported values as the mean of three independent runs.
[00639] The results of the SPR binding studies are shown in Table 4.
[00640]
[00641] Table 4: KD for heterodimeric antibody binding to Fc gamma receptors
Figure imgf000132_0001
[00642]
[00643] Table 4 summarized the KD data for exemplary heteromultimer binding to CD16a, CD16aV158, CS32a, CD32aR131 , CD32b and CD32bY163.
129 WSGF
These results show that WT Fc variants v875, v1379 ;
all Fc gamma receptors. These results also show that Fc knock out variants have impaired binding to some (v1380) or all (v1381 ) Fc gamma receptors.
[00644]
[00645] Example 24: Exemplary heteromultimers can bind to the human and cynomologous CD3 T cell receptor
[00646] The binding of exemplary heteromultiers for binding to the human CD3 and cynomologous receptors was examined by ELISA by the following method.
[00647] Human or cynomologous CD3 receptor antigen was diluted at 20 μg/ml in PBS, in Costar 3690 high binding half area high binding microplate, incubated overnight at 4 °C. Wells were washed 3 times with PBS and blocked with BSA 1 % in PBS for 30 min (50 ul/well). Primary heteromultimeric antibodies were diluted in BSA 1 % at the indicated concentrations and incubated for 2 hours at RT and wells washed 4 times with PBS-0.05% Tween 20. Secondary antibody (Jackson 1 15-036-062: anti-mouse or 709-036-098 anti-human Fcgamma specific) was diluted 1/5000 in BSA 1 % (25 μΙ/well) and incubated for one hour at RT and washed 4 times with PBS-0.05%Tween 20. TMB substrate was added (25 μΙ/well) for 25-30 min., and the reactions was stopped with 1 M H2S04 (12.5 μΙ/well) and OD read at 450 nm.
[00648] The ELISA data is shown in Figure 35 and illustrates binding to human CD3 (top panel) and binding to the cynomologous CD3 receptor (bottom panel) as determined by ELISA. Figure 35 shows that v4543, v4545, and v4548 show that highest degree of cross-reactivity to the cynomologous CD3 receptor (Figure 35 bottom panel).
[00649] Example 25: Expression and Purification of heteromultimers
[00650] Description of the methods used in the expression and purification of exemplary heteromultimer are described in Example 2.
[00651] Figure 31 A shows the SDS-PAGE analysis and relative purity of v875, v1380, v1379 and v891 following protein A and SEC purification, and
130 WSGF following 47 day storage at 4°C. The two visibile protc
sample indicate a breakdown product of this sample due to storage.
[00652] Figure 31 B shows the SDS-PAGE analysis and relative purity of additional exemplary hetermultimers including v875, v1653, v1654, v1655, v1656, v1660, v1800, and v1802 following protein A and SEC purification.
[00653] Example 26: Exemplary heteromultimers can be purified to > 99% heterodimer purity and < 1 % aggregates.
[00654] The purity of exemplary heteromultimers was tested by LC-MS. The heteromultimers were first purifed by protein A, protein L and SEC purification as described in Example 2. LC-MS analysis for heterodimer purity was performed as described below.
[00655] The purified samples were de-glycosylated with PNGase F for 6 hr at 37°C. Prior to MS analysis the samples were injected onto a Poros R2 column and eluted in a gradient with 20-90% ACN, 0.1 % FA in 3 minutes, resulting in one single peak.
[00656] The peak of the LC column was analyzed with a LTQ-Orbitrap XL mass spectrometer using the following setup: Cone Voltage: 50 V Tube lens: 215 V; FT Resolution: 7,500. The mass spectrum was integrated with the software Promass or Max Ent. to generate molecular weight profiles.
[00657] The LC-MS results of the Max Ent. molecular weight profiles for v875 are shown in Figure 32 and the results summarized in Table 5.
[00658]
[00659] Table 5: Heterodimer purity of v875
[00660]
Figure imgf000134_0001
[00661] Table 5 shows that following protein A and SEC purification, v875 consistend of 99.27% heterodimer purity (H1 H2) and less than 1 % of the anti-
CD3 homodimer (H2H2) and anti-CD19 homodimer (H1 H1 ).
[00662] The purity and percent aggregation of exemplary protein A and
SEC purified heteromultimers was determined by UPLC-SEC by the method described.
131 WSGF
[00663] UPLC-SEC analysis was performed using
column set to 30°C (2.5 mL, 4.6 x 150 mm, stainless steel, 1.7 μηη particles) at
0.4 ml/min. Run times consisted of 7 min and a total volume per injection of 2.8 mL with running buffers of 25 mM sodium phosphate, 150 mM sodium acetate, pH 7.1 ; and, 150 mM sodium phosphate, pH 6.4-7.1. Detection by absorbance was facilitated at190-400 nm and by fluorescence with excitation at 280 nm and emission collected from 300-360 nm. Peak integration was analyzed by
Empower 3 software.
[00664] The LC-MS and UPLC-SEC results for exemplary heteromultimers is summarized in Table 6. Table 6 shows that all heteromultimers have a
heterodimer purity >95% as determined by LC-MS, and all heteromultimers <
2% aggregates as determined by UPLC-SEC analysis.
[00665]
[00666] Table 6: Summary of LC-MS and UPLC-SEC analysis of exemplary heterodimers
Figure imgf000135_0001
#: Interference from an adjacent peak making exact quantification impossible.
True relative intensity % of this peak is likely less than presented value.
[00667] * ky LC-MS after treatment with neuraminidase
[00668] Example 27: Exemplary heteromultimers have a CH3 Tm that is greater than 75°C
[00669] CH3 domain stability of exemplary heteromultimers was examined by DSC using the following method. All DSC experiments were carried out
using a GE VP-Capillary instrument. The proteins were buffer-exchanged into
PBS (pH 7.4) and diluted to 0.3 to 0.7mg/mL with 0.137mL loaded into the
sample cell and measured with a scan rate of 1°C/min from 20 to 100°C. Data
132 WSGF was analyzed using the Origin software (GE Healthca
background subtracted.
[00670] The DSC results shown in Figures 33 A, B and C show that v875 has an estimated CH3 Tm > 76°C (Figure 33A), v1380 has an estimated CH3 Tm > 82.3°C (Figure 33B), and v1379 has an estimated CH3 Tm > 82.5°C (Figure 33C).
[00671] Example 28: Design, expression and purification of CD3/CD20 and additional CD3/CD19 heteromultimer constructs.
[00672] V5850 (corresponding to polypeptide sequences SEQ ID NOs: 203, 205 and 207), v5851 (corresponding to polypeptide sequences SEQ ID NOs: 209, 21 1 and 213), v5852 (corresponding to polypeptide sequences SEQ ID NOs: 215, 217 and 219), v6324 (corresponding to polypeptide sequences SEQ ID NOs: 221 , 223 and 225), v6325 (corresponding to polypeptide sequences SEQ ID NOs: 225, 227 and 229), v1813 (corresponding to polypeptide sequences SEQ ID NOs: 231 , 233 and 235), v1821 (corresponding to polypeptide sequences SEQ ID NOs: 237, 239 and 241 ), v1823 (corresponding to polypeptide sequences SEQ ID NOs: 243, 245 and 247) exemplify bispecific CD3/CD19 or CD3/CD20 hybrid heterodimeric Fc constructs. Bispecific hybrid variants are composed of a F(ab') on either chain A or B paired with an scFv-Fc on the alternate polypeptide chain. Chain A of the heterdimer Fc is comprised of the following mutations: T350V_L351Y_F405A_Y407V and Chain B of the heterdimer Fc is comprised of the following mutations:
T350V_T366L_K392L_T394W. v6324 exemplifies bispecific CD3/CD20 scFv heterodimeric Fc constructs. V1813, v1821 , and v1823 exemplify CD3/CD20 common light chain heterodimeric Fc constructs. Common light chain variants are composed of two different F(ab')s, each on complimentary heterodimer Fc, which share a single light chain. The specific variant composition is indicated in Table 7.
[00673] The anti-CD19 MOR208_scFv-Fc(VHVL) used in v5852 was generated by fusing the published variable heavy chain sequence to the variable light chain sequences indicated in table 7 with a (GGGGS)3 linker
133 WSGF between the heavy and light chain. The variable dome
B of the heterdimer Fc.
[00674] The anti-CD20 Ofatumumab_scFv-Fc(VHVL) used in v6324 and v6325 was generated by fusing the published variable heavy chain sequence to the variable light chain sequences indicated in Table 7 with a (GGGGS)3 linker between the heavy and light chain. The variable domains were fused to Chain B of the heterdimer Fc.
[00675] Cloning, expression and purification was performed as indicated in Example 2.
[00676] Yield and purity of the variants is indicated in Table 8. Heterodimer purity was determined by LCMS analysis according to Example 26. All the variants demonstrated heterodimer purity in excess of 73.8% with an average purity of 89.6% for all variants tested. The samples had low amounts of incorrectly paired homodimers ranging from 0 to 5.3% of the total product. Reported values represent the sum of all observed homodimer species. The presence of half-antibodies was more commonly observed than homodimers and ranged from 0 to 20.7% of the total product. Reported values represent the sum of all observed half-antibody species
[00677] Table 7. Composition of CD3/CD19 or CD20 hybrid variants
Figure imgf000137_0001
134 WSGF
Figure imgf000138_0001
[00678
[00679]
[00680]
[00681] Table 8. Variant expression and purity
Figure imgf000138_0002
[00682]
135 [00683] "unknown species detected by LCMS resulted
[00684] Example 29: CD3/CD20 and additional CD3/Cu l a neteromummer variants bind to T cells and B cells.
[00685] The ability of the exemplary CD3/CD20 heteromultimeters, v5850, v6324, v6325, v1813, v1821 , v1823 to bind to CD3 and CD20 cells were assessed via FACS analysis as per procedures described in Example 4. Additionally, the ability of an exemplary CD3/CD19 heteromultimers, v5851 and v5852 to bind to CD3 and CD19 cells were similarily assessed. An additional variant v875, a CD3/CD19 BiTE Fc antibody construct, were also prepared and tested as benchmarks. Representative binding curves for v875, v5850 and v5851 on Raji and Jurkat cells are shown in Fig 36A & 38B. The binding results for each variant expressed in kinetic constants Bmax and Kd are listed below in Tables 9 and 10. Table 8 describes the binding to the CD19 and CD20- expressing Raji B cells, while Table 10 describes binding to the CD3-expresssing Jurkat T cells. In Raji binding studies (Table 9) all the variants bound with a greater Bmax and a higher Kd compared to 875. In Jurkat binding studies (Table 10) all the variants, except v1823, bound with a higher Bmax than 875 and had a range of KD's.
[00686] Table 9 (Raji)
[00687]
v875 v4542 v5850 v5851 v5852 v6324 v6325 vl813 vl821 vl823
Bmax 2.78 2.96 4.24 3.88 na 6.91 6.44 6.40 4.71 4.14
(OD450)
KD 0.36 0.70 3.60 1.38 na 25.35 11.87 4.04 122.5 21.05
(nM)
[00688]
[00689] Table 10 (Jurkat)
v875 v4542 v5850 v5851 v5852 v6324 v6325 vl813 vl821 vl823
Bmax 1.59 2.27 2.06 2.51 2.21 2.32 2.51 2.54 2.1 1 0.88
(OD450)
KD 21.36 6.66 4.04 4.24 25.24 11.62 1.58 691.4 181.5 68.77
(nM) [00690]
[00691] Example 30: CD3/CD20 and additional CD3/Cu l a neteromummer variants bridge T cells and B cells.
[00692] The ability of the six exemplary CD3/CD20 heteromultimer variants— namely v5850, v6324, v6325, v1813, v1821 and v1823— and two exemplary CD3/CD19
heteromultimer variants— namely v5851 and v5852— to bridge T cells and B cells were tested via FACS analysis as per procedures described in Example 3. Additional constructs, namely v792 and v875, were also prepared and tested as controls. V792 has identical anti-Her2 F(ab') based on trastuzumab on chain A and chain B of the
heterodimer Fc with the following mutations T350V_L351Y_F405A_Y407V on chain A and T350V_T366L_K392L_T394W on chain B (drug bank accession number - DB00072)
[00693] Table 1 1 and 12 provides the percentage bridging between Jurkat- Jurkat, Raji- Raji, and Jurkat-Raji for each variant, each table represents an individual experiment. All variants were effective at bridging Jurkat and Raji cells. Furthermore, none of the variants bridged two jurkat cells and some Raji-Raji cell bridging was observed to different extents.
[00694] Table 1 1.
Figure imgf000140_0001
[00727]
[00728]
[00729] Table 12.
[00730]
Figure imgf000140_0002
Bridging 792 875 5852
[00737] Ju [00738] [00739] [00740] [00741] [00742] rkat/Jurkat .7 .5 .9 .8 .1
[00743] R [00744] [00745] [00746] [00747] [00748] aji/Raji .7 .6 .2 .5 .7
[00749] Ju [00750] [00751] [00752] [00753] [00754] rkat/Raji .9 5.7 0.4 .7 5.7
[00755]
[00756] Example 31 : Design, expression and purification of HER2/HER3
heteromultimeric constructs.
[00757] To assess various properties of HER2/HER3 bispecific heteromultimeric constructs, two formats and various controls were produced and purified as follows. The first construct was produced by fusing HER2- and HER3-binding scFvs onto a heterodimer Fc region (v878, HER2/HER3 Het-Fc). The second construct was prepared by fusion of Her2 and Her3 scFvs onto an albumin-based platform (v1090, AlbuCORE anti- Her3xHer2). Various controls, including a Her2 One-Armed het-Fc, a Her3 one-armed het- Fc and the control HSA-fusion protein v1087 were also produced as controls.
[00758] Design
[00759] The bispecific HER2/HER3 constructs were produced by fusion of Her2 and Her3 scFvs onto the het-Fc or the AlbuCORE platforms.
[00760] 1. Variant 878: a monovalent one-armed anti-Her2 antibody, where the Her2 binding domain is a scFv on chain A, and the Fc region is a heterodimer having the mutations L351Y_F405A_Y407V in Chain A, and T366L_K392M_T394W in Chain B. The epitope of antigen binding domain is domain 1 of Her2.
[00761] 2. Variant 879: a monovalent one-armed anti-Her3 antibody, where the Her3 binding domain is a scFv on chain B, and the Fc region is a heterodimer having the mutations L351Y_F405A_Y407V in Chain A, and T366L_K392M_T394W in Chain B.
[00762] 3. Variant 880: a bispecific anti-Her2, anti-Her3 antibody, where the Her2 binding domain is a scFv on chain A, the Her3 binding domain is a scFv on chain B and the Fc region is a heterodimer having the mutations L351^
and T366L_K392M_T394W in Chain B.
[00763] 4. Variant 1087: The MM-1 1 1 molecule is a single polypeptide fusion protein of two scFvs, anti-Her2 (B1 D2) and anti-Her3 (H3), linked to the C and N termini,
respectively, of a modified human serum albumin protein, and is produced by Merrimack. The resulting molecule is bispecific and bivalent. As a control, a version of the MM-1 1 1 molecule was constructed in which the anti-Her3 (H3) warhead was fused to the N terminus of albumin by a short AAS linker, while anti-Her2 (B1 D2) was fused to the C terminus through an AAAL linker to create the benchmark control variant 1087. This control variant lacks the C34S/N504Q mutation originally introduced by Merrimack in their MM-1 1 1 molecule, and has a polypeptide sequence corresponding to SEQ ID NO:258.
[00764] 5. Variant 1090: An albumin-based heteromultimer was formed by combining fragment 1 fused to antiHER3 at its N terminus through a GGGS linker [SEQ ID NO:260] and fragment 2 fused to antiHER2 (B1 D2) at its C terminus through a GGGS linker [SEQ ID NO:262] (variant number 1090). This molecule has warheads or cargo polypeptides in cis and is almost identical to v1087. The main differences between the 1087 polypeptide and the 1090 are that 1 ) the linkers used in the 1087 polypeptide were more hydrophobic, while the linkers used on the 1090 variant were polyGLY)(S) and 2) The 1090 variant lacks the C34S/N504Q mutation originally introduced by Merrimack.
[00765] Variants 878, 879, and 880 were expressed and purified as described in Example 2. Variants 1087 and 1090 were expressed and purified as described in
Example 10.
[00766] Example 32: Fc-based HER2/HER3 heteromultimeric constructs bind bispecifically to Her2 and Her3 receptors on MALME-3M cells.
[00767] The ability of the v878, v879 and v880 HER2 and HER3 constructs to bind to Her2 and Her3 was assessed using FACS analysis:
[00768] A dose range of the two one-arm monovalent variants (anti-HER2 one-arm, v878, and anti-HER2 one-arm, v879) and the bispecific anti-HER2/HER3 heterodimer, v880, was inclubated with MALME-3M melanoma cells followed by FACS analysis to determine the binding affinity of each molecule as described in Example 4. [00769] Figure 37A and 37B illustrates affinity on linea
the figures show, the HER2/HER3 het-Fc construct demonstrates Dotn Di-specmcity ana avidity.
[00770] Example 33: Albumin-based HER2/HER3 based constructs to bind
bispecifically to Her2 and Her3 receptors on MALME-3 cells.
[00771] The binding affinity of an albumin-based heteromultimer loaded in a cis configuration with anti-Her2 and anti-Her3 scFvs to MALME-3M cells were assessed and compared to the v1087 control.
[00772] The binding affinity of an exemplary albumin-based heteromultimer variant 1090 (anti-Her2 x anti-Her3 ABH2) for MALME-3M cells was evaluated using FACS with FITC-labeled anti-HSA antibodies, as described in Example 4.
[00773] Figure 38 depicts the binding of variant 1090 compared to the control 1087 in MALME-3M cells and indicates that v1090 has similar binding as v1087 to target MALME- 3M cells.
[00774] Example 34: Monovalent one-armed anti-CD3 antibodies are able to bridge Jurkat T cells and Raji B cells
[00775] The ability of monospecific anti-CD3 antibodies v870, v871 , v872 to bridge T cells and B cells was tested by FACS analysis as described in Example 3.
[00776] The results are summarized in Table 13 and show that v870, v871 and v872 bridge a higher percentage of Jurkat T and Raji B cells when compared to medium and human IgG controls. Table 13 also shows that v870, v871 and v872 bridge a lower percentage of Jurkat T and Raji B cells when compared to the bispecific anti-CD3 anti- CD^ antibody, v873.
[00777] Table 13: Monovalent anti-CD3 antibody bridging of Jurkat T and Raji B cells
Figure imgf000143_0001
v872 OAA_anti-CD3 (VL-VH OKT3) v873 bispecific CD19-CD3(VH-VL-BiTE) 27,6
Human IgG 6
Medium 7,3
[00778]
[00779] It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims.

Claims

WHAT IS CLAIMED IS:
1 ) An isolated multispecific heteromultimer construct comprising:
a first polypeptide construct comprising a first heavy chain polypeptide and a CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell;
a second polypeptide construct comprising a second heavy chain polypeptide which is different from said first heavy chain polypeptide, and an antigen binding polypeptide construct that binds to a target antigen on at least one B cell; wherein:
at least one of said CD3 binding polypeptide construct and said antigen binding polypeptide construct comprises a single chain Fv region;
said multispecific heteromultimer construct simultaneously engages said at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell; and
said first and second heavy chain polypeptides form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc, wherein:
said heterodimeric Fc is formed with stability at least comparable to a native homodimeric Fc, and
said heterodimeric Fc is formed with purity such that when said multispecific heteromultimer construct is coexpressed from a stable mammalian cell in an expression product, said expression product comprises at least about 70% of said multispecific heteromultimer, and less than 10% monomers or homodimers of said first or second polypeptide constructs.
2) The isolated multispecific heteromultimer of claim 1 , wherein said first or second polypeptide construct is devoid of at least one of immunoglobulin light chain, and immunoglobulin first constant (CH1 ) region. 3) The isolated multispecific heteromultimer of ar
heterodimer Fc region comprises a variant CH2 domain or ninge comprising amino acid modifications that prevents functionally effective binding to all the Fcgamma receptors.
4) The isolated multispecific heteromultimer of claim 3 , wherein wherien said variant CH2 domain or hinge comprising amino acid modification also prevents functionally effective binding to complement proteins (C1 q complex).
5) The isolated multispecific heteromultimer of any of claims 1 -2, wherein the heterodimer Fc region comprises a variant CH2 domain or hinge comprising amino acid modifications that enhance binding to the FcyRllb receptor.
6) An isolated multispecific heteromultimer construct comprising:
a first polypeptide construct comprising a first heavy chain polypeptide and a CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell;
a second polypeptide construct comprising a second heavy chain polypeptide which is different from said first heavy chain polypeptide, and an antigen binding polypeptide construct that binds to a target antigen on at least one B cell; wherein:
at least one of said CD3 binding polypeptide construct and said antigen binding polypeptide construct optionally comprises a single chain Fv region;
said first and second heavy chain polypeptides form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc, wherein:
said heterodimeric Fc is formed with stability at least comparable to a native homodimeric Fc, and
said heterodimeric Fc is formed with purity such that when said multispecific heteromultimer construct is coexpressed from a mammalian cell in an expression product, said expression product comprises greater than 70% of said multispecific heteromultimer, and less than 10% mon
first or second polypeptide constructs; and
said multispecific heteromultimer construct binds said at least one B cell with a valency greater than one, and said multispecific heteromultimer simultaneously engages said at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell.
7) An isolated multispecific heteromultimer construct comprising:
a first polypeptide construct comprising a first heavy chain polypeptide and a CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell;
a second polypeptide construct comprising a second heavy chain polypeptide which is different from said first heavy chain polypeptide, and a steric modulator construct which exhibits negligible receptor binding; wherein:
said multispecific heteromultimer construct simultaneously engages at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell; and
said first and second heavy chain polypeptides form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc, wherein:
said heterodimeric Fc is formed with stability at least comparable to a native homodimeric Fc, and
said heterodimeric Fc is formed with purity such that when said multispecific heteromultimer construct is coexpressed from a stable mammalian cell in an expression product, said expression product comprises at least about 75% of said multispecific heteromultimer, and less than 10% monomers or homodimers of said first or second polypeptide constructs.
8) An isolated multispecific heteromultimer construct comprising: a first polypeptide construct comprising a 1
a CD3 binding polypeptide construct that binds to a Cuo complex on at least one CD3 expressing cell;
a second polypeptide construct comprising a second heavy chain polypeptide which is different from said first heavy chain polypeptide, and wherein said second polypeptide construct does not comprise an antigen binding polypeptide construct; wherein:
said multispecific heteromultimer construct simultaneously engages at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell; and
said first and second heavy chain polypeptides form a heterodimeric Fc region comprising a variant immunoglobulin CH3 region comprising at least one amino acid mutation that promotes the formation of said heterodimeric Fc, wherein:
said heterodimeric Fc is formed with stability at least comparable to a native homodimeric Fc, and
said heterodimeric Fc is formed with purity such that when said multispecific heteromultimer construct is coexpressed from a stable mammalian cell in an expression product, said expression product comprises at least about 75% of said multispecific heteromultimer, and less than 10% monomers or homodimers of said first or second polypeptide constructs.
9) The isolated multispecific heteromultimer of any of claims 1 -8 wherein the heterodimer Fc region comprises a variant CH2 domain comprising amino acid modifications to promote selective binding of a Fcgamma receptor.
10) The isolated multispecific heteromultimer of claim 9, wherein the variant CH2 domain selectively binds Fcgammallb receptor greater than to wild-type CH2 domain.
1 1 ) The isolated multispecific heteromultimer of claim 9, wherein the variant CH2 domain selectively binds atleast one of Fcgammallla and Fcgammalla receptor greater than a wild-type CH2 domain. 12) The isolated multispecific heteromultimer according w any one οτ iaims ι - ι i , wherein the variant CH3 domain has a melting temperature (Tm) of about 73°C or greater.
13) The isolated multispecific heteromultimer according to any one of Claims 1 -12, wherein the heterodimer Fc region is formed with a purity greater than about 90%.
14) The isolated multispecific heteromultimer according to any one of Claims 1 -13, wherein the heterodimer Fc region is formed with a purity of about 95% or greater and the Tm is at least about 75°C.
15) The isolated multispecific heteromultimer according to any one of Claims 1 -13, wherein the heterodimer Fc region is formed with a purity of at least about 90% and the Tm is about 75°C.
16) The isolated multispecific heteromultimer according to any one of Claims 1 -15 wherein:
a. the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications L351Y, F405A, and Y407V, and the variant CH3 sequence of the second transporter polypeptide comprises the amino acid modifications T366L, K392M, and T394W; b. the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications L351Y, F405A, and Y407V, and the variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications T366L, K392L, and T394W; c. the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications T350V, L351Y, F405A, and Y407V, and the variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications T350V, T366L, K392M, and T394W; d. the variant CH3 sequence of the first heavy <
amino acid modifications T350V, L351Y, F40OM, ana Y ^U / V , ana tne variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications T350V, T366L, K392L, and T394W; e. the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications T366L, N390R, K392R, and T394W, and the variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications L351Y, S400E, F405A, and Y407V; or f. the variant CH3 sequence of the first heavy chain polypeptide comprises the amino acid modifications T350V, T366L, N390R, K392R, and T394W, and the variant CH3 sequence of the second heavy chain polypeptide comprises the amino acid modifications T350V, L351Y, S400E, F405A, and Y407V.
17) The isolated multispecific heteromultimer of any of claims 1 -16 wherein the heterodimer Fc is glycosylated.
18) The isolated multispecific heteromultimer of any of claims 1 -16 wherein the heterodimer Fc is afucosylated.
19) The isolated multispecific heteromultimer of any of claim 1 -16 wherein the heterodimer Fc is aglycosylated.
20) The isolated multispecific heteromultimer of any of claims 1 -6 and 9-19, wherein said antigen binding polypeptide construct that binds to a target antigen on at least one B cell comprises at least one target antigen binding domain derived from an antibody, a fibronectin, an affibody, anticalin, cysteine knot protein, DARPin, avimer, Kunitz domain or variant or derivative thereof.
21 ) The isolated multispecific heteromultimer of claim 20, wherein said antibody is a heavy chain antibody devoid of light chains. 22) The isolated multispecific heteromultimer of ar
wherein said antigen binding polypeptide construct comprises at least one uu 1 a binding domain.
23) The isolated multispecific heteromultimer of any one of claims 1 -6 and 9-21 , wherein said antigen binding polypeptide construct comprises at least one CD20 binding domain.
24) An isolated multispecific heteromultimer construct comprising:
a first polypeptide construct comprising a first transporter polypeptide fused to at least one CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell;
a second polypeptide construct comprising a second transporter polypeptide which is different from said first transporter polypeptide, fused to at least one antigen binding polypeptide construct that binds to a target antigen on at least one B cell;
wherein said first and second transporter polypeptides are derived from a protein by segmentation of said protein, each transporter polypeptide comprising an amino acid sequence with at least 90% identity to a segment of said protein, and wherein said transporter polypeptides self-assemble to form a quasi-native structure of said monomeric protein.
25) The isolated multispecific heteromultimer of claim 24 wherein said transporter polypeptides are not derived from an antibody.
26) The isolated multispecific heteromultimer of any one of claims 24-25, wherein each transporter polypeptide is an albumin derivative.
27) The isolated multispecific heteromultimer of claim 26, wherein said albumin is human serum albumin. 28) The isolated multispecific heteromultimer of ar
least one transporter polypeptide is an allo-albumin derivative.
29) The isolated multispecific heteromultimer of any of claims 24-25, and 28 wherein each transporter polypeptide is derived from a different alloalbumin.
30) An isolated multispecific heteromultimer construct comprising:
a first polypeptide construct comprising a first transporter polypeptide fused to at least one CD3 binding polypeptide construct that binds to a CD3 complex on at least one CD3 expressing cell;
a second polypeptide construct comprising a second transporter polypeptide which is different from said first transporter polypeptide, fused to at least one antigen binding polypeptide construct that binds to a target antigen on at least one B cell;
wherein said first and second transporter polypeptides are obtained by segmentation of albumin, and each transporter polypeptide comprising an amino acid sequence with at least 90% identity to a segment of albumin such that said transporter polypeptides self-assemble to form quasi-native albumin, and wherein said first cargo polypeptide does not have any binding domain present in said second cargo polypeptide.
31 ) The isolated multispecific heteromultimer of any of claims 24-30, wherein said multispecific heteromultimer construct simultaneously engages said at least one B cell and said at least one CD3 expressing cell such that the CD3 expressing cell is activated, thereby inducing killing of the B cell.
32) The isolated multispecific heteromultimer of any of claims 24-30, wherein said antigen binding polypeptide construct that binds to a target antigen on at least one B cell comprises at least one target antigen binding domain derived from an antibody, a fibronectin, an affibody, anticalin, cysteine knot protein, DARPin, avimer, Kunitz domain or variant or derivative thereof. 33) The isolated multispecific heteromultimer of ar
said antigen binding polypeptide construct comprises at least one uu 1 a Dinaing domain.
34) The isolated multispecific heteromultimer of any one of claims 24-32, wherein said antigen binding polypeptide construct comprises at least one CD20 binding domain.
35) The isolated multispecific heteromultimer of any of claims 1 -34, wherein said at least one CD3 binding polypeptide construct comprises at least one CD3 binding domain derived from a CD3 specific antibody, a nanobody, fibronectin, affibody, anticalin, cysteine knot protein, DARPin, avimer, Kunitz domain or variant or derivative thereof.
36) The isolated multispecific heteromultimer of claim 35, wherein said at least one CD3 binding domain comprises at least one amino acid modification that reduces immunogenicity as compared to a corresponding CD3 binding domain not comprising said modification.
37) The isolated multispecific heteromultimer of any of claims 1 -36, wherein said at least one CD3 binding domain comprises at least one amino acid modification that increases its stability as measured by Tm, as compared to a corresponding CD3 binding domain not comprising said modification.
38) The isolated multispecific heteromultimer of any of claims 36-37, wherein said CD3 specific antibody is a heavy chain antibody devoid of light chains.
39) The isolated multispecific heteromultimer of any of claims 36-37, wherein said at least one CD3 binding polypeptide construct comprises at least one CD3 binding domain derived from a non-antibody protein scaffold domain. 40) The isolated multispecific heteromultimer of ar
least one of said first and second polypeptide constructs Turiner comprises a singie- chain Fv polypeptide.
41 ) The isolated multispecific heteromultimer of any of claims 1 -40 wherein at least one of said first and second polypeptide constructs further comprises a single- chain Fab polypeptide.
42) The isolated multispecific heteromultimer of any of claims 1 -41 where in the CD3 expressing cell is a T-cell.
43) The isolated multispecific heteromultimer of claim 42, wherein said
heteromultimer binds to the T-cell with sufficient affinity and decorates the T cell at sufficient capacity that induces the T-cell to display B cell killing activity when the T cell and the B cell are bridged.
44) The isolated multispecific heteromultimer of any of claims 1 -43 where in the CD3 expressing cell is a human cell.
45) The isolated multispecific heteromultimer of any of claims 1 -44 where in the CD3 expressing cell is a non-human, mammalian cell.
46) The isolated multispecific heteromultimer of any of claims 1 -45 where in the at least one CD3 binding polypeptide construct binds to CD3 constructs across multiple species.
47) The isolated multispecific heteromultimer of any of claims 1 -46 wherein the at least one B cell is associated with a disease.
48) The isolated multispecific heteromultimer of claim 47 wherein the disease is a cancer selected from a carcinoma, a sarcoma, leukaemia, lymphoma and glioma. 49) The isolated multispecific heteromultimer of cl;
least one of squamous cell carcinoma, adenocarcinoma, transition cen carcinoma, osteosarcoma and soft tissue sarcoma.
50) The isolated multispecific heteromultimer of any one of claims 1 -49, wherein the at least one B cell is an autoimmune reactive cell that is a lymphoid or myeloid cell.
51 ) The isolated multispecific heteromultimer of any one of claims 1 -50, wherein said heteromultimer further comprises at least one binding domain that binds at least one of: EpCAM, EGFR, IGFR, HER-2 neu, HER-3, HER-4, PSMA, CEA, MUC-1 (mucin), MUC2, MUC3, MUC4, MUC5, MUC7, CCR4, CCR5, CD19, CD20, CD33, CD30, ganglioside GD3, 9-0-Acetyl-GD3, GM2, Poly SA, GD2, Carboanhydrase IX (MN/CA IX), CD44v6, Sonic Hedgehog (Shh), Wue-1 , Plasma Cell Antigen,
(membrane-bound), Melanoma Chondroitin Sulfate Proteoglycan (MCSP), CCR8, TNF-alpha precursor, STEAP, mesothelin, A33 Antigen, Prostate Stem Cell Antigen (PSCA), Ly-6; desmoglein 4, E-cadherin neoepitope, Fetal Acetylcholine Receptor, CD25, CA19-9 marker, CA-125 marker and Muellerian Inhibitory Substance (MIS) Receptor type II, sTn (sialylated Tn antigen; TAG-72), FAP (fibroblast activation antigen), endosialin, LG, SAS, EPHA4 CD63, CD3 BsAb immunocytokines TNF, IFNy, IL-2, and TRAIL.
52) The isolated multispecific heteromultimer of any of claims 1 -51 wherein said heteromultimer optionally comprises at least one linker.
53) The isolated multispecific heteromultimer of claim 52, wherein said at least one linker is a polypeptide comprising from about 1 to about 100 amino acids.
54) A set of expression vectors for expressing a multispecific heteromultimer of any one of claims 1 -53, comprising at least a first DNA sequence encoding said first polypeptide construct and at least a second DNA sequence encoding said second polypeptide construct. 55) A method of producing an expression product
heteromultimer of any one of claims 1 -53, in stable mammalian cells, the method comprising: transfecting at least one mammalian cell with:
at least a first DNA sequence encoding said first polypeptide construct and at least a second DNA sequence encoding said second polypeptide construct, such that said at least one first DNA sequence, said at least one second DNA sequence are transfected in said at least one mammalian cell in a pre-determined ratio to generate stable mammalian cells;
culturing said stable mammalian cells to produce said expression product comprising said multispecific heteromultimer.
56) The method of claim 55, wherein said predetermined ratio of the at least one first DNA sequence: at least one second DNA sequence is about 1 :1.
57) The method of any one of claims 55-56, wherein said mammalian cell is selected from the group consisting of a VERO, HeLa, HEK, NSO, Chinese Hamster Ovary (CHO), W138, BHK, COS-7, Caco-2 and MDCK cell, and subclasses and variants thereof.
58) A pharmaceutical composition comprising a multispecific heteromultimer as defined in any one of claims 1 -53, and a suitable excipient.
59) A process for the production of a pharmaceutical composition of claim 57, said process comprising:
a. culturing a host cell under conditions allowing the expression of a
heteromultimer as defined in one of claims 1 -53;
b. recovering the produced heteromultimer from the culture; and
producing the pharmaceutical composition.
60) A method for the prevention, treatment or amelioration of at least one of: a proliferative disease, a minimal residi
an inflammatory disease, an immunological disoraer, an autoimmune aisease, an infectious disease, viral disease, allergic reactions, parasitic reactions, graft-versus-host diseases or host-versus-graft diseases or cell malignancies, said method comprising administering to a subject in need of such a prevention, treatment or amelioration a pharmaceutical composition of claims 58.
61 ) A method of treating cancer in a mammal in need thereof, comprising administering to the mammal a composition comprising an effective amount of the pharmaceutical composition of claim 58, optionally in combination with other pharmaceutically active molecules.
62) The method of claim 61 , wherein said cancer is a solid tumor.
63) The method of claim 62, wherein said solid tumor is one or more of sarcoma, carcinoma, and lymphoma.
64) The method of claim 61 , wherein said cancer is a hematological cancer.
65) The method of claim 64, wherein said cancer is one or more of B-cell lymphoma, non-Hodgkin's lymphoma, and leukemia.
66) A method of treating cancer cells comprising providing to said cell a
composition comprising a heteromultimer provided in one of claims 1 -53.
67) The method of claim 66 further comprising providing said heteromultimer in conjugation with another therapeutic agent.
68) A method of treating a cancer non-responsive to at least one of a CD19 lytic antibody, a CD20 lytic antibody and blinatumomab, in a mammal in need thereof, comprising administering to the mammal a composite
amount of the pharmaceutical composition of claim 5o.
69) A method of treating a cancer cell regressive after treatment with
blinatumomab, comprising providing to said cancer cell a composition comprising an effective amount of the pharmaceutical composition of claim 58.
70) A method of treating an individual suffering from a disease characterized by expression of B cells, said method comprising providing to said individual an effective amount of a composition comprising an effective amount of the pharmaceutical composition of claim 58.
71 ) The method of claim 70, wherein said disease is not responsive to treatment with at least one of an anti-CD19 antibody and an anti-CD20 antibody.
72) A method of treating an autoimmune condition in a mammal in need thereof, comprising administering to said mammal a composition comprising an effective amount of the pharmaceutical composition provided in claim 58.
73) The method of claim 72, wherein said autoimmune condition is one or more of multiple sclerosis, rheumatoid arthritis, lupus erytematosus, psoriatic arthritis, psoriasis, vasculitis, uveitis, Crohn's disease, and type 1 diabetes.
74) A method of treating an inflammatory condition in a mammal in need thereof, comprising administering to said mammal a composition comprising an effective amount of the pharmaceutical composition comprising an heteromultimer provided in one of claims 1 -53.
75) A kit comprising a heteromultimer as defined in any one of claims 1 -53, and instructions for use thereof.
PCT/US2013/050411 2012-07-13 2013-07-13 Bispecific asymmetric heterodimers comprising anti-cd3 constructs WO2014012085A2 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
RU2015102193A RU2650868C2 (en) 2012-07-13 2013-07-13 Bispecific asymmetric heterodimers comprising anti-cd3 constructs
EP13816697.0A EP2872170A4 (en) 2012-07-13 2013-07-13 Bispecific asymmetric heterodimers comprising anti-cd3 constructs
CN201380037369.3A CN104640562A (en) 2012-07-13 2013-07-13 Bispecific asymmetric heterodimers comprising anti-cd3 constructs
CA2878843A CA2878843A1 (en) 2012-07-13 2013-07-13 Bispecific asymmetric heterodimers comprising anti-cd3 constructs
BR112015000798A BR112015000798A2 (en) 2012-07-13 2013-07-13 Bispecific asymmetric heterodimers comprising anti-cd3 constructs
KR20157003872A KR20150036606A (en) 2012-07-13 2013-07-13 Bispecific asymmetric heterodimers comprising anti-cd3 constructs
AU2013289883A AU2013289883B2 (en) 2012-07-13 2013-07-13 Bispecific asymmetric heterodimers comprising anti-CD3 constructs
JP2015521877A JP2015528003A (en) 2012-07-13 2013-07-13 Bispecific asymmetric heterodimer comprising anti-CD3 construct
HK15108247.7A HK1207575A1 (en) 2012-07-13 2015-08-25 Bispecific asymmetric heterodimers comprising anti-cd3 constructs -cd3

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201261671640P 2012-07-13 2012-07-13
US61/671,640 2012-07-13
US201361845948P 2013-07-12 2013-07-12
US61/845,948 2013-07-12

Publications (3)

Publication Number Publication Date
WO2014012085A2 true WO2014012085A2 (en) 2014-01-16
WO2014012085A3 WO2014012085A3 (en) 2014-03-27
WO2014012085A9 WO2014012085A9 (en) 2015-03-19

Family

ID=49916711

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/050411 WO2014012085A2 (en) 2012-07-13 2013-07-13 Bispecific asymmetric heterodimers comprising anti-cd3 constructs

Country Status (11)

Country Link
US (3) US20140154253A1 (en)
EP (1) EP2872170A4 (en)
JP (1) JP2015528003A (en)
KR (1) KR20150036606A (en)
CN (1) CN104640562A (en)
AU (1) AU2013289883B2 (en)
BR (1) BR112015000798A2 (en)
CA (1) CA2878843A1 (en)
HK (1) HK1207575A1 (en)
RU (1) RU2650868C2 (en)
WO (1) WO2014012085A2 (en)

Cited By (98)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015143079A1 (en) * 2014-03-19 2015-09-24 Regeneron Pharmaceuticals, Inc. Antibody compositions for tumor treatment
WO2015153765A1 (en) 2014-04-01 2015-10-08 Adimab, Llc Multispecific antibody analogs comprising a common light chain, and methods of their preparation and use
US9212225B1 (en) 2014-07-01 2015-12-15 Amphivena Therapeutics, Inc. Bispecific CD33 and CD3 binding proteins
EP2914634A4 (en) * 2012-11-02 2016-05-11 Zymeworks Inc Crystal structures of heterodimeric fc domains
US9359437B2 (en) 2013-02-01 2016-06-07 Regeneron Pharmaceuticals, Inc. Antibodies comprising chimeric constant domains
WO2016087416A1 (en) * 2014-12-03 2016-06-09 F. Hoffmann-La Roche Ag Multispecific antibodies
US9493578B2 (en) 2009-09-02 2016-11-15 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
US9499605B2 (en) 2011-03-03 2016-11-22 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs
WO2016204966A1 (en) * 2015-06-16 2016-12-22 Genentech, Inc. Anti-cd3 antibodies and methods of use
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
WO2017055314A1 (en) * 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Bispecific anti-cd19xcd3 t cell activating antigen binding molecules
US9650446B2 (en) 2013-01-14 2017-05-16 Xencor, Inc. Heterodimeric proteins
US9657102B2 (en) 2012-09-21 2017-05-23 Regeneron Pharmaceuticals, Inc. Anti-CD3 antibodies, bispecific antigen-binding molecules that bind CD3 and CD20, and uses thereof
EP3019622A4 (en) * 2013-07-12 2017-06-14 Zymeworks Inc. Bispecific cd3 and cd19 antigen binding contructs
WO2017101828A1 (en) * 2015-12-16 2017-06-22 苏州康宁杰瑞生物科技有限公司 Heterodimer molecule based on ch3 structural domain, and preparation method therefor and use thereof
US9701759B2 (en) 2013-01-14 2017-07-11 Xencor, Inc. Heterodimeric proteins
EP3094737A4 (en) * 2014-01-15 2017-08-09 Zymeworks Inc. Bi-specific cd3 and cd19 antigen-binding constructs
US9732155B2 (en) 2011-11-04 2017-08-15 Zymeworks Inc. Crystal structures of heterodimeric Fc domains
US9738722B2 (en) 2013-01-15 2017-08-22 Xencor, Inc. Rapid clearance of antigen complexes using novel antibodies
US9782478B1 (en) 2011-04-22 2017-10-10 Aptevo Research And Development Llc Prostate-specific membrane antigen binding proteins and related compositions and methods
US9822186B2 (en) 2014-03-28 2017-11-21 Xencor, Inc. Bispecific antibodies that bind to CD38 and CD3
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
US9879095B2 (en) 2010-08-24 2018-01-30 Hoffman-La Roche Inc. Bispecific antibodies comprising a disulfide stabilized-Fv fragment
US9890204B2 (en) 2009-04-07 2018-02-13 Hoffmann-La Roche Inc. Trivalent, bispecific antibodies
US9914785B2 (en) 2012-11-28 2018-03-13 Zymeworks Inc. Engineered immunoglobulin heavy chain-light chain pairs and uses thereof
CN107847559A (en) * 2015-05-13 2018-03-27 埃博灵克斯股份有限公司 T cell based on CD3 reactivity raises polypeptide
WO2018005954A3 (en) * 2016-07-01 2018-03-29 Resolve Therapeutics, Llc Optimized binuclease fusions and methods
US9994646B2 (en) 2009-09-16 2018-06-12 Genentech, Inc. Coiled coil and/or tether containing protein complexes and uses thereof
US10106624B2 (en) 2013-03-15 2018-10-23 Xencor, Inc. Heterodimeric proteins
US10106600B2 (en) 2010-03-26 2018-10-23 Roche Glycart Ag Bispecific antibodies
US10131710B2 (en) 2013-01-14 2018-11-20 Xencor, Inc. Optimized antibody variable regions
WO2018228442A1 (en) * 2017-06-14 2018-12-20 Dingfu Biotarget Co., Ltd. Proteinaceous heterodimer and use 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
US10279034B2 (en) 2017-04-11 2019-05-07 Eli Lilly And Company Anti-PD-L1-anti-TIM-3 bispecific antibodies
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
US10358479B2 (en) 2012-07-13 2019-07-23 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs
CN110218255A (en) * 2018-03-02 2019-09-10 广西医科大学 Nano antibody CD3/Nb47 of AntiCD3 McAb and the preparation method and application thereof
US10428155B2 (en) 2014-12-22 2019-10-01 Xencor, Inc. Trispecific antibodies
US10487155B2 (en) 2013-01-14 2019-11-26 Xencor, Inc. Heterodimeric proteins
US10501543B2 (en) 2016-10-14 2019-12-10 Xencor, Inc. IL15/IL15Rα heterodimeric Fc-fusion proteins
WO2019237081A1 (en) * 2018-06-07 2019-12-12 Cullinan Management, Inc. Multi-specific binding proteins and methods of use thereof
US10508154B2 (en) 2012-06-25 2019-12-17 Zymeworks Inc. Process and methods for efficient manufacturing of highly pure asymmetric antibodies in mammalian cells
WO2019244107A1 (en) * 2018-06-21 2019-12-26 Daiichi Sankyo Company, Limited Compositions including cd3 antigen binding fragments 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
JP2020501550A (en) * 2016-12-13 2020-01-23 デリニア,インコーポレーテッド Multivalent regulatory T cell regulator
US10544187B2 (en) 2013-03-15 2020-01-28 Xencor, Inc. Targeting regulatory T cells with heterodimeric proteins
US10556952B2 (en) 2015-03-30 2020-02-11 Regeneron Pharmaceuticals, Inc. Heavy chain constant regions with reduced binding to Fc gamma receptors
US10611825B2 (en) 2011-02-28 2020-04-07 Hoffmann La-Roche Inc. Monovalent antigen binding proteins
US10640555B2 (en) 2009-06-16 2020-05-05 Hoffmann-La Roche Inc. Bispecific antigen binding proteins
US10662244B2 (en) 2014-11-17 2020-05-26 Regeneron Pharmaceuticals, Inc. Methods for tumor treatment using CD3XCD20 bispecific antibody
WO2020132810A1 (en) * 2018-12-24 2020-07-02 Generon (Shanghai) Corporation Ltd. Multispecific antigen binding proteins capable of binding cd19 and cd3, and use thereof
US10738118B2 (en) 2015-05-29 2020-08-11 Amphivena Therapeutics, Inc. Methods of using bispecific CD33 and CD3 binding proteins
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
US10793621B2 (en) 2011-02-28 2020-10-06 Hoffmann-La Roche Inc. Nucleic acid encoding dual Fc antigen binding proteins
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
US10870701B2 (en) 2016-03-15 2020-12-22 Generon (Shanghai) Corporation Ltd. Multispecific fab fusion proteins and use thereof
US10875931B2 (en) 2010-11-05 2020-12-29 Zymeworks, Inc. Stable heterodimeric antibody design with mutations in the Fc domain
US10968276B2 (en) 2013-03-12 2021-04-06 Xencor, Inc. Optimized anti-CD3 variable regions
US10981992B2 (en) 2017-11-08 2021-04-20 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
US10982006B2 (en) 2018-04-04 2021-04-20 Xencor, Inc. Heterodimeric antibodies that bind fibroblast activation protein
EP3630169A4 (en) * 2017-05-23 2021-04-21 Dragonfly Therapeutics, Inc. A protein binding nkg2d, cd16 and a tumor-associated antigen
US11013800B2 (en) 2011-05-16 2021-05-25 Evive Biotech Ltd. Multi-specific Fab fusion proteins comprising a CD3-binding Fab fragment with N-terminal fusion to binding domains and methods of use
US11053316B2 (en) 2013-01-14 2021-07-06 Xencor, Inc. Optimized antibody variable regions
US11084863B2 (en) 2017-06-30 2021-08-10 Xencor, Inc. Targeted heterodimeric Fc fusion proteins containing IL-15 IL-15alpha and antigen binding domains
US11147886B2 (en) 2015-07-15 2021-10-19 Zymeworks Inc. Drug-conjugated bi-specific antigen-binding constructs
WO2021224913A1 (en) * 2020-05-04 2021-11-11 Immunorizon Ltd. Precursor tri-specific antibody constructs and methods of use thereof
US11203640B2 (en) 2017-07-10 2021-12-21 Eli Lilly And Company Checkpoint inhibitor bispecific antibodies
US11286300B2 (en) 2015-10-01 2022-03-29 Hoffmann-La Roche Inc. Humanized anti-human CD19 antibodies and methods of use
US11306156B2 (en) 2014-05-28 2022-04-19 Zymeworks Inc. Modified antigen binding polypeptide constructs and uses 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
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
EP4032910A1 (en) * 2021-01-22 2022-07-27 ETH Zurich Bispecific binding agent that binds to cd3 and a fluorophore
US11472890B2 (en) 2019-03-01 2022-10-18 Xencor, Inc. Heterodimeric antibodies that bind ENPP3 and CD3
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
EP3873930A4 (en) * 2018-10-23 2022-12-07 Magenta Therapeutics, Inc. Fc silenced antibody drug conjugates (adcs) 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
US11590223B2 (en) 2018-08-31 2023-02-28 Regeneron Pharmaceuticals, Inc. Dosing strategy that mitigates cytokine release syndrome for therapeutic antibodies
US11591401B2 (en) 2020-08-19 2023-02-28 Xencor, Inc. Anti-CD28 compositions
US11739144B2 (en) 2021-03-09 2023-08-29 Xencor, Inc. Heterodimeric antibodies that bind CD3 and CLDN6
US11780920B2 (en) 2020-06-19 2023-10-10 Hoffmann-La Roche Inc. Antibodies binding to CD3 and CD19
US11807692B2 (en) 2018-09-25 2023-11-07 Harpoon Therapeutics, Inc. DLL3 binding proteins and methods of use
US11834506B2 (en) 2017-02-08 2023-12-05 Dragonfly Therapeutics, Inc. Multi-specific binding proteins that bind NKG2D, CD16, and a tumor-associated antigen for activation of natural killer cells and therapeutic uses thereof to treat cancer
US11845799B2 (en) 2019-12-13 2023-12-19 Genentech, Inc. Anti-Ly6G6D antibodies and methods of use
US11859012B2 (en) 2021-03-10 2024-01-02 Xencor, Inc. Heterodimeric antibodies that bind CD3 and GPC3
US11866498B2 (en) 2018-02-08 2024-01-09 Genentech, Inc. Bispecific antigen-binding molecules and methods of use
US11884733B2 (en) 2018-02-08 2024-01-30 Dragonfly Therapeutics, Inc. Antibody variable domains targeting the NKG2D receptor
US11884732B2 (en) 2017-02-20 2024-01-30 Dragonfly Therapeutics, Inc. Proteins binding HER2, NKG2D and CD16
US11919956B2 (en) 2020-05-14 2024-03-05 Xencor, Inc. Heterodimeric antibodies that bind prostate specific membrane antigen (PSMA) and CD3
US11976125B2 (en) 2021-01-15 2024-05-07 Harpoon Therapeutics, Inc. B cell maturation antigen binding proteins

Families Citing this family (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2892059C (en) 2012-11-21 2023-02-14 Wuhan Yzy Biopharma Co., Ltd. Bispecific antibody
EP3406633B1 (en) 2013-07-25 2022-03-02 Cytomx Therapeutics Inc. Multispecific antibodies, multispecific activatable antibodies and methods of using the same
CN105940113A (en) * 2013-11-13 2016-09-14 酵活有限公司 Monovalent antigen binding constructs targeting egfr and/or her2 and uses thereof
US9611325B2 (en) 2014-07-21 2017-04-04 Wuhan Yzy Biopharma Co., Ltd. Construction and application of bispecific antibody HER2xCD3
CA2955947A1 (en) 2014-07-25 2016-01-28 Cytomx Therapeutics, Inc. Anti-cd3 antibodies, activatable anti-cd3 antibodies, multispecific anti-cd3 antibodies, multispecific activatable anti-cd3 antibodies, and methods of using the same
TN2017000106A1 (en) * 2014-09-26 2018-07-04 Macrogenics Inc Bi-specific monovalent diabodies that are capable of binding cd19 and cd3, and uses thereof
RU2754661C2 (en) 2015-05-01 2021-09-06 Дзе Реджентс Оф Дзе Юниверсити Оф Калифорния Glycan-dependent immunotherapy molecules
EP3294768B1 (en) * 2015-05-13 2019-08-21 Ablynx N.V. T cell recruiting polypeptides based on tcr alpha/beta reactivity
CN104892765B (en) * 2015-06-29 2018-04-10 深圳市国创纳米抗体技术有限公司 A kind of bispecific antibody of AntiCD3 McAb antigen and Her 2 antigen
TWI796283B (en) * 2015-07-31 2023-03-21 德商安美基研究(慕尼黑)公司 Antibody constructs for msln and cd3
TWI829617B (en) 2015-07-31 2024-01-21 德商安美基研究(慕尼黑)公司 Antibody constructs for flt3 and cd3
TWI744242B (en) 2015-07-31 2021-11-01 德商安美基研究(慕尼黑)公司 Antibody constructs for egfrviii and cd3
WO2017035430A2 (en) * 2015-08-27 2017-03-02 Kolltan Pharmaceuticals, Inc. Anti-alk antibodies and methods for use thereof
EA039859B1 (en) 2016-02-03 2022-03-21 Эмджен Рисерч (Мюник) Гмбх Bispecific antibody constructs binding egfrviii and cd3
DK3411402T3 (en) 2016-02-03 2022-02-07 Amgen Res Munich Gmbh BCMA AND CD3 BISPECIFIC T-CELL INVOLVING ANTIBODY CONSTRUCTIONS
MX2018009804A (en) * 2016-02-12 2018-11-09 Nantomics Llc High-throughput identification of patient-specific neoepitopes as therapeutic targets for cancer immunotherapies.
BR112018070676A2 (en) * 2016-04-15 2019-02-05 Zymeworks Inc multispecific antigen binding constructs that target immunotherapeutic agents
SG11201810331YA (en) 2016-05-20 2018-12-28 Harpoon Therapeutics Inc Single chain variable fragment cd3 binding proteins
CN106117358A (en) * 2016-06-16 2016-11-16 中山大学 AntiCD3 McAb single domain antibody
US20190218290A1 (en) * 2016-09-07 2019-07-18 Anti-Doping Lab Qatar Camel derived polypeptides binding human cd3, identified using a novel one-step method
CN108395482B (en) 2017-02-08 2021-02-05 西比曼生物科技(香港)有限公司 Construction of targeting CD20 antigen chimeric antigen receptor and activity identification of engineered T cell thereof
WO2018152547A1 (en) * 2017-02-20 2018-08-23 Adimab, Llc Proteins binding cd123, nkg2d and cd16
AU2018250641A1 (en) 2017-04-11 2019-10-31 Inhibrx, Inc. Multispecific polypeptide constructs having constrained CD3 binding and methods of using the same
WO2018199593A1 (en) * 2017-04-24 2018-11-01 재단법인 목암생명과학연구소 Bispecific antibody binding to her3 and cd3
CN111201242A (en) * 2017-06-22 2020-05-26 财团法人生物技术开发中心 Asymmetric heterodimeric FC-SCFV fusion anti-GLOBO H and anti-CD 3 bispecific antibodies and uses thereof in cancer therapy
MX2020002070A (en) 2017-08-22 2020-03-24 Sanabio Llc Soluble interferon receptors and uses thereof.
AU2018347607A1 (en) 2017-10-14 2020-03-26 Cytomx Therapeutics, Inc. Antibodies, activatable antibodies, bispecific antibodies, and bispecific activatable antibodies and methods of use thereof
CN107903324B (en) * 2017-11-15 2021-01-29 北京绿竹生物技术股份有限公司 Bispecific antibody capable of binding to human CD19 and CD3
CN108129566B (en) * 2017-12-31 2021-05-11 中国科学院武汉病毒研究所 C-type single domain antibody of targeting mesothelin and preparation method and application thereof
US20200392512A1 (en) * 2018-02-26 2020-12-17 Ablynx N.V. Nucleotide sequences encoding peptide linkers
AU2019227715B2 (en) 2018-02-28 2022-03-31 Ap Biosciences, Inc. Bifunctional proteins combining checkpoint blockade for targeted therapy
CN112969476A (en) * 2018-12-07 2021-06-15 江苏恒瑞医药股份有限公司 Multispecific protein molecules
EP3917969A4 (en) * 2019-01-28 2023-02-01 Wuxi Biologics Ireland Limited Novel bispecific cd3/cd20 polypeptide complexes
PE20220649A1 (en) * 2019-07-26 2022-04-28 Janssen Biotech Inc PROTEINS COMPRISING KALLYCREIN-RELATED PEPTIDASE 2 ANTIGEN-BINDING DOMAINS AND THEIR USES
CN112390882A (en) * 2019-08-19 2021-02-23 杨洋 Bispecific antibody targeting CD3 and CD20 and application thereof
CN110551222B (en) * 2019-08-27 2023-06-06 重庆市畜牧科学院 Novel bifunctional antibody and application thereof
US20230075633A1 (en) 2019-12-20 2023-03-09 Shandong Boan Biotechnology Co., Ltd. Optimized anti-cd3 arm in the generation of t-cell bispecific antibodies for immunotherapy
TW202140553A (en) 2020-01-13 2021-11-01 美商威特拉公司 Antibody molecules to c5ar1 and uses thereof
EP4259193A1 (en) * 2020-12-09 2023-10-18 Janux Therapeutics, Inc. Compositions and methods related to tumor activated antibodies targeting psma and effector cell antigens
KR20230142831A (en) 2021-01-13 2023-10-11 비스테라, 인크. Humanized Complement 5A Receptor 1 Antibodies and Methods of Using the Same

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6129914A (en) * 1992-03-27 2000-10-10 Protein Design Labs, Inc. Bispecific antibody effective to treat B-cell lymphoma and cell line
US20020062010A1 (en) * 1997-05-02 2002-05-23 Genentech, Inc. Method for making multispecific antibodies having heteromultimeric and common components
US6737056B1 (en) * 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US7183387B1 (en) * 1999-01-15 2007-02-27 Genentech, Inc. Polypeptide variants with altered effector function
US20040132101A1 (en) * 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
TWI353991B (en) * 2003-05-06 2011-12-11 Syntonix Pharmaceuticals Inc Immunoglobulin chimeric monomer-dimer hybrids
US8101720B2 (en) * 2004-10-21 2012-01-24 Xencor, Inc. Immunoglobulin insertions, deletions and substitutions
EP1820513A1 (en) * 2006-02-15 2007-08-22 Trion Pharma Gmbh Destruction of tumor cells expressing low to medium levels of tumor associated target antigens by trifunctional bispecific antibodies
PL2235064T3 (en) * 2008-01-07 2016-06-30 Amgen Inc Method for making antibody fc-heterodimeric molecules using electrostatic steering effects
ES2500066T3 (en) * 2008-01-25 2014-09-30 Amgen, Inc Ferroportin antibodies and methods of use
EP3375790A1 (en) * 2008-10-01 2018-09-19 Amgen Research (Munich) GmbH Cross-species-specific single domain bispecific single chain antibody
CA2750533A1 (en) * 2009-01-23 2010-07-29 Biogen Idec Ma Inc. Stabilized fc polypeptides with reduced effector function and methods of use
PT2519543T (en) * 2009-12-29 2016-10-07 Emergent Product Dev Seattle Heterodimer binding proteins and uses thereof
CN105001330B (en) * 2010-04-23 2020-05-01 弗·哈夫曼-拉罗切有限公司 Production of heteromultimeric proteins
JP6167040B2 (en) * 2010-11-05 2017-07-19 ザイムワークス,インコーポレイテッド Design of stable heterodimeric antibodies with mutations in the Fc domain
WO2013063702A1 (en) * 2011-11-04 2013-05-10 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the fc domain
US20140302037A1 (en) * 2013-03-15 2014-10-09 Amgen Inc. BISPECIFIC-Fc MOLECULES

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
None
See also references of EP2872170A4

Cited By (173)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9890204B2 (en) 2009-04-07 2018-02-13 Hoffmann-La Roche Inc. Trivalent, bispecific antibodies
US11673945B2 (en) 2009-06-16 2023-06-13 Hoffmann-La Roche Inc. Bispecific antigen binding proteins
US10640555B2 (en) 2009-06-16 2020-05-05 Hoffmann-La Roche Inc. Bispecific antigen binding proteins
US9493578B2 (en) 2009-09-02 2016-11-15 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
US9994646B2 (en) 2009-09-16 2018-06-12 Genentech, Inc. Coiled coil and/or tether containing protein complexes and uses thereof
US10106600B2 (en) 2010-03-26 2018-10-23 Roche Glycart Ag Bispecific antibodies
US9605061B2 (en) 2010-07-29 2017-03-28 Xencor, Inc. Antibodies with modified isoelectric points
US9879095B2 (en) 2010-08-24 2018-01-30 Hoffman-La Roche Inc. Bispecific antibodies comprising a disulfide stabilized-Fv fragment
US10875931B2 (en) 2010-11-05 2020-12-29 Zymeworks, Inc. Stable heterodimeric antibody design with mutations in the Fc domain
US10793621B2 (en) 2011-02-28 2020-10-06 Hoffmann-La Roche Inc. Nucleic acid encoding dual Fc antigen binding proteins
US10611825B2 (en) 2011-02-28 2020-04-07 Hoffmann La-Roche Inc. Monovalent antigen binding proteins
US9499605B2 (en) 2011-03-03 2016-11-22 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs
US10711051B2 (en) 2011-03-03 2020-07-14 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs
US10155803B2 (en) 2011-03-03 2018-12-18 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs
US9782478B1 (en) 2011-04-22 2017-10-10 Aptevo Research And Development Llc Prostate-specific membrane antigen binding proteins and related compositions and methods
US11013800B2 (en) 2011-05-16 2021-05-25 Evive Biotech Ltd. Multi-specific Fab fusion proteins comprising a CD3-binding Fab fragment with N-terminal fusion to binding domains and methods of use
US10851178B2 (en) 2011-10-10 2020-12-01 Xencor, Inc. Heterodimeric human IgG1 polypeptides with isoelectric point modifications
US10457742B2 (en) 2011-11-04 2019-10-29 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the Fc domain
US9732155B2 (en) 2011-11-04 2017-08-15 Zymeworks Inc. Crystal structures of heterodimeric Fc domains
US9988460B2 (en) 2011-11-04 2018-06-05 Zymeworks Inc. Crystal structures of heterodimeric Fc domains
US10508154B2 (en) 2012-06-25 2019-12-17 Zymeworks Inc. Process and methods for efficient manufacturing of highly pure asymmetric antibodies in mammalian cells
US10358479B2 (en) 2012-07-13 2019-07-23 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs
US11248037B2 (en) 2012-07-13 2022-02-15 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs
US9657102B2 (en) 2012-09-21 2017-05-23 Regeneron Pharmaceuticals, Inc. Anti-CD3 antibodies, bispecific antigen-binding molecules that bind CD3 and CD20, and uses thereof
US11072656B2 (en) 2012-09-21 2021-07-27 Regeneran Pharmaceuticals, Inc. Anti-CD3 antibodies, bispecific antigen-binding molecules that bind CD3 and CD20, and uses thereof
US11155621B2 (en) 2012-09-21 2021-10-26 Regeneran Pharmaceuticals, Inc. Anti-CD3 antibodies, bispecific antigen-binding molecules that bind CD3 and CD20, and uses thereof
EP2914634A4 (en) * 2012-11-02 2016-05-11 Zymeworks Inc Crystal structures of heterodimeric fc domains
US11078296B2 (en) 2012-11-28 2021-08-03 Zymeworks Inc. Engineered immunoglobulin heavy chain-light chain pairs and uses thereof
US9914785B2 (en) 2012-11-28 2018-03-13 Zymeworks Inc. Engineered immunoglobulin heavy chain-light chain pairs and uses thereof
US10487155B2 (en) 2013-01-14 2019-11-26 Xencor, Inc. Heterodimeric proteins
US9701759B2 (en) 2013-01-14 2017-07-11 Xencor, Inc. Heterodimeric proteins
US10738133B2 (en) 2013-01-14 2020-08-11 Xencor, Inc. Heterodimeric proteins
US11634506B2 (en) 2013-01-14 2023-04-25 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
US10738132B2 (en) 2013-01-14 2020-08-11 Xencor, Inc. Heterodimeric proteins
US10472427B2 (en) 2013-01-14 2019-11-12 Xencor, Inc. Heterodimeric proteins
US11718667B2 (en) 2013-01-14 2023-08-08 Xencor, Inc. Optimized antibody variable regions
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
US10988537B2 (en) 2013-02-01 2021-04-27 Regeneren Pharmaceuticals, Inc. Antibodies comprising chimeric constant domains
US9359437B2 (en) 2013-02-01 2016-06-07 Regeneron Pharmaceuticals, Inc. Antibodies comprising chimeric constant domains
US10106610B2 (en) 2013-02-01 2018-10-23 Regeneron Pharmaceuticals, Inc. Antibodies comprising chimeric constant domains
US10968276B2 (en) 2013-03-12 2021-04-06 Xencor, Inc. Optimized anti-CD3 variable regions
US11814423B2 (en) 2013-03-15 2023-11-14 Xencor, Inc. Heterodimeric proteins
US10519242B2 (en) 2013-03-15 2019-12-31 Xencor, Inc. Targeting regulatory T cells with heterodimeric proteins
US11299554B2 (en) 2013-03-15 2022-04-12 Xencor, Inc. Heterodimeric proteins
US9605084B2 (en) 2013-03-15 2017-03-28 Xencor, Inc. Heterodimeric proteins
US10106624B2 (en) 2013-03-15 2018-10-23 Xencor, Inc. Heterodimeric proteins
US10287364B2 (en) 2013-03-15 2019-05-14 Xencor, Inc. Heterodimeric proteins
US10544187B2 (en) 2013-03-15 2020-01-28 Xencor, Inc. Targeting regulatory T cells with heterodimeric proteins
US10858417B2 (en) 2013-03-15 2020-12-08 Xencor, Inc. Heterodimeric proteins
EP3019622A4 (en) * 2013-07-12 2017-06-14 Zymeworks Inc. Bispecific cd3 and cd19 antigen binding contructs
US10323099B2 (en) 2013-10-11 2019-06-18 Hoffmann-La Roche Inc. Multispecific domain exchanged common variable light chain antibodies
EP3094737A4 (en) * 2014-01-15 2017-08-09 Zymeworks Inc. Bi-specific cd3 and cd19 antigen-binding constructs
KR102435081B1 (en) * 2014-03-19 2022-08-24 리제너론 파아마슈티컬스, 인크. Antibody compositions for tumor treatment
CN106459214A (en) * 2014-03-19 2017-02-22 瑞泽恩制药公司 Antibody compositions for tumor treatment
JP2017509643A (en) * 2014-03-19 2017-04-06 リジェネロン・ファーマシューティカルズ・インコーポレイテッドRegeneron Pharmaceuticals, Inc. Antibody composition for tumor therapy
JP2019196377A (en) * 2014-03-19 2019-11-14 リジェネロン・ファーマシューティカルズ・インコーポレイテッドRegeneron Pharmaceuticals, Inc. Antibody compositions for tumor treatment
CN113045669A (en) * 2014-03-19 2021-06-29 瑞泽恩制药公司 Methods and antibody compositions for tumor therapy
TWI701042B (en) * 2014-03-19 2020-08-11 美商再生元醫藥公司 Methods and antibody compositions for tumor treatment
KR20160127143A (en) * 2014-03-19 2016-11-02 리제너론 파아마슈티컬스, 인크. Antibody compositions for tumor treatment
TWI754319B (en) * 2014-03-19 2022-02-01 美商再生元醫藥公司 Methods and antibody compositions for tumor treatment
US10550193B2 (en) 2014-03-19 2020-02-04 Regeneron Pharmaceuticals, Inc. Methods and antibody compositions for tumor treatment
WO2015143079A1 (en) * 2014-03-19 2015-09-24 Regeneron Pharmaceuticals, Inc. Antibody compositions for tumor treatment
US11434300B2 (en) 2014-03-19 2022-09-06 Regeneron Pharmaceuticals, Inc. Methods and antibody compositions for tumor treatment
EP3594238A1 (en) * 2014-03-19 2020-01-15 Regeneron Pharmaceuticals, Inc. Antibody compositions for tumor treatment
EA035164B1 (en) * 2014-03-19 2020-05-08 Ридженерон Фармасьютикалз, Инк. Antibody compositions for tumor treatment
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
US11840579B2 (en) 2014-03-28 2023-12-12 Xencor, Inc. Bispecific antibodies that bind to CD38 and CD3
WO2015153765A1 (en) 2014-04-01 2015-10-08 Adimab, Llc Multispecific antibody analogs comprising a common light chain, and methods of their preparation and use
EP3825326A1 (en) 2014-04-01 2021-05-26 Adimab, LLC Multispecific antibody analogs comprising a common light chain, and methods of their preparation and use
EP4050026A1 (en) 2014-04-01 2022-08-31 Adimab, LLC Method of obtaining or identifying one or more common light chains for use in preparing a multispecific antibody
US11306156B2 (en) 2014-05-28 2022-04-19 Zymeworks Inc. Modified antigen binding polypeptide constructs and uses thereof
US9803029B2 (en) 2014-07-01 2017-10-31 Amphivena Therapeutics, Inc. Bispecific CD33 and CD3 binding proteins
US10626190B2 (en) 2014-07-01 2020-04-21 Amphivena Therapeutics, Inc. Bispecific CD33 and CD3 binding proteins
US9212225B1 (en) 2014-07-01 2015-12-15 Amphivena Therapeutics, Inc. Bispecific CD33 and CD3 binding proteins
WO2016004108A3 (en) * 2014-07-01 2016-04-07 Amphivena Therapeutics, Inc. Bispecific cd33 and cd3 binding proteins
US10662244B2 (en) 2014-11-17 2020-05-26 Regeneron Pharmaceuticals, Inc. Methods for tumor treatment using CD3XCD20 bispecific antibody
US10913803B2 (en) 2014-11-26 2021-02-09 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
AU2017216517B2 (en) * 2014-11-26 2019-09-26 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US11225528B2 (en) 2014-11-26 2022-01-18 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
US9850320B2 (en) 2014-11-26 2017-12-26 Xencor, Inc. Heterodimeric antibodies to CD3 X CD20
US11111315B2 (en) 2014-11-26 2021-09-07 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
US11945880B2 (en) 2014-11-26 2024-04-02 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
US11859011B2 (en) 2014-11-26 2024-01-02 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
AU2015353409B2 (en) * 2014-11-26 2019-05-09 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
US9856327B2 (en) 2014-11-26 2018-01-02 Xencor, Inc. Heterodimeric antibodies to CD3 X CD123
US10526417B2 (en) 2014-11-26 2020-01-07 Xencor, Inc. Heterodimeric antibodies that bind CD3 and CD38
CN107001482A (en) * 2014-12-03 2017-08-01 豪夫迈·罗氏有限公司 Multi-specificity antibody
US10633457B2 (en) 2014-12-03 2020-04-28 Hoffmann-La Roche Inc. Multispecific antibodies
CN107001482B (en) * 2014-12-03 2021-06-15 豪夫迈·罗氏有限公司 Multispecific antibodies
WO2016087416A1 (en) * 2014-12-03 2016-06-09 F. Hoffmann-La Roche Ag Multispecific antibodies
US10428155B2 (en) 2014-12-22 2019-10-01 Xencor, Inc. Trispecific antibodies
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
US11518807B2 (en) 2015-03-30 2022-12-06 Regeneron Pharmaceuticals, Inc. Heavy chain constant regions with reduced binding to Fc gamma receptors
US10556952B2 (en) 2015-03-30 2020-02-11 Regeneron Pharmaceuticals, Inc. Heavy chain constant regions with reduced binding to Fc gamma receptors
CN107847559B (en) * 2015-05-13 2022-07-01 埃博灵克斯股份有限公司 T cell recruitment polypeptides based on CD3 responsiveness
CN107847559A (en) * 2015-05-13 2018-03-27 埃博灵克斯股份有限公司 T cell based on CD3 reactivity raises polypeptide
US11753469B2 (en) 2015-05-29 2023-09-12 Anji Bruno, Llc Methods of using bispecific CD33 and CD3 binding proteins
US10738118B2 (en) 2015-05-29 2020-08-11 Amphivena Therapeutics, Inc. Methods of using bispecific CD33 and CD3 binding proteins
US11007267B2 (en) 2015-06-16 2021-05-18 Genentech, Inc. Anti-CD3 antibodies and methods of use
WO2016204966A1 (en) * 2015-06-16 2016-12-22 Genentech, Inc. Anti-cd3 antibodies and methods of use
JP2018526972A (en) * 2015-06-16 2018-09-20 ジェネンテック, インコーポレイテッド Anti-CD3 antibody and method of use
EP3916018A1 (en) * 2015-06-16 2021-12-01 Genentech, Inc. Anti-cd3 antibodies and methods of use
US11147886B2 (en) 2015-07-15 2021-10-19 Zymeworks Inc. Drug-conjugated bi-specific antigen-binding constructs
US11352426B2 (en) 2015-09-21 2022-06-07 Aptevo Research And Development Llc CD3 binding polypeptides
US11286300B2 (en) 2015-10-01 2022-03-29 Hoffmann-La Roche Inc. Humanized anti-human CD19 antibodies and methods of use
WO2017055314A1 (en) * 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Bispecific anti-cd19xcd3 t cell activating antigen binding molecules
EP3913000A1 (en) * 2015-10-02 2021-11-24 F. Hoffmann-La Roche AG Bispecific anti-cd19xcd3 t cell activating antigen binding molecules
US10227410B2 (en) 2015-12-07 2019-03-12 Xencor, Inc. Heterodimeric antibodies that bind CD3 and PSMA
US11623957B2 (en) 2015-12-07 2023-04-11 Xencor, Inc. Heterodimeric antibodies that bind CD3 and PSMA
US11168149B2 (en) 2015-12-16 2021-11-09 Jiangsu Alphamab Biopharmaceuticals Co., Ltd. Heterodimer molecule based on CH3 domain, and preparation method and use thereof
WO2017101828A1 (en) * 2015-12-16 2017-06-22 苏州康宁杰瑞生物科技有限公司 Heterodimer molecule based on ch3 structural domain, and preparation method therefor and use thereof
US10870701B2 (en) 2016-03-15 2020-12-22 Generon (Shanghai) Corporation Ltd. Multispecific fab fusion proteins and use thereof
US11236170B2 (en) 2016-06-14 2022-02-01 Xencor, Inc. Bispecific checkpoint inhibitor antibodies
US11492407B2 (en) 2016-06-14 2022-11-08 Xencor, Inc. Bispecific checkpoint inhibitor antibodies
US10787518B2 (en) 2016-06-14 2020-09-29 Xencor, Inc. Bispecific checkpoint inhibitor antibodies
US10316088B2 (en) 2016-06-28 2019-06-11 Xencor, Inc. Heterodimeric antibodies that bind somatostatin receptor 2
US11225521B2 (en) 2016-06-28 2022-01-18 Xencor, Inc. Heterodimeric antibodies that bind somatostatin receptor 2
EP3478830B1 (en) * 2016-07-01 2024-04-10 Resolve Therapeutics, LLC Optimized binuclease fusions and methods
WO2018005954A3 (en) * 2016-07-01 2018-03-29 Resolve Therapeutics, Llc Optimized binuclease fusions and methods
US10793632B2 (en) 2016-08-30 2020-10-06 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
US10501543B2 (en) 2016-10-14 2019-12-10 Xencor, Inc. IL15/IL15Rα heterodimeric Fc-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
JP7228515B2 (en) 2016-12-13 2023-02-24 デリニア,インコーポレーテッド Multivalent regulatory T cell regulatory factor
JP2020501550A (en) * 2016-12-13 2020-01-23 デリニア,インコーポレーテッド Multivalent regulatory T cell regulator
US11834506B2 (en) 2017-02-08 2023-12-05 Dragonfly Therapeutics, Inc. Multi-specific binding proteins that bind NKG2D, CD16, and a tumor-associated antigen for activation of natural killer cells and therapeutic uses thereof to treat cancer
US11884732B2 (en) 2017-02-20 2024-01-30 Dragonfly Therapeutics, Inc. Proteins binding HER2, NKG2D and CD16
US10279034B2 (en) 2017-04-11 2019-05-07 Eli Lilly And Company Anti-PD-L1-anti-TIM-3 bispecific antibodies
US11351251B2 (en) 2017-04-11 2022-06-07 Eli Lilly And Company Anti-PD-L1-anti-TIM-3 bispecific antibodies
EP3630169A4 (en) * 2017-05-23 2021-04-21 Dragonfly Therapeutics, Inc. A protein binding nkg2d, cd16 and a tumor-associated antigen
WO2018228442A1 (en) * 2017-06-14 2018-12-20 Dingfu Biotarget Co., Ltd. Proteinaceous heterodimer and use thereof
US11845806B2 (en) 2017-06-14 2023-12-19 Dingfu Biotarget Co., Ltd. Proteinaceous heterodimer and use 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
US11203640B2 (en) 2017-07-10 2021-12-21 Eli Lilly And Company Checkpoint inhibitor bispecific antibodies
US10981992B2 (en) 2017-11-08 2021-04-20 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
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
US11884733B2 (en) 2018-02-08 2024-01-30 Dragonfly Therapeutics, Inc. Antibody variable domains targeting the NKG2D receptor
US11939384B1 (en) 2018-02-08 2024-03-26 Dragonfly Therapeutics, Inc. Antibody variable domains targeting the NKG2D receptor
US11866498B2 (en) 2018-02-08 2024-01-09 Genentech, Inc. Bispecific antigen-binding molecules and methods of use
CN110218255B (en) * 2018-03-02 2020-12-04 广西医科大学 CD 3-resistant nano antibody CD3/Nb47 and preparation method and application thereof
CN110218255A (en) * 2018-03-02 2019-09-10 广西医科大学 Nano antibody CD3/Nb47 of AntiCD3 McAb and the preparation method and application thereof
US10982006B2 (en) 2018-04-04 2021-04-20 Xencor, Inc. Heterodimeric antibodies that bind fibroblast activation protein
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
WO2019237081A1 (en) * 2018-06-07 2019-12-12 Cullinan Management, Inc. Multi-specific binding proteins and methods of use thereof
WO2019244107A1 (en) * 2018-06-21 2019-12-26 Daiichi Sankyo Company, Limited Compositions including cd3 antigen binding fragments and uses thereof
US11590223B2 (en) 2018-08-31 2023-02-28 Regeneron Pharmaceuticals, Inc. Dosing strategy that mitigates cytokine release syndrome for therapeutic antibodies
US11807692B2 (en) 2018-09-25 2023-11-07 Harpoon Therapeutics, Inc. DLL3 binding proteins and methods of use
US11358999B2 (en) 2018-10-03 2022-06-14 Xencor, Inc. IL-12 heterodimeric Fc-fusion proteins
EP3873930A4 (en) * 2018-10-23 2022-12-07 Magenta Therapeutics, Inc. Fc silenced antibody drug conjugates (adcs) and uses thereof
WO2020135335A1 (en) * 2018-12-24 2020-07-02 Generon (Shanghai) Corporation Ltd. Multispecific antigen binding proteins capable of binding cd19 and cd3, and use thereof
WO2020132810A1 (en) * 2018-12-24 2020-07-02 Generon (Shanghai) Corporation Ltd. Multispecific antigen binding proteins capable of binding cd19 and cd3, and use thereof
US11472890B2 (en) 2019-03-01 2022-10-18 Xencor, Inc. Heterodimeric antibodies that bind ENPP3 and CD3
US11845799B2 (en) 2019-12-13 2023-12-19 Genentech, Inc. Anti-Ly6G6D antibodies and methods of use
WO2021224913A1 (en) * 2020-05-04 2021-11-11 Immunorizon Ltd. Precursor tri-specific antibody constructs and methods of use thereof
US11919956B2 (en) 2020-05-14 2024-03-05 Xencor, Inc. Heterodimeric antibodies that bind prostate specific membrane antigen (PSMA) and CD3
US11780920B2 (en) 2020-06-19 2023-10-10 Hoffmann-La Roche Inc. Antibodies binding to CD3 and CD19
US11919958B2 (en) 2020-08-19 2024-03-05 Xencor, Inc. Anti-CD28 compositions
US11591401B2 (en) 2020-08-19 2023-02-28 Xencor, Inc. Anti-CD28 compositions
US11976125B2 (en) 2021-01-15 2024-05-07 Harpoon Therapeutics, Inc. B cell maturation antigen binding proteins
EP4032910A1 (en) * 2021-01-22 2022-07-27 ETH Zurich Bispecific binding agent that binds to cd3 and a fluorophore
WO2022157352A1 (en) * 2021-01-22 2022-07-28 Eth Zurich Bispecific antibody that binds to cd3 and a fluorophore
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

Also Published As

Publication number Publication date
BR112015000798A2 (en) 2017-06-27
AU2013289883A1 (en) 2015-02-19
CA2878843A1 (en) 2014-01-16
US20190248897A1 (en) 2019-08-15
EP2872170A2 (en) 2015-05-20
EP2872170A4 (en) 2016-06-22
KR20150036606A (en) 2015-04-07
CN104640562A (en) 2015-05-20
WO2014012085A9 (en) 2015-03-19
JP2015528003A (en) 2015-09-24
AU2013289883B2 (en) 2018-11-01
US20210317212A1 (en) 2021-10-14
RU2650868C2 (en) 2018-04-17
WO2014012085A3 (en) 2014-03-27
RU2015102193A (en) 2016-08-27
HK1207575A1 (en) 2016-02-05
US20140154253A1 (en) 2014-06-05

Similar Documents

Publication Publication Date Title
US20210317212A1 (en) Bispecific Asymmetric Heterodimers Comprising Anti-CD3 Constructs
US11639376B2 (en) Constructs having a SIRP-α domain or variant thereof
US20160355588A1 (en) Bispecific CD3 and CD19 Antigen Binding Constructs
US20140072581A1 (en) Immunoglobulin Constructs Comprising Selective Pairing of the Light and Heavy Chains
DK2681245T3 (en) MULTIVALENT HEAT-MULTIMED SCAFFOLD DESIGN AND CONSTRUCTIONS
KR20210094588A (en) Antibodies that bind to CD3
EP2953972A1 (en) Method for the selection of antibodies against bcma
KR20230052293A (en) Anti-CD28 composition
EP2906237B1 (en) Multivalent heteromultimer scaffold design and constructs
EP2874652A2 (en) Immunoglobulin constructs comprising selective pairing of the light and heavy chains

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13816697

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 2878843

Country of ref document: CA

Ref document number: 2015521877

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2013816697

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20157003872

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2015102193

Country of ref document: RU

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2013289883

Country of ref document: AU

Date of ref document: 20130713

Kind code of ref document: A

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13816697

Country of ref document: EP

Kind code of ref document: A2

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112015000798

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112015000798

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20150113