WO2017210058A1 - Polythérapie avec des anticorps bispécifiques de redirection des lymphocytes t et des inhibiteurs de point de contrôle - Google Patents

Polythérapie avec des anticorps bispécifiques de redirection des lymphocytes t et des inhibiteurs de point de contrôle Download PDF

Info

Publication number
WO2017210058A1
WO2017210058A1 PCT/US2017/034250 US2017034250W WO2017210058A1 WO 2017210058 A1 WO2017210058 A1 WO 2017210058A1 US 2017034250 W US2017034250 W US 2017034250W WO 2017210058 A1 WO2017210058 A1 WO 2017210058A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
antibody
cell
interferon
virus
Prior art date
Application number
PCT/US2017/034250
Other languages
English (en)
Inventor
Edmund A. Rossi
Chien-Hsing Chang
David M. Goldenberg
Original Assignee
Ibc Pharmaceuticals, 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 claimed from US15/169,903 external-priority patent/US9670286B2/en
Application filed by Ibc Pharmaceuticals, Inc. filed Critical Ibc Pharmaceuticals, Inc.
Publication of WO2017210058A1 publication Critical patent/WO2017210058A1/fr

Links

Classifications

    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/212IFN-alpha
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39541Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against normal tissues, cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6853Carcino-embryonic antigens
    • 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
    • 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
    • C07K16/2803Immunoglobulins [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
    • 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
    • C07K16/2818Immunoglobulins [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 CD28 or CD152
    • 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
    • C07K16/2833Immunoglobulins [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 MHC-molecules, e.g. HLA-molecules
    • 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
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3007Carcino-embryonic Antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • 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
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing 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
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction

Definitions

  • the present invention concerns combinations of two or more agents for inducing an immune response to diseases, such as cancer or infectious disease.
  • agents may include: (i) leukocyte redirecting bispecific antibodies, (ii) antibody- drug conjugates, (iii) interferons such as such as interferon-a, interferon- ⁇ or interferon- ⁇ (most preferably interferon- ⁇ ), and/or (iv) checkpoint inhibitor antibodies.
  • Any combination of two or more such agents may be utilized in the subject methods and compositions.
  • the combinations may be administered simultaneously or sequentially.
  • Such combinations may comprise any two agents, any three agents, or all four types of agents.
  • the present invention concerns compositions and methods of use of novel anti-PDl antibodies.
  • such anti-PDl antibodies may be chimeric, humanized, or fully human.
  • the present invention concerns compositions and methods of use of leukocyte-redirecting complexes.
  • Leukocytes of use may include T cells, NK cells, monocytes, and neutrophils.
  • the complexes comprise bispecific antibodies with one binding site for an antigen expressed on a T cell, NK cell, monocyte, or neutrophil and another binding site for an antigen expressed on a diseased cell or pathogen.
  • the complexes are made as DOCK-AND-LOCK® complexes, in which the components are attached together using the binding interaction between
  • DDD dimerization and docking domain
  • PKA human protein kinase A
  • AD anchor domain
  • AKAPs A-kinase anchor proteins
  • the subject complexes may comprise one or more antibodies or antigen- binding antibody fragments that bind to an antigen expressed on T cells, NK cells, monocytes or neutrophils, such as ADAM 17, CD2, CD3, CD4, CD5, CD6, CD8, CDl la, CDl lb, CD14, CD16, CD25, CD28, CD30, CD32a, CD40, CD40L, CD44, CD45, CD56, CD57, CD64, CD69, CD74, CD89, CD90, CD 137, CD 177, CEACAM6, CEACAM8, HLA-DR alpha chain, KIR or SLC44A2, most preferably CD3 or CD 16, and one or more antibodies or antibody fragments that bind to an antigen on a target cell, such as CD 19, CD20, CD22, CD33, CD66e (CEACAM5), CEACAM6, CD74, EpCAM, HER2/neu, EGF receptor, Trop- 2, MUC5ac, or another tumor-associated antigen (TAA), or an antigen expressed on a
  • bispecific antibodies bsAbs
  • the predominant bispecific antibodies developed to date contain a first binding site specific to CD3 for T-cell recruitment and activation and a second binding site for a targeted disease- associated antigen, such as CD19 (Bassan, 2012, Blood 120:5094-95).
  • the bispecific antibody brings CD3 + T cells into direct contact with targeted disease cells and induces cell- mediated cytotoxicity (Bassan, 2012).
  • Anti-CD3 X anti-CD19 bispecific antibodies have been reported to produce a complete and durable molecular remission at very low
  • Leukocyte redirecting bsAbs are not limited to T cells.
  • the bispecific killer engagers comprising scFvs against the NK cell antigen CD 16 and a tumor associated antigen (e.g., CD19, CD22, CD33, CD74) have also shown potent anti-cancer activity (e.g., Miller, Hematology Soc Hematol Educ Pogram 2013 :247-53).
  • Other alternatives include trispecific killer engagers (TriKEs), such as anti-CD 16 x anti-CD 19 x anti-CD22 (Miller, 2013; Gleason et al., 2012, Mol Cancer Ther 11 :2674-84).
  • An anti-CD16 x anti-CD33 BiKE was used to treat AML and myelodysplastic syndrome (Miller, 2013; Wiernik et al., 2013, Clin Cancer Res 19:3844-55).
  • a CD16 x CD33 BiKE led to potent tumor cell killing and cytokine production by K cells.
  • Inhibition of ADAM17 enhanced the CD16 x CD33 BiKE response (Miller, 2013).
  • Bispecific antibodies can be produced by the quadroma method, which involves the fusion of two different hybridomas, each producing a monoclonal antibody recognizing a different antigenic site (Milstein and Cuello, Nature 1983; 305:537-540).
  • the fused hybridomas are capable of synthesizing two different heavy chains and two different light chains, which can associate randomly to give a heterogeneous population of 10 different antibody structures of which only one of them, amounting to 1/8 of the total antibody molecules, will be bispecific, and therefore must be further purified from the other forms.
  • Fused hybridomas are often less stable cytogenetically than the parent hybridomas, making the generation of a production cell line more problematic.
  • bispecific antibodies Another method for producing bispecific antibodies uses heterobifunctional cross- linkers to chemically tether two different monoclonal antibodies, so that the resulting hybrid conjugate will bind to two different targets (Staerz, et al. Nature 1985; 314:628-631; Perez, et al. Nature 1985; 316:354-356).
  • Bispecific antibodies generated by this approach are essentially heteroconjugates of two IgG molecules, which diffuse slowly into tissues and are rapidly removed from the circulation.
  • Bispecific antibodies can also be produced by reduction of each of two parental monoclonal antibodies to the respective half molecules, which are then mixed and allowed to reoxidize to obtain the hybrid structure (Staerz and Bevan.
  • Discrete V H and V L domains of antibodies produced by recombinant DNA technology may pair with each other to form a dimer (recombinant Fv fragment) with binding capability (U.S. Pat. No. 4,642,334).
  • a dimer recombinant Fv fragment
  • binding capability U.S. Pat. No. 4,642,334
  • Cognate V H and V L domains can be joined with a peptide linker of appropriate composition and length (usually consisting of more than 12 amino acid residues) to form a single-chain Fv (scFv) with binding activity.
  • scFv single-chain Fv
  • bispecific antibodies targeting CD3 and CD 19 are in clinical development.
  • DART® Dual-Affinity Re-Targeting
  • DART® Dual-Affinity Re-Targeting
  • BITE® and DART® exhibit fast blood clearance due to their small size (-55 kDa), which requires frequent administration to maintain therapeutic levels of the bispecific antibodies.
  • Interferons are critical role players in the antitumor and antimicrobial host defense, and have been extensively explored as therapeutic agents for cancer and infectious disease (Billiau et al., 2006, Cytokine Growth Factor Rev 17:381-409; Pestka et al., 2004, Immunol Rev 202:8-32).
  • type I and II interferons IFN- ⁇ / ⁇ and ⁇
  • their use in clinic settings have been limited because of the short circulation half-life, systemic toxicity, and suboptimal responses in patients (Pestka et al., 2004, Immunol Rev 202:8-32; Miller et al., 2009, Ann N Y Acad Sci 1182:69-79).
  • IFN-a.2 The therapeutic effectiveness of IFNs has been validated to date by the approval of IFN-a.2 for treating hairy cell leukemia, chronic myelogenous leukemia, malignant melanoma, follicular lymphoma, condylomata acuminata, AIDs-related Kaposi sarcoma, and chronic hepatitis B and C; IFN- ⁇ for treating multiple sclerosis; and IFN- ⁇ for treating chronic granulomatous disease and malignant osteopetrosis.
  • IFN- ⁇ for treating multiple sclerosis
  • IFN- ⁇ for treating chronic granulomatous disease and malignant osteopetrosis.
  • ADCs Antibody-drug conjugates
  • ADCs are a potent class of therapeutic constructs that allow targeted delivery of cytotoxic agents to target cells, such as cancer cells. Because of the targeting function, these compounds show a much higher therapeutic index compared to the same systemically delivered agents.
  • ADCs have been developed as intact antibodies or antibody fragments, such as scFvs. The antibody or fragment is linked to one or more copies of drug via a linker that is stable under physiological conditions, but that may be cleaved once inside the target cell.
  • ADCs approved for therapeutic use include gemtuzumab ozogamicin for AML (subsequently withdrawn from the market), brentuximab vedotin for ALCL and Hodgkin lymphoma, and trastuzumab emtansine for HER2-positive metastatic breast cancer (Verma et al., 2012, N Engl J Med 367-. ⁇ &3-91; Bross et al., 2001, Clin Cancer Res 7: 1490-96; Francisco et al., 2003, Blood 102: 1458-65).
  • ADCs inotuzumab ozogamicin (Pfizer), glembatumomab vedotin (Celldex Therapeutics), SAR3419 (Sanofi-Aventis), SAR56658 (Sanofi-Aventis), AMG-172 (Amgen), AMG-595 (Amgen), BAY-94-9343 (Bayer), BIIB015 (Biogen personal), BT062 (Biotest), SGN-75 (Seattle Genetics), SGN-CD19A (Seattle Genetics), vorsetuzumab mafodotin (Seattle Genetics), ABT-414 (Abb Vie), ASG-5ME (Agensys), ASG-22ME (Agensys), ASG-16M8F (Agensys), IMGN-529 (ImmunoGen), FMGN-853 (ImmunoGen), MDX-1203 (Medarex),
  • Immune checkpoints function as endogenous inhibitory pathways for immune system function that act to maintain self-tolerance and to modulate the duration and extent of immune response to antigenic stimulation (Pardoll, 2012). However, it appears that tumor tissues and possibly certain pathogens may co-opt the checkpoint system to reduce the effectiveness of host immune response, resulting in tumor growth and/or chronic infection (see, e.g., Pardoll, 2012, Nature Reviews Cancer 12:252-64; Nirschl & Drake, 2013, Clin Cancer Res 19:4917-24).
  • Checkpoint molecules include CTLA4 (cytotoxic T lymphocyte antigen-4), PDl (programmed cell death protein 1), PD-Ll (programmed cell death ligand 1), LAG-3 (lymphocyte activation gene-3), TIM-3 (T cell immunoglobulin and mucin protein-3) and several others (Pardoll, 2012, Nature Reviews Cancer 12:252-64; Nirschl & Drake, 2013, Clin Cancer Res 19:4917-24). Antibodies against several of the checkpoint proteins
  • CLA4, PDl, PD-Ll are in clinical trials and have shown unexpected efficacy againts tumors that were resistant to standard treatments.
  • the present invention relates to combination therapy with two or more agents selected from the group consisting of leukocyte-redirecting complexes, interferons, checkpoint inhibitor antibodies, and antibody-drug conjugates (ADCs).
  • the first three types of agents may be used to induce or enhance the immune response against disease- associated antigens, such as tumor-associated antigens (TAAs) or pathogen (microorganism)- expressed antigens, including HIV and other pathogenic viruses.
  • ADCs may be used in combination with any or all of the immunomodulatory agents to reduce tumor burden and enhance overall efficacy of treatment.
  • the complexes preferably are bispecific antibodies (bsAbs), with one binding site against a leukocyte expressed antigen and a second binding site that binds to a target antigen on a tumor cell or pathogen (i.e., micro-organism).
  • bsAbs bispecific antibodies
  • Exemplary T-cell antigens are selected from the group consisting of CD2, CD3, CD4, CD5, CD6, CD8, CD25, CD28, CD30, CD40, CD40L, CD44, CD45, CD69 and CD90.
  • Exemplary antigens expressed on NK cells are selected from the group consisting of CD8, CD16, CD56, CD57, ADAM 17, KIR and CD137.
  • Exemplary monocyte antigens are selected from the group consisting of CD74, HLA-DR alpha chain, CD14, CD16, CD64 and CD89.
  • Exemplary neutrophil antigens are selected from the group consisting of CEACAM6, CEACAM8, CD16b, CD32a, CD89, CD177, CDl la, CDl lb and SLC44A2.
  • the T-cell antigen is CD3, or the NK cell antigen is CD 16.
  • Target antigens for the second antibody may be selected from the group consisting of alpha-fetoprotein (AFP), a4 integrin, B7, carbonic anhydrase IX, complement factors Clq, Clr, Cls, C2a, C2b, C3, C3a, C3b, C4, C4a, C4b, C5a, C5aR, C5b, C5, C6, C7, C8, C9n, CCCL19, CCCL21, CD1, CDla, CD2, CD3R, CD4, CD5, CD8, CD11A, CD14, CD15, CD16, CD18, CD19, CD20, CD21, CD22, CD23, CD25, CD29, CD30, CD32b, CD33, CD37, CD38, CD40, CD40L, CD44, CD45, CD46, CD52, CD54, CD55, CD59, CD64, CD66a-e, CD67, CD70, CD74, CD79a, CD79b, CD80, CD83
  • An exemplary design for a leukocyte redirecting bsAb disclosed in the Examples below combined an anti-CD3 scFv with an anti-CD 19 F(ab) 2 to form a construct designated (19)-3s, which specifically targeted B cells.
  • Other bsAbs combining anti-CD3 with antibody fragments against other tumor-associated antigens, discussed in more detail below, are of use in targeted leukocyte immunotherapy of various solid tumors.
  • the advantages of this design include bivalent binding to tumor cells, a larger size (-130 kDa) to preclude rapid renal clearance, and potent leukocyte mediated cytotoxicity.
  • the bsAbs mediate the formation of immunological synapses between leukocytes and cognate target cells, induce leukocyte activation and proliferation in the presence of target cells, redirect potent leukocyte mediated killing of target cells in vitro and inhibit growth of human tumors in vivo.
  • a preferred embodiment concerns leukocyte redirecting bispecific antibodies produced as trivalent D L® complexes, with longer T 1/2 , better pharmacokinetic properties and increased in vivo stability.
  • Methods for production and use of DNL® complexes, comprising dimers of DDD moieties from human PKA regulatory subunits RIa, Rip, Rlla or RIip, bound to AD moieties from AKAPs, are well known (see, e.g., U.S. Patent Nos.
  • DNL® complexes comprising virtually any combination of effectors may be constructed and used.
  • the antibodies of use can be of various isotypes, preferably human IgGl, IgG2, IgG3 or IgG4, more preferably comprising human IgGl hinge and constant region sequences.
  • the antibodies or fragments thereof can be chimeric human-mouse, humanized (human framework and murine hypervariable (CDR) regions), or fully human, as well as variations thereof, such as half-IgG4 antibodies (referred to as "unibodies”), as described by van der Neut Kolfschoten et al. ⁇ Science 2007; 317: 1554-1557).
  • the antibodies or fragments thereof may be designed or selected to comprise human constant region sequences that belong to specific allotypes, which may result in reduced immunogenicity when administered to a human subject.
  • Preferred allotypes for administration include a non-Glml allotype (nGlml), such as Glm3, Glm3,l, Glm3,2 or Glm3,l,2. More preferably, the allotype is selected from the group consisting of the nGlml, Glm3, nGlml,2 and Km3 allotypes.
  • compositions and/or use of leukocyte- redirecting complexes in combination with one or more checkpoint inhibitor antibodies are antagonistic for checkpoint inhibitor function.
  • Many such antibodies are known in the art, such as pembrolizumab (MK-3475, Merck), nivolumab (BMS-936558, Bristol-Myers Squibb), pidilizumab (CT-011, CureTech Ltd.), AMP-224 (Merck), MDX- 1105 (Medarex), MEDI4736 (Medlmmune), atezolizumab (MPDL3280A) (Genentech), BMS-936559 (Bristol-Myers Squibb), ipilimumab (Bristol-Myers Squibb), durvalumab (Astrazeneca) and tremelimumab (Pfizer).
  • Anti-K R antibodies such as lirlumab (Innate Pharma) and IPH2101 (Innate Pharma) may perform similar functions in NK cells.
  • the checkpoint inhibitor antibody may be a novel chimeric or humanized anti-PDl antibody, as described in the Examples below. However, any known checkpoint inhibitor antibody may be used in combination with one or more of the other agents.
  • Interferons of use are known in the art and may include interferon-a, interferon- ⁇ , interferon- ⁇ , interferon- 2 or interferon ⁇ .
  • the interferon is interferon-a.
  • the subject interferon may be administered as free interferon, PEGylated interferon, an interferon fusion protein or interferon conjugated to an antibody.
  • one or more of the immunomodulatory agents discussed above may be used in combination with an antibody-drug conjugate (ADC).
  • ADCs are particularly effective for reducing tumor burden without significant systemic toxicity and may act to improve the effectiveness of the immune response induced by leukocyte retargeting bsAb, interferon and/or checkpoint inhibitor antibody.
  • Exemplary ADCs of use may include ADCs approved for therapeutic use, which include gemtuzumab ozogamicin for AML (subsequently withdrawn from the market), brentuximab vedotin for ALCL and Hodgkin lymphoma, and trastuzumab emtansine for HER2-positive metastatic breast cancer (Verma et al., 2012, NEnglJMed 367: 1783-91; Bross et al., 2001, Clin Cancer Res 7: 1490- 96; Francisco et al., 2003, Blood 102: 1458-65).
  • ADCs approved for therapeutic use include gemtuzumab ozogamicin for AML (subsequently withdrawn from the market), brentuximab vedotin for ALCL and Hodgkin lymphoma, and trastuzumab emtansine for HER2-positive metastatic breast cancer (Verma et al., 2012, NEnglJMed 367:
  • ADCs inotuzumab ozogamicin (Pfizer), glembatumomab vedotin (Celldex Therapeutics), SAR3419 (Sanofi-Aventis), SAR56658 (Sanofi-Aventis), AMG-172 (Amgen), AMG-595 (Amgen), BAY-94-9343 (Bayer), BIIB015 (Biogen personal), BT062 (Biotest), SGN-75 (Seattle Genetics), SGN-CD19A (Seattle Genetics), vorsetuzumab mafodotin (Seattle Genetics), ABT-414 (Abb Vie), ASG-5ME (Agensys), ASG-22ME (Agensys), ASG-16M8F (Agensys), IMGN-529 (ImmunoGen), IMGN-853 (ImmunoGen), MDX-1203 (Medarex
  • an ADC is used in combination with an immunomodulator
  • the ADC is administered prior to the immunomodulator.
  • the subject combination therapy may be of use for treating cancer. It is anticipated that any type of tumor and any type of tumor antigen may be targeted. Exemplary types of cancers that may be targeted include acute lymphoblastic leukemia, acute myelogenous leukemia, biliary cancer, B-cell leukemia, B-cell lymphoma, biliary cancer, bone cancer, brain cancer, breast cancer, triple-negative breast cancer, cervical cancer, Burkitt lymphoma, chronic lymphocytic leukemia, chronic myelogenous leukemia, colorectal cancer, endometrial cancer, esophageal cancer, gall bladder cancer, gastric cancer, gastrointestinal tract cancer, glioma, hairy cell leukemia, head and neck cancer, Hodgkin's lymphoma, liver cancer, lung cancer, medullary thyroid cancer, melanoma, multiple myeloma, ovarian cancer, non-Hodgkin's lymphoma, pancreatic cancer,
  • Tumor-associated antigens that may be targeted by leukocyte redirecting bsAbs and/or by ADCs include, but are not limited to, alpha-fetoprotein (AFP), a4 integrin, B7, carbonic anhydrase IX, complement factors Clq, Clr, Cls, C2a, C2b, C3, C3a, C3b, C4, C4a, C4b, C5a, C5aR, C5b, C5, C6, C7, C8, C9n, CCCL19, CCCL21, CD1, CDla, CD2, CD3R, CD4, CD5, CD8, CD11A, CD14, CD15, CD16, CD18, CD19, CD20, CD21, CD22, CD23, CD25, CD29, CD30, CD32b, CD33, CD37, CD38, CD40, CD40L, CD44, CD45, CD46, CD52, CD54, CD55, CD59, CD64, CD66a-e, CD67
  • Exemplary antibodies that may be used for cancer therapy include, but are not limited to, hA19 (anti-CD19, U.S. Patent No. 7, 109,304), hRl (anti-IGF-lR, U.S. Patent No. 9,441,043), hPAM4 (anti-MUC5ac, U.S. Patent No. 7,282,567), hA20 (anti-CD20, U.S. Patent No. 7,151, 164), MMMU31 (anti-AFP, U.S. Patent No. 7,300,655), hLLl (anti-CD74, U.S. Patent No. 7,312,318), hLL2 (anti-CD22, U.S. Patent No.
  • the subject combination therapy may include combinations with multiple antibodies that are immunostimulatory, anti-tumor or anti-infectious agent.
  • Alternative antibodies that may be used for treatment of various disease states include, but are not limited to, abciximab (anti-glycoprotein Ilb/IIIa), alemtuzumab (anti- CD52), bevacizumab (anti-VEGF), cetuximab (anti-EGFR), gemtuzumab (anti-CD33), ibritumomab (anti-CD20), panitumumab (anti-EGFR), rituximab (anti-CD20), tositumomab (anti-CD20), trastuzumab (anti-ErbB2), lambrolizumab (anti-PDl receptor), nivolumab (anti- PDl receptor), ipilimumab (anti-CTLA4), abagovomab (anti-CA-125), adecatumumab (anti- EpCAM), atlizumab (anti-IL-6 receptor), benralizumab (anti-CD 125
  • Patent 8,333,971 Ab 75, Ab 76, Ab 77 (Paulik et al., 1999, Biochem Pharmacol 58: 1781-90), as well as the anti-HIV antibodies described and sold by Polymun (Vienna, Austria), also described in U.S. Patent 5,831,034, U.S. Patent 5,911,989, and Vcelar et al., AIDS 2007; 21(16):2161-2170 and Joos et al., Antimicrob. Agents Chemother. 2006; 50(5): 1773-9.
  • the subject combination therapy may be of use to treat subjects infected with pathogenic organisms, such as bacteria, viruses or fungi.
  • pathogenic organisms such as bacteria, viruses or fungi.
  • fungi include Microsporum, Trichophyton, Epidermophyton, Sporothrix schenckii, Cryptococcus neoformans, Coccidioides immitis, Histoplasma capsulatum, Blastomyces dermatitidis or Candida albican.
  • viruses include human immunodeficiency virus (HIV), herpes virus, cytomegalovirus, rabies virus, influenza virus, human papilloma virus, hepatitis B virus, hepatitis C virus, Sendai virus, feline leukemia virus, Reo virus, polio virus, human serum parvo-like virus, simian virus 40, respiratory syncytial virus, mouse mammary tumor virus, Varicella-Zoster virus, dengue virus, rubella virus, measles virus, adenovirus, human T-cell leukemia viruses, Epstein-Barr virus, murine leukemia virus, mumps virus, vesicular stomatitis virus, Sindbis virus, lymphocytic choriomeningitis virus or blue tongue virus.
  • HCV human immunodeficiency virus
  • herpes virus cytomegalovirus
  • rabies virus influenza virus
  • human papilloma virus hepatitis B virus
  • Exemplary bacteria include Bacillus anthracis, Streptococcus agalactiae, Legionella pneumophilia, Streptococcus pyogenes, Escherichia coli, Neisseria gonorrhoeae, Neisseria meningitidis, Pneumococcus spp., Hemophilis influenzae B, Treponema pallidum, Lyme disease spirochetes, Pseudomonas aeruginosa, Mycobacterium leprae, Brucella abortus, Mycobacterium tuberculosis or a Mycoplasma.
  • Exemplary use of ADCs against infectious agents are disclosed in Johannson et al. (2006, AIDS 20: 1911-15) and Chang et al., 2012, PLos One 7:e41235).
  • Known antibodies against pathogens include, but are not limited to, P4D10 (anti- HIV), CR6261 (anti-influenza), exbivirumab (anti -hepatitis B), felvizumab (anti -respiratory syncytial virus), foravirumab (anti-rabies virus), motavizumab (anti-respiratory syncytial virus), palivizumab (anti-respiratory syncytial virus), panobacumab (anti-Pseudomonas), rafivirumab (anti-rabies virus), regavirumab (anti-cytomegalovirus), sevirumab (anti- cytomegalovirus), tivirumab (anti-hepatitis B), and urtoxazumab (anti-i. coli).
  • Immunomodulators may include, but are not limited to, a cytokine, a chemokine, a stem cell growth factor, a lymphotoxin, an hematopoietic factor, a colony stimulating factor (CSF), erythropoietin, thrombopoietin, tumor necrosis factor-a (TNF), TNF- ⁇ , granulocyte-colony stimulating factor (G-CSF), granulocyte macrophage-colony stimulating factor (GM-CSF), interferon-a, interferon- ⁇ , interferon- ⁇ , interferon- ⁇ , interferon- ⁇ , stem cell growth factor designated "SI factor", human growth hormone, N-methionyl human growth hormone, bovine growth hormone, parathyroid hormone, thyroxine, insulin, proinsulin, relaxin, prorelaxin, follicle stimulating hormone (FSH), thyroid stimulating hormone (TSH
  • a leukocyte- redirecting bispecific antibody or antibody fragment may be attached to an immunomodulator, such as a cytokine.
  • an immunomodulator such as a cytokine.
  • Cytokine complexes are disclosed, for example, in U.S. Patent Nos. 7,906,118 and 8,034,3522, the Examples section of each incorporated herein by reference.
  • T-cell binding component of the leukocyte redirecting bsAb binds to the CD3 antigen
  • other antigens expressed on effector T cells are known and may be targeted by the leukocyte redirecting complex.
  • Exemplary T-cell antigens include, but are not limited to, CD2, CD3, CD4, CD5, CD6, CD8, CD25, CD28, CD30, CD40, CD40L, CD44, CD45, CD69 and CD90.
  • exemplary antigens may be selected from CD8, CD16, CD56, CD57, ADAM 17, and CD137 for K cells; CD74, HLA-DR alpha chain, CD14, CD16, CD64 and CD89 for monocytes; and CEACAM6, CEACAM8, CD16b, CD32a, CD89, CD177, CDl la, CDl lb and SLC44A2 for neutrophils.
  • FIG. 1 Schematic diagram of formation of DOCK- AND-LOCK® complex comprising anti-CD 19 F(ab) 2 x anti-CD3 scFv.
  • FIG. 2A Immune synapse formation between Daudi Burkitt lymphoma and T cells, mediated by (19)-3s. Freshly isolated T cells were combined with Daudi cells at an E:T ratio of 2.5: 1. Cells were treated with 0, 1 or 5 ⁇ / ⁇ . of (19)-3s for 30 min at room temperature prior to analysis by flow cytometry. Anti-CD20-FITC and anti-CD7-APC were used to identify Daudi and T cells, respectively. Co-binding was indicated as the % of CD207CD7 + events. After treatment with (19)-3s, 45.5% of flow events were CD20/CD7 dual-positive, indicating synapsed Daudi and T cells.
  • FIG. 2B Conditions were as in FIG. 2(A), except for the absence of (19)-3s antibody. Compared to FIG. 2(A), only 2% of flow events were CD20/CD7 dual-positive without antibody.
  • FIG. 2C Addition of (19)-3s resulted in association of >90% of the Daudi with T cells.
  • FIG. 3A Jurkat (T cells) and Daudi (B cells) were combined at a 1 : 1 ratio, treated with 0.1 ⁇ g/mL (19)-3s for 30 minutes and stained with anti-CD20-FITC, prior to analysis by fluorescence microscopy.
  • FIG. 3B Jurkat (T cells) and Daudi (B cells) were combined at a 1 : 1 ratio, treated with 0.1 ⁇ g/mL (19)-3s for 30 minutes and stained with anti-CD20-FITC and anti-CD3-PE, prior to analysis by fluorescence microscopy.
  • FIG. 3C The merged image of FIG. 3 A and 3B reveals synapse formation between green-stained Daudi and red-stained Jurkat cells.
  • FIG. 3D Synapse formation was not evident in the absence of (19)-3s.
  • FIG. 4 Dose response analysis of (19)-3s mediated cell-to-cell association of Daudi and Jurkat cells as a function of increasing concentrations of (19)-3s.
  • FIG. 5A Comparison of cell-to-cell association mediated by BITE® and DARTTM.
  • FIG. 5B Comparison of cell-to-cell association mediated by (19)-3s.
  • FIG. 6A Synapse formation between T cells and Capan-1 pancreatic cancer cells mediated by (19)-3s control bsAb. CFSE-labeled Capan-1 cells were coincubated with
  • FIG. 6B Synapse formation between T cells and Capan-1 pancreatic cancer cells mediated by (Ml)-3s MUC5AC bsAb. CFSE-labeled Capan-1 cells were coincubated with PKH26-labeled Jurkat in the presence of the bsAb.
  • FIG. 6C Synapse formation between T cells and Capan-1 pancreatic cancer cells mediated by (El)-3s Trop-2 targeting bsAb. CFSE-labeled Capan-1 cells were coincubated with PKH26-labeled Jurkat in the presence of the bsAb.
  • FIG. 7A T-cell activation by (19)-3s. Upregulation of CD69 expression is an early event in T-cell activation. Daudi cells combined with PBMCs were treated overnight with the indicated antibodies, and stained with anti-CD3-PE and anti-CD69-APC, prior to analysis by flow cytometry. CD69 expression was evaluated following gating of T cells by forward vs. side scattering and anti-CD3 staining. Combination of Daudi cells with an equal number of PBMCs resulted in 1.6% CD69+ T cells. Addition of 3 ng/mL (19)-3s induced 27% CD69+ T cells.
  • FIG. 7B T-cell activation by (19)-3s. Daudi cells combined with purified T cells were treated overnight with the indicated antibodies, and stained with anti-CD3-PE and anti- CD69-APC, prior to analysis by flow cytometry. CD69 expression was evaluated following gating of T cells by forward vs. side scattering and anti-CD3 staining. Treatment of Daudi and purified T cells with (Ml)-3s or hA19-Fab-DDD2 did not increase the number of CD69+ T cells ( ⁇ 4%), compared to the untreated cell mixture. Alternatively, (19)-3s induced robust T-cell activation, producing 80% CD69+ cells.
  • FIG. 7C T-cell activation by (19)-3s.
  • Purified T cells alone were treated overnight with the indicated antibodies, and stained with anti-CD3-PE and anti-CD69-APC, prior to analysis by flow cytometry.
  • CD69 expression was evaluated following gating of T cells by forward vs. side scattering and anti-CD3 staining. Without the addition of Daudi (target) cells, (19)-3s did not induce CD69 expression and T-cell activation.
  • FIG. 8A Induction of T-cell proliferation by (19)-3s.
  • PBMCs were incubated with 3 nM or 30 pM of (19)-3s, compared to IL-2/PHA positive control and (14)-3s (non-target- binding control).
  • FIG. 8B Induction of T-cell proliferation by (19)-3s. T cell proliferation was not observed in PBMCs depleted of B cells, indicating that target cells (B cells) are required for T-cell activation and proliferation.
  • FIG. 9A In vitro cytotoxicity of (19)-3s T-cell redirecting bsAbs. Dose-response curves for cytotoxicity to Nalm-6, Raji, Ramos and Namalwa cancer cells were determined for the (19)-3s DNL® bsAb complex.
  • FIG. 9B In vitro cytotoxicity of (19)-3s T-cell redirecting bsAbs. Dose-response curves for cytotoxicity to Nalm-6, Raji, Ramos and Namalwa cancer cells were determined for the (14)-3s (non-targeting) DNL® bsAb complex. [056] FIG. 9C. Consistent results were observed using PBMCs, or T cells, obtained from two different donors and Nalm-6 cancer cells.
  • FIG. 10A In vitro cytotoxicity of (20)-3 s, (22)-3 s and (C2)-3 s T-cell redirecting bsAbs. Dose-response curves were determined for cytotoxicity to Namalwa cells induced by (20)-3s, (22)-3s and (C2)-3s T-cell redirecting bsAbs.
  • FIG. 10B In vitro cytotoxicity of (20)-3s, (22)-3s and (C2)-3s T-cell redirecting bsAbs. Dose-response curves were determined for cytotoxicity to Jeko cells induced by (20)- 3s, (22)-3s and (C2)-3s T-cell redirecting bsAbs.
  • FIG. IOC In vitro cytotoxicity of (20)-3 s, (22)-3 s and (C2)-3 s T-cell redirecting bsAbs. Dose-response curves were determined for cytotoxicity to Daudi cells induced by (20)-3s, (22)-3s and (C2)-3s T-cell redirecting bsAbs.
  • FIG. 11 A In vitro cytotoxicity of T-cell redirecting bsAbs in solid tumor cell lines. Dose-response curves were determined for cytotoxicity to the LS174T colon adenocarcinoma cell line for the (14)-3s bsAb, compared to non-targeting (19)-3s bsAb.
  • FIG. 11B In vitro cytotoxicity of T-cell redirecting bsAbs in solid tumor cell lines. Dose-response curves were determined for cytotoxicity to the Capan-1 pancreatic
  • FIG. llC In vitro cytotoxicity of T-cell redirecting bsAbs in solid tumor cell lines. Dose-response curves were determined for cytotoxicity to the NCI-N87 gastric carcinoma cell line for the (El)-3s and (15)-3s bsAbs, compared to non-targeting (19)-3s bsAb.
  • FIG. 12 Summary of in vitro cytotoxicity data for T-cell redirecting bsAbs in cancer cell lines.
  • FIG. 13A In vivo retargeting of Raji lymphoma xenografts using (19)-3s bsAb.
  • FIG. 13B In vivo retargeting of Raji lymphoma xenografts using (19)-3s bsAb.
  • NOD/SCID mice bearing Raji Burkitt lymphoma (1 x 10 6 cells) xenografts, reconstituted with human PBMCs (5 x 10 6 cells) and treated with (19)-3s for only 1 week, administered as indicated by the arrows. Cells were treated with a single dose of 130 ⁇ g.
  • FIG. 13C In vivo retargeting of Raji lymphoma xenografts using (19)-3s bsAb.
  • NOD/SCID mice bearing Raji Burkitt lymphoma (1 x 10 6 cells) xenografts, reconstituted with human PBMCs (5 x 10 6 cells) and treated with (19)-3s for only 1 week, administered as indicated by the arrows. Cells were treated 3x with 43 ⁇ g per dose.
  • FIG. 13D In vivo retargeting of Raji lymphoma xenografts using (19)-3s bsAb.
  • NOD/SCID mice bearing Raji Burkitt lymphoma (1 x 10 6 cells) xenografts, reconstituted with human PBMCs (5 x 10 6 cells) and treated with (19)-3s for only 1 week, administered indicated by the arrows. Cells were treated 5x with 26 ⁇ g per dose.
  • FIG. 14 A Effect of repeated dosing on in vivo retargeting of Raji lymphoma xenografts using (19)-3s bsAb. NOD/SCID mouse xenografts were prepared as indicated the legend to FIG. 13. The (19)-3s was administered as indicated by the arrows. FIG. 14A shows untreated control.
  • FIG. 14B Effect of repeated dosing on in vivo retargeting of Raji lymphoma xenografts using (19)-3s bsAb.
  • the (19)-3s was administered as indicated by the arrows.
  • FIG. 14C Effect of repeated dosing on in vivo retargeting of Raji lymphoma xenografts using (19)-3s bsAb.
  • the (19)-3s was administered as indicated by the arrows.
  • FIG. 14D Effect of repeated dosing on in vivo retargeting of Raji lymphoma xenografts using (19)-3s bsAb.
  • the (19)-3s was administered as indicated by the arrows.
  • FIG. 14E Effect of repeated dosing on in vivo retargeting of Raji lymphoma xenografts using (19)-3s bsAb.
  • the (19)-3s was administered as indicated by the arrows.
  • FIG. 14F Effect of repeated dosing on in vivo retargeting of Raji lymphoma xenografts using (19)-3s bsAb.
  • the (19)-3s was administered as indicated by the arrows.
  • FIG. 15A In vivo efficacy of T-cell retargeting bsAbs in solid tumor xenografts.
  • NOD/SCID mouse xenografts were prepared with LS174T colon adenocarcinoma. Mice were administered T cells only without bsAb.
  • FIG. 15B In vivo efficacy of T-cell retargeting bsAbs in solid tumor xenografts.
  • NOD/SCID mouse xenografts were prepared with LS174T colon adenocarcinoma. Mice w treated with (El)-3s bsAb as indicated.
  • FIG. 15C In vivo efficacy of T-cell retargeting bsAbs in solid tumor xenografts.
  • NOD/SCID mouse xenografts were prepared with Capan-1 pancreatic carcinoma. Mice w administered PBMCs only without bsAb.
  • FIG. 15D In vivo efficacy of T-cell retargeting bsAbs in solid tumor xenografts.
  • NOD/SCID mouse xenografts were prepared with Capan-1 pancreatic carcinoma. Mice were treated with (14)-3s bsAb as indicated.
  • FIG. 16A In vivo inhibition of tumor growth by (El)-3s DNL® complex in the presence or absence of interferon- ⁇ .
  • Capan-1 pancreatic carcinoma xenografts in NOD/SCID mice were treated with anti-Trop-2 x anti-CD3 bsAb with or without added interferon- ⁇ .
  • the interferon-a was added in the form of a Trop-2 targeting DNL® complex.
  • FIG. 16B In vivo inhibition of tumor growth by (El)-3s DNL® complex in the presence or absence of interferon- ⁇ .
  • Capan-1 pancreatic carcinoma xenografts in NOD/SCID mice were treated with anti-Trop-2 x anti-CD3 bsAb with or without added interferon- ⁇ .
  • the interferon-a was added as the commercially available PEGASYS® (peginterferon alfa-2a).
  • FIG. 17 Survival curves for NOD/SCID mice treated with (El)-3s with or without interferon- ⁇ . Controls were untreated or treated with interferon- ⁇ alone.
  • FIG. 18 In vivo inhibition of tumor growth by (El)-3s DNL® complex in the presence or absence of interferon- ⁇ , compared to TF12 control.
  • Capan-1 pancreatic carcinoma xenografts in NOD/SCID mice were treated with anti-Trop-2 x anti-CD3 bsAb with or without added interferon- ⁇ , added as PEGASYS®, compared to untreated control, TF12 control or PEGASYS® alone.
  • FIG. 19 Survival curves for NOD/SCID mice treated with (El)-3s with or without interferon- ⁇ (PEGASYS®). Controls were untreated or treated with PEGASYS® alone or TF12 alone.
  • FIG. 20 In vivo inhibition of tumor growth by (El)-3s DNL® complex in the presence or absence of interferon- ⁇ , compared to TF12 control.
  • NCI-N87 human gastric cancer xenografts in NOD/SCID mice were treated with anti-Trop-2 x anti-CD3 bsAb with or without added interferon- ⁇ , added as PEGASYS®, compared to untreated control, TF12 control or PEGASYS® alone.
  • FIG. 21 Survival curves for NOD/SCID mice with NCI-N87 gastric cancer xenografts treated with (El)-3s with or without interferon- ⁇ (PEGASYS®). Controls were untreated or treated with PEGASYS® alone or TF12 alone.
  • FIG. 22 Schematic representation of the nascent El -3 polypeptide.
  • LP leader peptide that is removed in mature protein
  • VH heavy chain variable domain
  • VK kappa light chain variable domain
  • LI linker peptide 1
  • L2 linker peptide 2
  • L3 linker pepide 3
  • 6H hexa-histidine
  • FIG. 23A Ex vivo T cell redirected killing of BxPC3 human pancreatic cancer solid tumor cell line.
  • FIG. 23B Ex vivo T cell redirected killing of Capan-1 human pancreatic cancer solid tumor cell line.
  • FIG. 23C Ex vivo T cell redirected killing of NCI-N87 human gastric cancer solid tumor cell line.
  • FIG. 24 In vivo T cell redirected therapy of NCI-N87 gastric carcinoma in NOD- SCID mice.
  • FIG. 25 Binding of 5G9.G1.B 11 to PDl expressed on activated Jurkat cells.
  • Jurkat T cells (5 mL at 5 x 10 5 cells/mL) were not stimulated or stimulated with PHA (1 ⁇ g/mL), PMA (50 ng/mL), or both PHA (1 ⁇ g/mL) and PMA (50 ng/mL) for 48 h and analyzed for the expression of CD69 by flow cytometry.
  • Expression of CD69 is a marker for activated T cells; EH12 is a positive control for anti-PDl
  • FIG. 26 Quantitation of IL-2 by ELISA. A notable increase in IL-2 produced from T cells in a mixed lymphocyte assay was observed for 5G9.G1.B11 dose-independently.
  • FIG. 27A The amino acid sequence determined for the VK (SEQ ID NO:40) of 5G9.G1.B 11, with the 3 CDRs underlined.
  • FIG. 27B The amino acid sequence determined for the VH (SEQ ID NO:41) of 5G9.G1.B 11, with the 3 CDRs underlined.
  • FIG. 27C CDR sequences of 5G9.G1.B 11 were, for the heavy chain,
  • GFAFSSNDMS (SEQ ID NO:42), TI S GGGINT YYPD S VKG (SEQ ID NO:43) and
  • RSNYAWFAY (SEQ ID NO:44) and for the light chain, RASESVDTYGISFMN (SEQ ID NO:45), PNQGS (SEQ ID NO:46) and QQSKEVPWT (SEQ ID NO:47).
  • FIG. 28A Binding of 2G9 to recombinant PD 1 -Fc.
  • FIG. 28B Binding of 2G9 to SpEFX-2Dl, but not SpESX cells.
  • the SpESX cell line which does not express PDl, was transfected with human PDl to obtain SpESX-2Dl, which overexpresses PDl .
  • FIG. 29 Blockade of biotinlyated PDl binding to PD-L1 on MDA-MB-231 by anti- PDl antibodies.
  • CDR sequences are underlined.
  • Framework residues (FRs) where the human amino acid residue is substituted with the corresponding parental mouse amino acid residue are highlighted in bold font.
  • FIG. 30 Amino acid sequence of light (SEQ ID NO:48) and heavy (SEQ ID NO:49) chains of humanized anti-PDl antibody (hPDl).
  • FIG. 31 D E sequence encoding light chain (SEQ ID NO:50) and heavy chain (SEQ ID NO:51) of humanized anti-PDl antibody.
  • FIG. 32 Binding of humanized vs. chimeric anti-PDl to recombinant human PD1- His.
  • FIG. 33 Binding of humanized vs. chimeric anti-PDl to cells transfected with human PD1 (2D1 cells).
  • FIG. 34 Combination therapy with anti-Trop-2 x anti-CD3 bsAb and humanized or chimeric anti-PDl antibody.
  • a “therapeutic agent” is an atom, molecule, or compound that is useful in the treatment of a disease.
  • therapeutic agents include antibodies, antibody fragments, peptides, drugs, toxins, enzymes, nucleases, hormones, immunomodulators, antisense oligonucleotides, small interfering RNA (siRNA), chelators, boron compounds, photoactive agents, dyes, and radioisotopes.
  • an "antibody” as used herein refers to a full-length (i.e., naturally occurring or formed by normal immunoglobulin gene fragment recombinatorial processes) immunoglobulin molecule (e.g., an IgG antibody) or an immunologically active (i.e., specifically binding) portion of an immunoglobulin molecule, like an antibody fragment.
  • An “antibody” includes monoclonal, polyclonal, bispecific, multispecific, murine, chimeric, humanized and human antibodies.
  • a "naked antibody” is an antibody or antigen binding fragment thereof that is not attached to a therapeutic or diagnostic agent.
  • the Fc portion of an intact naked antibody can provide effector functions, such as complement fixation and ADCC (see, e.g., Markrides, Pharmacol Rev 50:59-87, 1998).
  • Other mechanisms by which naked antibodies induce cell death may include apoptosis. (Vaswani and Hamilton, Ann Allergy Asthma Immunol 81 : 105- 119, 1998.)
  • an “antibody fragment” is a portion of an intact antibody such as F(ab') 2 , F(ab) 2 , Fab', Fab, Fv, scFv, dAb and the like. Regardless of structure, an antibody fragment binds with the same antigen that is recognized by the full-length antibody.
  • antibody fragments include isolated fragments consisting of the variable regions, such as the "Fv” fragments consisting of the variable regions of the heavy and light chains or recombinant single chain polypeptide molecules in which light and heavy variable regions are connected by a peptide linker ("scFv proteins").
  • Single-chain antibodies often abbreviated as “scFv” consist of a polypeptide chain that comprises both a V H and a V L domain which interact to form an antigen- binding site.
  • the V H and V L domains are usually linked by a peptide of 1 to 25 amino acid residues.
  • Antibody fragments also include diabodies, triabodies and single domain antibodies (dAb).
  • a "chimeric antibody” is a recombinant protein that contains the variable domains including the complementarity determining regions (CDRs) of an antibody derived from one species, preferably a rodent antibody, while the constant domains of the antibody molecule are derived from those of a human antibody.
  • the constant domains of the chimeric antibody may be derived from that of other species, such as a cat or dog.
  • a "humanized antibody” is a recombinant protein in which the CDRs from an antibody from one species; e.g., a rodent antibody, are transferred from the heavy and light variable chains of the rodent antibody into human heavy and light variable domains, including human framework region (FR) sequences.
  • the constant domains of the antibody molecule are derived from those of a human antibody.
  • FR amino acid residues from the parent (e.g., murine) antibody may be substituted for the corresponding human FR residues.
  • a "human antibody” is an antibody obtained from transgenic mice that have been genetically engineered to produce specific human antibodies in response to antigenic challenge.
  • elements of the human heavy and light chain locus are introduced into strains of mice derived from embryonic stem cell lines that contain targeted disruptions of the endogenous heavy chain and light chain loci.
  • the transgenic mice can synthesize human antibodies specific for human antigens, and the mice can be used to produce human antibody-secreting hybridomas. Methods for obtaining human antibodies from transgenic mice are described by Green et al., Nature Genet. 7: 13 (1994), Lonberg et al., Nature 368 : 856 (1994), and Taylor et al., Int. Immun. 6: 579 (1994).
  • a human antibody also can be constructed by genetic or chromosomal transfection methods, as well as phage display technology, all of which are known in the art. (See, e.g., McCafferty et al., 1990, Nature 348 :552-553 for the production of human antibodies and fragments thereof in vitro, from immunoglobulin variable domain gene repertoires from unimmunized donors).
  • antibody variable domain genes are cloned in-frame into either a major or minor coat protein gene of a filamentous bacteriophage, and displayed as functional antibody fragments on the surface of the phage particle.
  • the filamentous particle contains a single-stranded DNA copy of the phage genome, selections based on the functional properties of the antibody also result in selection of the gene encoding the antibody exhibiting those properties. In this way, the phage mimics some of the properties of the B cell.
  • Phage display can be performed in a variety of formats, for their review, see, e.g. Johnson and Chiswell, Current Opinion in Structural Biology 3 :5564-571 (1993).
  • Human antibodies may also be generated by in vitro activated B cells. (See, U.S. Pat. Nos. 5,567,610 and 5,229,275).
  • antibody fusion protein is a recombinantly produced antigen-binding molecule in which an antibody or antibody fragment is linked to another protein or peptide, such as the same or different antibody or antibody fragment or a DDD or AD peptide.
  • the fusion protein may comprise a single antibody component, a multivalent or multispecific combination of different antibody components or multiple copies of the same antibody component.
  • the fusion protein may additionally comprise an antibody or an antibody fragment and a therapeutic agent. Examples of therapeutic agents suitable for such fusion proteins include immunomodulators and toxins.
  • One preferred toxin comprises a ribonuclease (RNase), preferably a recombinant RNase.
  • a preferred immunomodulator might be an interferon, such as interferon-a, interferon- ⁇ or interferon- ⁇ .
  • a "multispecific antibody” is an antibody that can bind simultaneously to at least two targets that are of different structure, e.g., two different antigens, two different epitopes on the same antigen, or a hapten and/or an antigen or epitope.
  • a “multivalent antibody” is an antibody that can bind simultaneously to at least two targets that are of the same or different structure. Valency indicates how many binding arms or sites the antibody has to a single antigen or epitope; i.e., monovalent, bivalent, trivalent or multivalent. The multivalency of the antibody means that it can take advantage of multiple interactions in binding to an antigen, thus increasing the avidity of binding to the antigen.
  • Specificity indicates how many antigens or epitopes an antibody is able to bind; i.e., monospecific, bispecific, trispecific, multispecific.
  • a natural antibody e.g., an IgG
  • Multispecific, multivalent antibodies are constructs that have more than one binding site of different specificity.
  • bispecific antibody is an antibody that can bind simultaneously to two targets which are of different structure.
  • Bispecific antibodies bsAb and bispecific antibody fragments (bsFab) may have at least one arm that specifically binds to, for example, a T cell, an K cell, a monocyte or a neutrophil, and at least one other arm that specifically binds to an antigen produced by or associated with a diseased cell, tissue, organ or pathogen, for example a tumor-associated antigen.
  • bsAb bispecific antibody fragments
  • bsFab bispecific antibody fragments
  • a variety of bispecific antibodies can be produced using molecular engineering.
  • An antibody preparation, or a composition described herein, is said to be administered in a "therapeutically effective amount" if the amount administered is physiologically significant.
  • An agent is physiologically significant if its presence results in a detectable change in the physiology of a recipient subject.
  • an antibody preparation is physiologically significant if its presence invokes an antitumor response or mitigates the signs and symptoms of an infectious disease state.
  • a physiologically significant effect could also be the evocation of a humoral and/or cellular immune response in the recipient subject leading to growth inhibition or death of target cells.
  • T-cell antigens include CD2, CD3, CD4, CD5, CD6, CD8, CD25, CD28, CD30, CD40, CD40L, CD44, CD45, CD69 and CD90.
  • Other exemplary antigens may be selected from CD8, CD16, CD56, CD57, ADAM17, and CD137 for K cells; CD74, HLA- DR alpha chain, CD 14, CD 16, CD64 and CD89 for monocytes; and CEACAM6,
  • the anti-T-cell antibody binds to CD3, or the anti- K antibody binds to CD 16.
  • TAAs tumor-associated antigens
  • An exemplary preferred TAA is CD 19.
  • bispecific anti-CD3 x anti-CD 19 antibodies are known in the art and presently in clinical development, such as BITE® (Bispecific T-cell Engager) (e.g., Nagorsen et al., 2009, Leukemia & Lymphoma 50:886-91; Amann et al., 2009, J Immunother 32:453- 64; Baeuerle and Reinhardt, 2009, Cancer Res 69:4941-44) and DART® (see, e.g., Moore et al., 2011, Blood 117:4542-51; Veri et al., 2010, Arthritis Rheum 62: 1933-43).
  • BITE® Bispecific T-cell Engager
  • DART® see, e.g., Moore et al., 2011, Blood 117:4542-51; Veri et al., 2010, Arthritis Rheum 62: 1933-43.
  • Blinatumomab is a BITE® antibody comprising V H and V L domains of anti-CD3 and anti-CD 19 antibody fragments, connected with a 5-amino acid linker and expressed as a single polypeptide chain that anneals to itself to form the antigen-binding sites. It is thought that blinatumomab acts by bringing the T-cell-specific CD3 and B-cell specific CD 19 antigens into close proximity, to initiate a T-cell cytotoxic response against the juxtaposed B cells, which does not require T-cell specificity to the cancer cells (e.g., Portell et al., 2013, Clin Pharmacol 5(Suppl 1): 5- 11).
  • blinatumomab induced 50% cell lysis of MEC-1 cells at a concentration of 10 pg/mL (Topp et al., 2012, Blood 120:5185-87; Bassan et al., 2012, Blood 120:5094-95).
  • the anti-CD19 portion of blinatumomab was derived from the HD37 hybridoma (see, e.g., U.S. Patent No. 7,575,923, the Examples section of which is incorporated herein by reference), which is publicly available (e.g., Santa Cruz Biotechnology Cat. No. sc-18894).
  • the anti-CD3 portion of blinatumomab was derived from the TR66 hybridoma (U.S. Patent No. 7,575,923; Traunecker et al., 1991, EMBO J. 10:3655-59), also publicly available (e.g., Enzo Life Sciences, catalog No. ALX-804-822-C100).
  • amino acid sequence of the anti-CD3 moiety used as part of a DNL® complex, is as disclosed below in SEQ ID NO:22 to SEQ ID NO:27.
  • the person of ordinary skill will realize that any known anti-CD3 antibody may be utilized in the claimed methods and compositions.
  • the antibody moieties of use are humanized or human.
  • a variety of antibodies against CD 19 that may be used in the claimed methods and compositions are publicly known and/or commercially available, such as from Santa Cruz Biotechnology (catalog Nos. sc-390244, sc-373897, sc-18894, sc-18896, etc.); ABCAM® (catalog Nos. ab25232, abl34114, abl40981, abl255, etc.); ABBIOTECTM (catalog Nos. 252262, 252248, 250585, 251063, etc.) and many other vendors.
  • the anti-CD 19 antibody moiety is a humanized A19 antibody, comprising the light chain CDR sequences CDR1 KASQSVDYDGDSYLN (SEQ ID NO: 14); CDR2 DASNLVS (SEQ ID NO: 15); and CDR3 QQSTEDPWT (SEQ ID NO: 16) and the heavy chain CDR sequences CDR1 SYWMN (SEQ ID NO: 17); CDR2 QIWPGDGDTNYNGKFKG (SEQ ID NO: 18) and CDR3 RETTTVGRYYYAMDY (SEQ ID NO: 19).
  • DART® anti-CD3 x anti-CD 19 bispecific antibodies
  • DART® which also incorporates the anti-CD19 Fv sequences of HD37 and the anti-CD3 Fv sequences of TR66
  • DART® bispecific antibodies were more potent at inducing B cell lysis than single-chain, bispecific antibodies (BITE®) bearing identical anti- CD ⁇ and anti-CD3 variable region sequences, with EC 50 values in the pg/mL range (Moore et al., 2011).
  • any known anti-CD3 x anti-CD 19 bispecific antibody may be used to induce an immune response against disease-associated cells or pathogens.
  • Catumaxomab is an anti-CD3 x anti-EpCAM bispecific antibody that has been approved in Europe for treatment of malignant ascites associated with metastasizing cancer (Chames & Baty, 2009, MAbs 1 :539-47).
  • catumaxomab was able to kill tumor cells at a concentration range of 10 pM and was reported to lead to total eradication of melanoma tumors (Chames & Baty, 2009).
  • Human clinical trials with ovarian cancer patients with malignant ascites also showed a statistically significant efficacy (Chames & Baty, 2009).
  • anti-tumor bsAbs is not limited to anti-CD3 x anti-CD 19, but has also included anti-HER2 x anti-CD64 (MDX- 210, MDX-H210), anti-EGFR x anti-CD64 (MDX-447), anti-CD30 x anti-CD16 (HRS- 3/A9), anti-HER2 x anti-CD3 (Her2Bi), anti-CD20 x anti-CD3 (CD20Bi, Bi20), anti-EpCAM x anti-CD3 (catumaxomab, MT110), anti-HER2 x anti-CD3 (ertumaxomab), and anti-NG2 x anti-CD28 (rM28) (Chames & Baty, 2009).
  • anti-HER2 x anti-CD64 MDX- 210, MDX-H210
  • anti-EGFR x anti-CD64 MDX-447
  • anti-CD30 x anti-CD16 HRS- 3/A9
  • an anti-CD3 x anti-CD 19 bispecific antibody or other leukocyte redirecting bsAb is made as a DNL® construct, as disclosed in Example 1 below.
  • the person of ordinary skill will realize that the subject leukocyte redirecting bispecific antibodies are not limited to anti-CD3 x anti-CD 19 constructs, but may comprise antibodies against any known disease-associated antigens attached to an anti-CD3 antibody moiety. Alternatively, antibodies against other T-cell antigens besides CD3, or other antigens expressed on NK cells, monocytes or neutrophils may also be used.
  • T-cell antigens include, but are not limited to, CD2, CD3, CD4, CD5, CD6, CD8, CD25, CD28, CD30, CD40, CD40L, CD44, CD45, CD69 and CD90.
  • Other exemplary antigens may be selected from CD8, CD16, CD56, CD57, ADAM17, KIR and CD137 for K cells; CD74, HLA-DR alpha chain, CD 14, CD 16, CD64 and CD89 for monocytes; and CEACAM6, CEACAM8, CD16b, CD32a, CD89, CD177, CDl la, CDl lb and SLC44A2 for neutrophils.
  • Antibodies against each of the leukocyte antigens are publicly known and/or publicly available (see, e.g., ABCAM® catalog numbers abl31276, abl39266, ab8360, ab51312, ab846, abl33616, ab75877, abl33255, abl09217, ab93278, abl7147, abl 15851, abl28955, abl3463, ab85986; Santa Cruz Biotechnology catalog numbers sc-46683, sc-59047; Enzo Life Sciences, Inc. catalog number ALX-805-037-C100; Sino Biological Inc. catalog numbers 12211-RP02, 11150-R074; Millipore catalog numbers 04-1102, 04-1102,
  • anti-leukocyte antibodies were publicly available and could have been used in the subject leukocyte redirecting bsAbs. As discussed below, numerous antibodies against a wide variety of disease-associated antigens were publicly known and/or commercially available and could have been used in the subject leukocyte redirecting bispecific antibodies.
  • Other exemplary leukocyte redirecting bsAbs of potential use include FBTA05 (anti-CD20 x anti-CD3) and TRBS07 (anti-GD2 x anti-CD3).
  • leukocyte redirecting bsAbs, antibody-drug conjugates and/or checkpoint inhibitor antibodies may be used in combination with one or more interferons, such as interferon-a, interferon- ⁇ or interferon- ⁇ .
  • interferons such as interferon-a, interferon- ⁇ or interferon- ⁇ .
  • Human interferons are well known in the art and the amino acid sequences of human interferons may be readily obtained from public databases (e.g., GenBank Accession Nos. AAA52716.1; AAA52724;
  • Human interferons may also be commercially obtained from a variety of vendors (e.g., Cell Signaling Technology, Inc., Danvers, MA; Genentech, South San Francisco, CA; EMD Millipore, Billerica, MA).
  • Interferon- ⁇ has been reported to have anti-tumor activity in animal models of cancer (Ferrantini et al., 1994, J Immunol 153 :4604-15) and human cancer patients
  • IFNa can exert a variety of direct anti-tumor effects, including down-regulation of oncogenes, up-regulation of tumor suppressors, enhancement of immune recognition via increased expression of tumor surface MHC class I proteins, potentiation of apoptosis, and sensitization to chemotherapeutic agents (Gutterman et al., 1994, PNAS USA 91 : 1198-205; Matarrese et al., 2002, Am J Pathol 160: 1507-20; Mecchia et al., 2000, Gene Ther 7: 167-79; Sabaawy et al., 1999, IntJ Oncol 14: 1143-51; Takaoka et al, 2003, Nature 424:516-23).
  • IFNa can have a direct and potent anti-proliferative effect through activation of STAT1 (Grimley et al., 1998 Blood 91 :3017-27).
  • Interferon-a2b has been conjugated to anti-tumor antibodies, such as the hL243 anti-HLA-DR antibody and depletes lymphoma and myeloma cells in vitro and in vivo (Rossi et al., 2011, Blood 118: 1877-84).
  • IFNa can inhibit angiogenesis (Sidky and Borden, 1987, Cancer Res 47:5155-61) and stimulate host immune cells, which may be vital to the overall antitumor response but has been largely under-appreciated (Belardelli et al., 1996, Immunol Today 17:369-72).
  • IFNa has a pleiotropic influence on immune responses through effects on myeloid cells (Raefsky et al, 1985, J Immunol 135:2507-12; Luft et al, 1998, J Immunol 161 : 1947-53), T-cells (Carrero et al, 2006, J Exp Med 203 :933-40; Pilling et al., 1999, Eur J Immunol 29: 1041-50), and B-cells (Le et al, 2001, Immunity 14:461-70).
  • IFNa induces the rapid differentiation and activation of dendritic cells (Belardelli et al, 2004, Cancer Res 64:6827-30; Paquette et al., 1998, J Leukoc Biol 64:358-67; Santini et al., 2000, J Exp Med 191 : 1777-88) and enhances the cytotoxicity, migration, cytokine production and antibody-dependent cellular cytotoxicity (ADCC) of NK cells (Biron et al., 1999, Ann Rev Immunol 17: 189-220; Brunda et al. 1984, Cancer Res 44:597-601).
  • ADCC antibody-dependent cellular cytotoxicity
  • Interferon- ⁇ has been reported to be efficacious for therapy of a variety of solid tumors. Patients treated with 6 million units of IFN- ⁇ twice a week for 36 months showed a decreased recurrence of hepatocellular carcinoma after complete resection or ablation of the primary tumor in patients with HCV-related liver cancer (Ikeda et al., 2000, Hepatology 32:228-32). Gene therapy with interferon- ⁇ induced apoptosis of glioma, melanoma and renal cell carcinoma (Yoshida et al., 2004, Cancer Sci 95:858-65). Endogenous IFN- ⁇ has been observed to inhibit tumor growth by inhibiting angiogenesis in vivo (Jablonska et al., 2010, J Clin Invest. 120: 1151-64.)
  • IFN-a2 The therapeutic effectiveness of IFNs has been validated to date by the approval of IFN-a2 for treating hairy cell leukemia, chronic myelogenous leukemia, malignant melanoma, follicular lymphoma, condylomata acuminata, AIDs-related Kaposi sarcoma, and chronic hepatitis B and C; IFN- ⁇ for treating multiple sclerosis; and IFN- ⁇ for treating chronic granulomatous disease and malignant osteopetrosis.
  • IFN- ⁇ for treating hairy cell leukemia, chronic myelogenous leukemia, malignant melanoma, follicular lymphoma, condylomata acuminata, AIDs-related Kaposi sarcoma, and chronic hepatitis B and C
  • IFN- ⁇ for treating multiple sclerosis
  • IFN- ⁇ for treating chronic granulomatous disease and malignant osteopetrosis.
  • Interferons are critical role players in the antitumor and antimicrobial host defense, and have been extensively explored as therapeutic agents for cancer and infectious disease (Billiau et al., 2006, Cytokine Growth Factor Rev 17:381-409; Pestka et al., 2004, Immunol Rev 202:8-32).
  • type I and II interferons IFN- ⁇ / ⁇ and ⁇
  • their use in clinic settings have been limited because of the short circulation half-life, systemic toxicity, and suboptimal responses in patients (Pestka et al., 2004, Immunol Rev 202:8-32; Miller et al., 2009, Ann N Y Acad Sci 1182:69-79).
  • IFN- s designated as type III interferons, are a newly described group of cytokines that consist of IFN- ⁇ , 2, 3 (also referred to as interleukin-29, 28 A, and 28B, respectively), that are genetically encoded by three different genes located on chromosome 19 (Kotenko et al., 2003, Nat Immunol 4:69-77; Sheppard et al., 2003, Nat Immunol 4:63-8).
  • IFN- 2 and - ⁇ 3 are is highly homologous, with 96% amino acid identity, while IFN- ⁇ shares approximately 81% homology with IFN- 2 and - ⁇ 3 (Sheppard et al., 2003, Nat Immunol 4:63-8).
  • IFN- s activate signal transduction via the JAK/STAT pathway similar to that induced by type I IFN, including the activation of JAKl and TYK2 kinases, the phosphorylation of STAT proteins, and the activation of the transcription complex of IFN- stimulated gene factor 3 (ISGF3) (Witte et al., 2010, Cytokine Growth Factor Rev 21 :237-51; Zhou et al., 2007, J Virol 81 :7749-58).
  • IGF3 IFN- stimulated gene factor 3
  • IFN- ⁇ / ⁇ signals through two extensively expressed type I interferon receptors, and the resulting systemic toxicity associated with IFN- ⁇ / ⁇
  • IFN- s signal through a heterodimeric receptor complex consisting of unique IFN- ⁇ receptor 1 (IFN- Rl) and IL-10 receptor 2 (IL-10R2).
  • IFN- Rl unique IFN- ⁇ receptor 1
  • IL-10R2 IL-10 receptor 2
  • IFN- ⁇ has a very restricted expression pattern with the highest levels in epithelial cells, melanocytes, and hepatocytes, and the lowest level in primary central nervous system (CNS) cells.
  • CNS central nervous system
  • Blood immune system cells express high levels of a short IFN- ⁇ receptor splice variant (sIFN- Rl) that inhibits IFN- ⁇ action.
  • sIFN- Rl short IFN- ⁇ receptor splice variant
  • IFN-a and IFN- ⁇ induce expression of a common set of ISGs (interferon-stimulated genes) in hepatocytes, unlike IFN-a, administration of IFN- ⁇ did not induce STAT activation or ISG expression in purified lymphocytes or monocytes (Dickensheets et al., 2013, J Leukoc Biol. 93, published online 12/20/12). It was suggested that IFN- ⁇ may be superior to IFN-a for treatment of chronic HCV infection, as it is less likely to induce leukopenias that are often associated with IFN-a therapy (Dickensheets et al., 2013).
  • IFN- s display structural features similar to IL-10-related cytokines, but functionally possess type I IFN-like anti-viral and anti-proliferative activity (Witte et al., 2009, Genes Immun 10:702-14; Ank et al., 2006, J Virol 80:4501-9; Robek et al., 2005, J Virol 79:3851- 4).
  • IFN- ⁇ and - ⁇ 2 have been demonstrated to reduce viral replication or the cytopathic effect of various viruses, including DNA viruses (hepatitis B virus (Robek et al., 2005, J Virol 79:3851-4, Doyle et al., 2006, Hepatology 44:896-906) and herpes simplex virus 2 (Ank et al., 2008, J Immunol 180:2474-85)), ss (+) RNA viruses (EMCV; Sheppard et al., 2003, Nat Immunol 4:63-8) and hepatitis C virus (Robek et al., 2005, J Virol 79:3851-4, Doyle et al., 2006, Hepatology 44:896-906; Marcello et al., 2006, Gastroenterol 131 : 1887- 98; Pagliaccetti et al., 2008, J Biol Chem 283 :30079-89), ss (-) RNA viruses (ves
  • IFN- 3 has been identified from genetic studies as a key cytokine in HCV infection (Ge et al., 2009, Nature 461 : 399-401), and has also shown potent activity against EMCV (Dellgren et al., 2009, Genes Immun 10: 125-31).
  • IFN- s The anti-proliferative activity of IFN- s has been established in several human cancer cell lines, including neuroendocrine carcinoma BON1 (Zitzmann et al., 2006, Biochem Biophys Res Commun 344: 1334-41), glioblastoma LN319 (Meager et al., 2005, Cytokine 31 : 109-18), immortalized keratinocyte HaCaT (Maher et al., 2008, Cancer Biol Ther 7: 1109- 15), melanoma F01 (Guenterberg et al., 2010, Mol Cancer Ther 9:510-20), and esophageal carcinoma TE-11 (Li et al., 2010, Eur J Cancer 46: 180-90).
  • IFN- s induce both tumor apoptosis and destruction through innate and adaptive immune responses, suggesting that local delivery of IFN- ⁇ might be a useful adjunctive strategy in the treatment of human malignancies (Numasaki et al., 2007, J Immunol 178:5086-98).
  • a Fab-linked interferon- ⁇ was demonstrated to have potent anti-tumor and anti-viral activity in targeted cells (Liu et al., 2013, PLoS One 8:e63940).
  • PEGylated IFN- ⁇ (PEG-IFN- ⁇ ) has been provisionally used for patients with chronic hepatitis C virus infection.
  • PEG-IFN- ⁇ has been provisionally used for patients with chronic hepatitis C virus infection.
  • antiviral activity was observed at all dose levels (0.5-3.0 ⁇ g/kg), and viral load reduced 2.3 to 4.0 logs when PEG-IFN- ⁇ was administrated to genotype 1 HCV patients who relapsed after IFN-a therapy (Muir et al., 2010, Hepatology 52:822-32).
  • the subject leukocyte redirecting bispecific antibodies, ADCs and/or checkpoint inhibitor mAbs may be used in combination with one or more interferons, such as interferon-a, interferon- ⁇ , interferon- ⁇ , interferon- 2, or interferon ⁇ .
  • interferons such as interferon-a, interferon- ⁇ , interferon- ⁇ , interferon- 2, or interferon ⁇ .
  • the interferon may be administered prior to, concurrently with, or after the other agent.
  • the interferon may be either conjugated to or separate from the other agent.
  • checkpoint inhibitors In contrast to the majority of anti-cancer agents, checkpoint inhibitors do not target tumor cells directly, but rather target lymphocyte receptors or their ligands in order to enhance the endogenous antitumor activity of the immune system. (Pardoll, 2012, Nature Reviews Cancer 12:252-264) Because such antibodies act primarily by regulating the immune response to diseased cells, tissues or pathogens, they may be used in combination with other therapeutic modalities, such as the subject leukocyte redirecting bispecific antibodies, ADCs and/or interferons to enhance the anti-tumor effect of such agents. Because checkpoint activation may also be associated with chronic infections (Nirschl & Drake, 2013, Clin Cancer Res 19:4917-24), such combination therapies may also be of use to treat infectious disease.
  • PDl Programmed cell death protein 1
  • CD279 encodes a cell surface membrane protein of the immunoglobulin superfamily, which is expressed in B cells and NK cells (Shinohara et al., 1995, Genomics 23 :704-6; Blank et al., 2007, Cancer Immunol Immunother 56:739-45; Finger et al., 1997, Gene 197: 177-87; Pardoll, 2012, Nature Reviews Cancer 12:252-264).
  • the major role of PDl is to limit the activity of T cells in peripheral tissues during inflammation in response to infection, as well as to limit autoimmunity (Pardoll, 2012, Nature Reviews Cancer 12:252-264).
  • PDl expression is induced in activated T cells and binding of PDl to one of its endogenous ligants acts to inhibit T-cell activation by inhibiting stimulatory kinases (Pardoll, 2012, Nature Reviews Cancer 12:252-264). PDl also acts to inhibit the TCR "stop signal" (Pardoll, 2012, Nature Reviews Cancer 12:252-264). PDl is highly expressed on T reg cells and may increase their proliferation in the presence of ligand (Pardoll, 2012, Nature Reviews Cancer 12:252-264).
  • Anti-PDl antibodies have been used for treatment of melanoma, non-small-cell lung cancer, bladder cancer, prostate cancer, colorectal cancer, head and neck cancer, triple- negative breast cancer, leukemia, lymphoma and renal cell cancer (Topalian et al., 2012, N EnglJMed 366:2443-54; Lipson et al., 2013, Clin Cancer Res 19:462-8; Berger et al., 2008, Clin Cancer Res 14:3044-51; Gildener-Leapman et al., 2013, Oral Oncol 49: 1089-96;
  • anti-PDl antibodies include lambrolizumab (MK-3475, MERCK), nivolumab (BMS-936558, BRISTOL-MYERS SQUIBB), AMP-224 (MERCK), and pidilizumab (CT-011, CURETECH LTD.).
  • Anti-PDl antibodies are commercially available, for example from ABCAM® (AB137132), BIOLEGE D® (EH12.2H7, RMP1-14) and AFFYMETRIX EBIOSCIENCE (J105, Jl 16, MIH4).
  • Another anti-PDl antibody of use is defined by the heavy chain CDR sequences GFAFSS DMS (SEQ ID NO:42), TISGGGINTYYPDSVKG (SEQ ID NO:43) and RSNYAWFAY (SEQ ID NO:44) and the light chain CDR sequences RASESVDTYGISFMN (SEQ ID NO:45), PNQGS (SEQ ID NO:46) and QQSKEVPWT (SEQ ID NO:47).
  • the antibody may be used in chimeric, humanized, or fully human form, as discussed below.
  • P-L1 Programmed cell death 1 ligand 1
  • CD274 and B7-H1 are ligands for PD1, found on activated T cells, B cells, myeloid cells and macrophages.
  • anti -tumor therapies have focused on anti-PD-Ll antibodies.
  • the complex of PD1 and PD-L1 inhibits proliferation of CD8+ T cells and reduces the immune response (Topalian et al., 2012, NEngl J Med 366:2443-54; Brahmer et al., 2012, NEngJMed 366:2455-65).
  • Anti-PD-Ll antibodies have been used for treatment of non-small cell lung cancer, melanoma, colorectal cancer, renal-cell cancer, pancreatic cancer, gastric cancer, ovarian cancer, breast cancer, and hematologic malignancies (Brahmer et al., N EngJMed 366:2455-65; Ott et al., 2013, Clin Cancer Res 19:5300-9; Radvanyi et al., 2013, Clin Cancer Res 19:5541; Menzies & Long, 2013, Ther Adv Med Oncol 5:278-85; Berger et al., 2008, Clin Cancer Res 14: 13044-51).
  • anti-PD-Ll antibodies include MDX-1105 (MEDAREX), MEDI4736 (MEDFMMUNE) MPDL3280A (GE ENTECH) and BMS-936559 (BRISTOL-MYERS SQUIBB).
  • Anti-PD-Ll antibodies are also commercially available, for example from
  • CTLA4 Cytotoxic T-lymphocyte antigen 4
  • CD 152 Cytotoxic T-lymphocyte antigen 4
  • CTLA4 acts to inhibit T-cell activation and is reported to inhibit helper T-cell activity and enhance regulatory T-cell immunosuppressive activity (Pardoll, 2012, Nature Reviews Cancer 12:252- 264).
  • CTLA4 inhibits T cell activation by outcompeting CD28 in binding to CD80 and CD86, as well as actively delivering inhibitor signals to the T cell (Pardoll, 2012, Nature Reviews Cancer 12:252-264).
  • Anti-CTL4A antibodies have been used in clinical trials for treatment of melanoma, prostate cancer, small cell lung cancer, non-small cell lung cancer (Robert & Ghiringhelli, 2009, Oncologist 14:848-61; Ott et al., 2013, Clin Cancer Res 19:5300; Weber, 2007, Oncologist 12:864-72; Wada et al., 2013, J Transl Med 11 :89).
  • a significant feature of anti-CTL4A is the kinetics of anti-tumor effect, with a lag period of up to 6 months after initial treatment required for physiologic response (Pardoll, 2012, Nature Reviews Cancer 12:252-264). In some cases, tumors may actually increase in size after treatment initiation, before a reduction is seen (Pardoll, 2012, Nature Reviews Cancer 12:252-264).
  • anti-CTLA4 antibodies include ipilimumab (Bristol-Myers Squibb) and tremelimumab (PFIZER).
  • Anti-CTLA4 antibodies are commercially available, for example from ABCAM® (AB134090), SINO BIOLOGICAL INC. (11159-H03H, 11159-H08H), and THERMO SCIENTIFIC PIERCE (PA5-29572, PA5-23967, PA5-26465, MA1-12205, MA1- 35914).
  • Ipilimumab has recently received FDA approval for treatment of metastatic melanoma (Wada et al., 2013, J Transl Med 11 :89).
  • an immune checkpoint inhibitor antibody may preferably be administered at about 0.3-10 mg/kg, or the maximum tolerated dose, administered about every three weeks or about every six weeks.
  • the checkpoint inhibitor antibody may be administered by an escalating dosage regimen including administering a first dosage at about 3 mg/kg, a second dosage at about 5 mg/kg, and a third dosage at about 9 mg/kg.
  • the escalating dosage regimen includes administering a first dosage of checkpoint inhibitor antibody at about 5 mg/kg and a second dosage at about 9 mg/kg.
  • Another stepwise escalating dosage regimen may include administering a first dosage of checkpoint inhibitor antibody about 3 mg/kg, a second dosage of about 3 mg/kg, a third dosage of about 5 mg/kg, a fourth dosage of about 5 mg/kg, and a fifth dosage of about 9 mg/kg.
  • a stepwise escalating dosage regimen may include administering a first dosage of 5 mg/kg, a second dosage of 5 mg/kg, and a third dosage of 9 mg/kg.
  • Exemplary reported dosages of checkpoint inhibitor mAbs include 3 mg/kg ipilimumab administered every three weeks for four doses; 10 mg/kg ipilimumab every three weeks for eight cycles; 10 mg/kg every three weeks for four cycles then every 12 weeks for a total of three years; 10 mg/kg MK-3475 every two or every three weeks; 2 mg/kg MK-3475 every three weeks; 15 mg/kg tremilimumab every three months; 0.1, 0.3, 1, 3 or 10 mg/kg nivolumab every two weeks for up to 96 weeks; 0.3, 1, 3, or 10 mg/kg BMS-936559 every two weeks for up to 96 weeks (Kyi & Postow, October 23, 2013, FEBS Lett [Epub ahead of print]; Callahan & Wolchok, 2013, JLeukoc Biol 94:41-53).
  • agents that stimulate immune response to tumors and/or pathogens may be used in combination with leukocyte redirecting bispecific antibodies alone or in further combination with an interferon, such as interferon-a, and/or an antibody-drug conjugate for improved cancer therapy.
  • an interferon such as interferon-a, and/or an antibody-drug conjugate for improved cancer therapy.
  • co-stimulatory pathway modulators that may be used in combination include, but are not limited to, agatolimod, belatacept, blinatumomab, CD40 ligand, anti-B7-l antibody, anti-B7-2 antibody, anti-B7-H4 antibody, AG4263, eritoran, anti-OX40 antibody, ISF-154, and SGN-70; B7-1, B7-2, ICAM-1, ICAM- 2, ICAM-3, CD48, LFA-3, CD30 ligand, CD40 ligand, heat stable antigen, B7h, OX40 ligand, LIGHT, CD70 and CD24.
  • anti-KIR antibodies may also be used in combination with leukocyte-redirecting bsAbs, interferons, ADCs and/or checkpoint inhibitor antibodies.
  • NK cells mediate anti-tumor and anti-infectious agent activity by spontaneous cytotoxicity and by ADCC when activated by antibodies (Kohrt et al., 2013, Blood, [Epub ahead of print 12/10/13]). The degree of cytotoxic response is determined by a balance of inhibitory and activating signals received by the NK cells (Kohrt et al., 2013).
  • KIR immunoglobulin-like receptor
  • Anti-KIR antibodies such as lirlumab (Innate Pharma) and IPH2101 (Innate Pharma) have demonstrated anti-tumor activity in multiple myeloma (Benson et al., 2012, Blood 120:4324-33).
  • anti-KIR antibodies prevent the tolerogenic interaction of NK cells with target cells and augments the NK cell cytotoxic response to tumor cells (Kohrt et al., 2013).
  • anti-KIR antibodies In vivo, in combination with rituximab (anti-CD20), anti-KIR antibodies at a dose of 0.5 mg/kg induced enhanced NK cell-mediated, rituximab-dependent cytotoxicity against lymphoma tumors (Kohrt et al., 2013).
  • Anti-KIR mAbs may be combined with ADCs, leukocyte-redirecting bsAbs, interferons and/or checkpoint inhibitor antibodies to potentiate cytotoxicity to tumor cells or pathogenic organisms.
  • monoclonal antibodies can be obtained by injecting mice with a composition comprising an antigen, removing the spleen to obtain B- lymphocytes, fusing the B-lymphocytes with myeloma cells to produce hybridomas, cloning the hybridomas, selecting positive clones which produce antibodies to the antigen, culturing the clones that produce antibodies to the antigen, and isolating the antibodies from the hybridoma cultures.
  • MAbs can be isolated and purified from hybridoma cultures by a variety of well-established techniques. Such isolation techniques include affinity chromatography with Protein-A Sepharose, size-exclusion chromatography, and ion-exchange chromatography. See, for example, Coligan at pages 2.7.1-2.7.12 and pages 2.9.1-2.9.3. Also, see Baines et al, "Purification of Immunoglobulin G (IgG)," in METHODS IN MOLECULAR BIOLOGY, VOL. 10, pages 79-104 (The Humana Press, Inc. 1992).
  • the antibodies can be sequenced and subsequently prepared by recombinant techniques. Humanization and chimerization of murine antibodies and antibody fragments are well known to those skilled in the art. The use of antibody components derived from humanized, chimeric or human antibodies obviates potential problems associated with the immunogenicity of murine constant regions.
  • a chimeric antibody is a recombinant protein in which the variable regions of a human antibody have been replaced by the variable regions of, for example, a mouse antibody, including the complementarity-determining regions (CDRs) of the mouse antibody.
  • Chimeric antibodies exhibit decreased immunogenicity and increased stability when administered to a subject.
  • CDRs complementarity-determining regions
  • a chimeric or murine monoclonal antibody may be humanized by transferring the mouse CDRs from the heavy and light variable chains of the mouse immunoglobulin into the
  • variable domains of a human antibody The mouse framework regions (FR) in the chimeric monoclonal antibody are also replaced with human FR sequences.
  • additional modification might be required in order to restore the original affinity of the murine antibody. This can be accomplished by the replacement of one or more human residues in the FR regions with their murine counterparts to obtain an antibody that possesses good binding affinity to its epitope. See, for example, Tempest et al., Biotechnology 9:266 (1991) and Verhoeyen et al, Science 239: 1534 (1988).
  • those human FR amino acid residues that differ from their murine counterparts and are located close to or touching one or more CDR amino acid residues would be candidates for substitution.
  • the phage display technique may be used to generate human antibodies ⁇ e.g., Dantas-Barbosa et al., 2005, Genet. Mol. Res. 4: 126-40).
  • Human antibodies may be generated from normal humans or from humans that exhibit a particular disease state, such as cancer (Dantas-Barbosa et al., 2005).
  • the advantage to constructing human antibodies from a diseased individual is that the circulating antibody repertoire may be biased towards antibodies against disease-associated antigens.
  • Fab fragment antigen binding protein
  • RNAs were converted to cDNAs and used to make Fab cDNA libraries using specific primers against the heavy and light chain immunoglobulin sequences (Marks et al., 1991, J. Mol. Biol. 222:581-97). Library construction was performed according to Andris-Widhopf et al. (2000, In: PHAGE DISPLAY
  • Fab fragments were digested with restriction endonucleases and inserted into the bacteriophage genome to make the phage display library.
  • libraries may be screened by standard phage display methods, as known in the art (see, e.g., Pasqualini and Ruoslahti, 1996, Nature 380:364-366; Pasqualini, 1999, The Quart. J. Nucl. Med. 43 : 159-162).
  • Phage display can be performed in a variety of formats, for their review, see e.g. Johnson and Chiswell, Current Opinion in Structural Biology 3 :5564-571 (1993). Human antibodies may also be generated by in vitro activated B cells. See U.S. Patent Nos.
  • transgenic animals that have been genetically engineered to produce human antibodies may be used to generate antibodies against essentially any immunogenic target, using standard immunization protocols.
  • Methods for obtaining human antibodies from transgenic mice are disclosed by Green et al, Nature Genet. 7: 13 (1994), Lonberg et al, Nature 3(55:856 (1994), and Taylor et al, Int. Immun. 6:579 (1994).
  • a non- limiting example of such a system is the XENOMOUSE® ⁇ e.g., Green et al., 1999, J.
  • the XENOMOUSE® was transformed with germline-configured YACs (yeast artificial chromosomes) that contained portions of the human IgH and Igkappa loci, including the majority of the variable region sequences, along accessory genes and regulatory sequences.
  • the human variable region repertoire may be used to generate antibody producing B cells, which may be processed into hybridomas by known techniques.
  • a XENOMOUSE® immunized with a target antigen will produce human antibodies by the normal immune response, which may be harvested and/or produced by standard techniques discussed above.
  • a variety of strains of XENOMOUSE® are available, each of which is capable of producing a different class of antibody.
  • Transgenically produced human antibodies have been shown to have therapeutic potential, while retaining the
  • compositions and methods are not limited to use of the XENOMOUSE® system but may utilize any transgenic animal that has been genetically engineered to produce human antibodies.
  • VK variable light chain
  • the V genes of an antibody from a cell that expresses a murine antibody can be cloned by PCR amplification and sequenced.
  • the cloned V L and V H genes can be expressed in cell culture as a chimeric Ab as described by Orlandi et al, (Proc. Natl Acad. Sci. USA, 86: 3833 (1989)).
  • a humanized antibody can then be designed and constructed as described by Leung et al. (Mol. Immunol, 32: 1413 (1995)).
  • cDNA can be prepared from any known hybridoma line or transfected cell line producing a murine antibody by general molecular cloning techniques (Sambrook et al., Molecular Cloning, A laboratory manual, 2 nd Ed (1989)).
  • the VK sequence for the antibody may be amplified using the primers VKIBACK and VKIFOR (Orlandi et al, 1989) or the extended primer set described by Leung et al. (BioTechniques, 15: 286 (1993)).
  • the V H sequences can be amplified using the primer pair VHIBACK/VHIFOR (Orlandi et al, 1989) or the primers annealing to the constant region of murine IgG described by Leung et al.
  • Humanized V genes can be constructed by a combination of long oligonucleotide template syntheses and PCR amplification as described by Leung et al. (Mol Immunol, 32: 1413 (1995)).
  • PCR products for VK can be subcloned into a staging vector, such as a pBR327-based staging vector, VKpBR, that contains an Ig promoter, a signal peptide sequence and convenient restriction sites.
  • PCR products for V H can be subcloned into a similar staging vector, such as the pBluescript-based VHpBS.
  • Expression cassettes containing the VK and V H sequences together with the promoter and signal peptide sequences can be excised from VKpBR and VHpBS and ligated into appropriate expression vectors, such as pKh and pGlg, respectively (Leung et al., Hybridoma, 13:469 (1994)).
  • the expression vectors can be co-transfected into an appropriate cell and supernatant fluids monitored for production of a chimeric, humanized or human antibody.
  • the VK and V H expression cassettes can be excised and subcloned into a single expression vector, such as pdHL2, as described by Gillies et al (J. Immunol Methods 125: 191 (1989) and also shown in Losman et al., Cancer, 80:2660 (1997)).
  • expression vectors may be transfected into host cells that have been pre-adapted for transfection, growth and expression in serum-free medium.
  • Exemplary cell lines that may be used include the Sp/EEE, Sp/ESF and Sp/ESF-X cell lines (see, e.g., U.S. Patent Nos. 7,531,327; 7,537,930 and 7,608,425; the Examples section of each of which is incorporated herein by reference). These exemplary cell lines are based on the Sp2/0 myeloma cell line, transfected with a mutant Bcl-EEE gene, exposed to methotrexate to amplify transfected gene sequences and pre-adapted to serum-free cell line for protein expression.
  • Antibody fragments which recognize specific epitopes can be generated by known techniques.
  • Antibody fragments are antigen binding portions of an antibody, such as F(ab') 2, Fab', F(ab) 2 , Fab, Fv, scFv and the like.
  • F(ab') 2 fragments can be produced by pepsin digestion of the antibody molecule and Fab ' fragments can be generated by reducing disulfide bridges of the F(ab') 2 fragments.
  • Fab ' expression libraries can be constructed (Huse et al, 1989, Science, 246: 1274-1281) to allow rapid and easy identification of monoclonal Fab' fragments with the desired specificity.
  • F(ab) 2 fragments may be generated by papain digestion of an antibody.
  • a single chain Fv molecule comprises a VL domain and a VH domain.
  • the VL and VH domains associate to form a target binding site.
  • These two domains are further covalently linked by a peptide linker (L).
  • L peptide linker
  • Single domain antibodies may be obtained, for example, from camels, alpacas or llamas by standard immunization techniques. (See, e.g.,
  • the VHH may have potent antigen-binding capacity and can interact with novel epitopes that are inacessible to conventional VH-VL pairs.
  • Alpaca serum IgG contains about 50% camelid heavy chain only IgG antibodies (HCAbs) (Maass et al., 2007).
  • Alpacas may be immunized with known antigens, such as T F- ⁇ , and VHHs can be isolated that bind to and neutralize the target antigen (Maass et al., 2007).
  • PCR primers that amplify virtually all alpaca VHH coding sequences have been identified and may be used to construct alpaca VHH phage display libraries, which can be used for antibody fragment isolation by standard biopanning techniques well known in the art (Maass et al., 2007).
  • anti-pancreatic cancer VHH antibody fragments may be utilized in the claimed compositions and methods.
  • An antibody fragment can be prepared by proteolytic hydrolysis of the full length antibody or by expression in E. coli or another host of the DNA coding for the fragment.
  • An antibody fragment can be obtained by pepsin or papain digestion of full length antibodies by conventional methods. These methods are described, for example, by Goldenberg, U.S. Patent Nos. 4,036,945 and 4,331,647 and references contained therein. Also, see Nisonoff et al, Arch Biochem. Biophys. 89: 230 (1960); Porter, Biochem. J. 73: 119 (1959), Edelman et al, in METHODS IN ENZYMOLOGY VOL. 1, page 422 (Academic Press 1967), and Coligan at pages 2.8.1-2.8.10 and 2.10.-2.10.4.
  • Immunogenicity of therapeutic antibodies is associated with increased risk of infusion reactions and decreased duration of therapeutic response (Baert et al., 2003, NEngl J Med 348:602-08).
  • the extent to which therapeutic antibodies induce an immune response in the host may be determined in part by the allotype of the antibody (Stickler et al., 2011, Genes and Immunity 12:213-21).
  • Antibody allotype is related to amino acid sequence variations at specific locations in the constant region sequences of the antibody.
  • the allotypes of IgG antibodies containing a heavy chain ⁇ -type constant region are designated as Gm allotypes (1976, J Immunol 111: 1056-59).
  • Glml For the common IgGl human antibodies, the most prevalent allotype is Glml (Stickler et al., 2011, Genes and Immunity 12:213-21). However, the Glm3 allotype also occurs frequently in Caucasians (Stickler et al., 2011). It has been reported that Glml antibodies contain allotypic sequences that tend to induce an immune response when administered to non- Glml (nGlml) recipients, such as Glm3 patients (Stickler et al., 2011). Non-Glml allotype antibodies are not as immunogenic when administered to Glml patients (Stickler et al., 2011).
  • the human Glml allotype comprises the amino acids aspartic acid at Kabat position 356 and leucine at Kabat position 358 in the CH3 sequence of the heavy chain IgGl.
  • the nGlml allotype comprises the amino acids glutamic acid at Kabat position 356 and methionine at Kabat position 358.
  • Both Glml and nGlml allotypes comprise a glutamic acid residue at Kabat position 357 and the allotypes are sometimes referred to as DEL and EEM allotypes.
  • a non-limiting example of the heavy chain constant region sequences for Glml and nGlml allotype antibodies is shown for the exemplary antibodies rituximab (SEQ ID NO:20) and veltuzumab (SEQ ID NO:21).
  • veltuzumab and rituximab are, respectively, humanized and chimeric IgGl antibodies against CD20, of use for therapy of a wide variety of hematological malignancies and/or autoimmune diseases.
  • Table 1 compares the allotype sequences of rituximab vs. veltuzumab.
  • rituximab (Glml7,l) is a DEL allotype IgGl, with an additional sequence variation at Kabat position 214 (heavy chain CHI) of lysine in rituximab vs. arginine in veltuzumab.
  • veltuzumab is less immunogenic in subjects than rituximab ⁇ see, e.g., Morchhauser et al., 2009, J Clin Oncol 27:3346-53; Goldenberg et al., 2009, Blood 113: 1062-70; Robak & Robak, 2011, BioDrugs 25 : 13-25), an effect that has been attributed to the difference between humanized and chimeric antibodies.
  • the difference in allotypes between the EEM and DEL allotypes likely also accounts for the lower immunogenicity of veltuzumab.
  • the allotype of the antibody In order to reduce the immunogenicity of therapeutic antibodies in individuals of nGlml genotype, it is desirable to select the allotype of the antibody to correspond to the Glm3 allotype, characterized by arginine at Kabat 214, and the nGlml,2 null-allotype, characterized by glutamic acid at Kabat position 356, methionine at Kabat position 358 and alanine at Kabat position 431. Surprisingly, it was found that repeated subcutaneous administration of Glm3 antibodies over a long period of time did not result in a significant immune response.
  • the human IgG4 heavy chain in common with the Glm3 allotype has arginine at Kabat 214, glutamic acid at Kabat 356, methionine at Kabat 359 and alanine at Kabat 431. Since immunogenicity appears to relate at least in part to the residues at those locations, use of the human IgG4 heavy chain constant region sequence for therapeutic antibodies is also a preferred embodiment. Combinations of Glm3 IgGl antibodies with IgG4 antibodies may also be of use for therapeutic administration.
  • antibodies are used that recognize and/or bind to antigens that are expressed at high levels on target cells and that are expressed predominantly or exclusively on diseased cells versus normal tissues.
  • Exemplary antibodies of use for therapy of, for example, cancer include but are not limited to LL1 (anti-CD74), LL2 or RFB4 (anti- CD22), veltuzumab (hA20, anti-CD20), rituxumab (anti-CD20), obinutuzumab (GA101, anti- CD20), lambrolizumab (anti-PDl), nivolumab (anti-PDl),MK-3475 (anti-PDl), AMP-224 (anti-PDl), pidilizumab (anti-PDl), MDX-1105 (anti-PD-Ll), MEDI4736 (anti-PD-Ll), MPDL3280A (anti-PD-Ll), BMS-936559 (anti-PD-Ll), ipilimumab (
  • Such antibodies are known in the art (e.g., U.S. Patent Nos. 5,686,072; 5,874,540; 6, 107,090; 6, 183,744; 6,306,393; 6,653,104; 6,730.300; 6,899,864; 6,926,893; 6,962,702; 7,074,403; 7,230,084; 7,238,785; 7,238,786; 7,256,004; 7,282,567; 7,300,655; 7,312,318; 7,585,491; 7,612,180; 7,642,239; and U.S. Patent Application Publ. No. 20050271671;
  • hPAM4 U.S. Patent No. 7,282,567
  • hA20 U.S. Patent No. 7, 151,164
  • hA19 U.S. Patent No. 7,109,304
  • hFMMU- 31 U.S. Patent No. 7,300,655
  • hLLl U.S. Patent No. 7,312,318,
  • hLL2 U.S. Patent No. 5,789,554
  • hMu-9 U.S. Patent No. 7,387,772
  • hL243 U.S. Patent No.
  • alpha-fetoprotein AFP
  • carbonic anhydrase IX B7, CCL19, CCL21, CSAp, ⁇ -2/neu, BrE3, CDl, CDla, CD2, CD3, CD4, CD5, CD8, CDl lA, CD14, CD15, CD16, CD18, CD19, CD20 (e.g., C2B8, hA20, 1F5 MAbs), CD21, CD22, CD23, CD25, CD29, CD30, CD32b, CD33, CD37, CD38, CD40, CD40L, CD44, CD45, CD46, CD52, CD54, CD55, CD59, CD64, CD67, CD70, CD74, CD79a, CD80, CD83, CD95, CD126, CD133, CD138, CD147, CD154, CEACAM5, CEACAM6, CTLA4, DLL3, DLL4, VEGF (e.g., AVASTIN®,
  • CD Cluster Designation
  • the CD66 antigens consist of five different glycoproteins with similar structures, CD66a-e, encoded by the carcinoembryonic antigen (CEA) gene family members, BCG, CGM6, NCA, CGM1 and CEA, respectively. These CD66 antigens (e.g., CEACAM6) are expressed mainly in granulocytes, normal epithelial cells of the digestive tract and tumor cells of various tissues. Also included as suitable targets for cancers are cancer testis antigens, such as NY-ESO-1 (Theurillat et al., Int. J. Cancer 2007; 120(11):2411-7), as well as CD79a in myeloid leukemia (Kozlov et al., Cancer Genet.
  • CEACAM6 carcinoembryonic antigen
  • cancers such as CD133 in prostate cancer (Maitland et al., Ernst Schering Found. Sympos. Proc. 2006; 5: 155-79), non-small-cell lung cancer (Donnenberg et al., J. Control Release 2007; 122(3):385-91), and glioblastoma (Beier et al., Cancer Res. 2007; 67(9):4010-5), and CD44 in colorectal cancer (Dalerba er al., Proc. Natl. Acad. Sci. USA 2007; 104(24)10158-63), pancreatic cancer (Li et al., Cancer Res. 2007; 67(3): 1030-7), and in head and neck squamous cell carcinoma (Prince et al., Proc. Natl. Acad. Sci. USA 2007; 104(3)973-8).
  • cancer types such as CD133 in prostate cancer (Maitland et al., Ernst Schering Found. Sympos. Proc. 2006; 5
  • lymphocytes produce natural antibodies to histones.
  • antibodies against histones may be of use in the subject combinations.
  • Known anti-histone antibodies include, but are not limited to, BWA-3 (anti-histone H2A/H4), LG2-1 (anti-histone H3), MRA12 (anti-histone HI), PRl-1 (anti-histone H2B), LG11-2 (anti-histone H2B), and LG2- 2 (anti-histone H2B) (see, e.g., Monestier et al., 1991, Eur J Immunol 21 : 1725-31; Monestier et al., 1993, Molec Immunol 30: 1069-75).
  • Macrophage migration inhibitory factor is an important regulator of innate and adaptive immunity and apoptosis. It has been reported that CD74 is the endogenous receptor for MIF (Leng et al., 2003, J Exp Med 197: 1467-76).
  • the therapeutic effect of antagonistic anti-CD74 antibodies on MIF-mediated intracellular pathways may be of use for treatment of a broad range of disease states, such as cancers of the bladder, prostate, breast, lung, colon and chronic lymphocytic leukemia (e.g., Meyer-Siegler et al., 2004, BMC Cancer 12:34; Shachar & Haran, 2011, Leuk Lymphoma 52: 1446-54).
  • Milatuzumab hLLl
  • An example of a most-preferred antibody/antigen pair is LL1, an anti-CD74 MAb (invariant chain, class II-specific chaperone, Ii) (see, e.g., U.S. Patent Nos. 6,653,104;
  • the CD74 antigen is highly expressed on B-cell lymphomas (including multiple myeloma) and leukemias, certain T-cell lymphomas, melanomas, colonic, lung, and renal cancers, glioblastomas, and certain other cancers (Ong et al., Immunology 95:296-302 (1999)).
  • B-cell lymphomas including multiple myeloma
  • leukemias certain T-cell lymphomas
  • melanomas melanomas
  • colonic, lung, and renal cancers glioblastomas
  • glioblastomas and certain other cancers
  • the diseases that are preferably treated with anti-CD74 antibodies include, but are not limited to, non-Hodgkin's lymphoma, Hodgkin's disease, melanoma, lung, renal, colonic cancers, glioblastome multiforme, histiocytomas, myeloid leukemias, and multiple myeloma.
  • the therapeutic combinations can be used against pathogens, since antibodies against pathogens are known.
  • antibodies and antibody fragments which specifically bind markers produced by or associated with infectious lesions, including viral, bacterial, fungal and parasitic infections, for example caused by pathogens such as bacteria, rickettsia, mycoplasma, protozoa, fungi, and viruses, and antigens and products associated with such microorganisms have been disclosed, inter alia, in Hansen et al., U.S. Pat. No. 3,927,193 and Goldenberg U.S. Pat. Nos.
  • Legionella spp. (Cat. #01-90-03), Listeria spp. (Cat. #01-90-90), Vibrio cholera (Cat. #01-90- 50), Shigella spp. (Cat. #16-90-01), and Campylobacter spp. (Cat. #01-92-93).
  • the pathogens are selected from the group consisting of HIV virus, Mycobacterium tuberculosis, Streptococcus agalactiae, methicillin-resistant Staphylococcus aureus, Legionella pneumophilia, Streptococcus pyogenes, Escherichia coli, Neisseria gonorrhoeae, Neisseria meningitidis, Pneumococcus, Cryptococcus neoformans, Histoplasma capsulatum, Hemophilis influenzae B, Treponema pallidum, Lyme disease spirochetes, Pseudomonas aeruginosa, Mycobacterium leprae, Brucella abortus, rabies virus, influenza virus, cytomegalovirus, herpes simplex virus I, herpes simplex virus II, human serum parvo-like virus, respiratory syncytial virus, varicella
  • Toxoplasma gondii Trypanosoma rangeli, Trypanosoma cruzi, Trypanosoma rhodesiensei, Trypanosoma brucei, Schistosoma mansoni, Schistosoma japoni cum, Babesia bovis, Elmeria tenella, Onchocerca volvulus, Leishmania tropica, Trichinella spiralis, Theileria parva, Taenia hydatigena, Taenia ovis, Taenia saginata, Echinococcus granulosus, Mesocestoides corti, Mycoplasma arthritidis, M. hyorhinis, M. orale, M. arginini, Acholeplasma laidlawii, M. salivarium and M. pneumoniae, as disclosed in U.S. Patent No. 6,440,416, the Examples section of which is incorporated herein by reference.
  • the claimed methods and compositions may utilize any of a variety of antibodies known in the art.
  • Antibodies of use may be commercially obtained from a number of known sources.
  • a variety of antibody secreting hybridoma lines are available from the American Type Culture Collection (ATCC, Manassas, VA).
  • ATCC American Type Culture Collection
  • VA Manassas
  • a large number of antibodies against various disease targets, including but not limited to tumor- associated antigens, have been deposited at the ATCC and/or have published variable region sequences and are available for use in the claimed methods and compositions. See, e.g., U.S. Patent Nos.
  • antibody sequences or antibody-secreting hybridomas against almost any disease-associated antigen may be obtained by a simple search of the ATCC, NCBI and/or USPTO databases for antibodies against a selected disease-associated target of interest.
  • the antigen binding domains of the cloned antibodies may be amplified, excised, ligated into an expression vector, transfected into an adapted host cell and used for protein production, using standard techniques well known in the art (see, e.g., U. S. Patent Nos. 7,531,327; 7,537,930; 7,608,425 and 7,785,880, the Examples section of each of which is incorporated herein by reference).
  • the antibody complexes bind to a MHC class I, MHC class II or accessory molecule, such as CD40, CD54, CD80 or CD86.
  • the antibody complex also may bind to a leukocyte activation cytokine, or to a cytokine mediator, such as F-KB.
  • one of the two different targets may be a cancer cell receptor or cancer-associated antigen, particularly one that is selected from the group consisting of B-cell lineage antigens (CD19, CD20, CD21, CD22, CD23, etc.), VEGF, VEGFR, EGFR, carcinoembryonic antigen (CEA), placental growth factor (P1GF), tenascin, ⁇ -2/neu, EGP-1, EGP-2, CD25, CD30, CD33, CD38, CD40, CD45, CD52, CD74, CD80, CD138, DLL3, DLL4, NCA66, CEACAM1, CEACAM6 (carcinoembryonic antigen-related cellular adhesion molecule 6), mesothelin, MUC1, MUC2, MUC3, MUC4, MUC5ac, IL-6, a-fetoprotein (AFP), A3, CA125, colon-specific antigen-p (CSAp), folate receptor, ULA-DR, human
  • HIV-1 human immunodeficiency virus I
  • Known anti-HIV antibodies include the anti-envelope antibody described by Johansson et al.
  • Antibodies against malaria parasites can be directed against the sporozoite, merozoite, schizont and gametocyte stages. Monoclonal antibodies have been generated against sporozoites (cirumsporozoite antigen), and have been shown to neutralize sporozoites in vitro and in rodents (N. Yoshida et al., Science 207:71-73, 1980). Several groups have developed antibodies to T. gondii, the protozoan parasite involved in toxoplasmosis (Kasper et al., J. Immunol. 129: 1694-1699, 1982; Id., 30:2407-2412, 1983).
  • Antibodies have been developed against schistosomular surface antigens and have been found to act against schistosomulae in vivo or in vitro (Simpson et al., Parasitology, 83 : 163-177, 1981; Smith et al., Parasitology, 84:83-91, 1982: Gryzch et al., J. Immunol., 129:2739-2743, 1982; Zodda et al., J. Immunol. 129:2326-2328, 1982; Dissous et al., J. Immunol., 129:2232-2234, 1982)
  • Trypanosoma cruzi is the causative agent of Chagas' disease, and is transmitted by blood-sucking reduviid insects.
  • An antibody has been generated that specifically inhibits the differentiation of one form of the parasite to another (epimastigote to trypomastigote stage) in vitro, and which reacts with a cell-surface glycoprotein; however, this antigen is absent from the mammalian (bloodstream) forms of the parasite (Sher et al., Nature, 300:639-640, 1982).
  • Anti-fungal antibodies are known in the art, such as anti-Sclerotinia antibody (U.S. Patent 7,910,702); antiglucuronoxylomannan antibody (Zhong and Priofski, 1998, Clin Diag Lab Immunol 5:58-64); anti-Candida antibodies (Matthews and Burnie, 2001, Curr Opin Investig Drugs 2:472-76); and anti-glycosphingolipid antibodies (Toledo et al., 2010, BMC Microbiol 10:47).
  • Suitable antibodies have been developed against most of the microorganism (bacteria, viruses, protozoa, fungi, other parasites) responsible for the majority of infections in humans, and many have been used previously for in vitro diagnostic purposes. These antibodies, and newer antibodies that can be generated by conventional methods, are appropriate for use in the present invention.
  • the antibodies or fragments thereof may be conjugated to one or more therapeutic or diagnostic agents.
  • the therapeutic agents do not need to be the same but can be different, e.g. a drug and a radioisotope.
  • 131 I can be incorporated into a tyrosine of an antibody or fusion protein and a drug attached to an epsilon amino group of a lysine residue.
  • Therapeutic and diagnostic agents also can be attached, for example to reduced SH groups and/or to carbohydrate side chains. Many methods for making covalent or non-covalent conjugates of therapeutic or diagnostic agents with antibodies or fusion proteins are known in the art and any such known method may be utilized.
  • a therapeutic or diagnostic agent can be attached at the hinge region of a reduced antibody component via disulfide bond formation.
  • such agents can be attached using a heterobifunctional cross-linker, such as N-succinyl 3-(2-pyridyldithio)propionate (SPDP). Yu et al, Int. J. Cancer 56: 244 (1994).
  • SPDP N-succinyl 3-(2-pyridyldithio)propionate
  • the therapeutic or diagnostic agent can be conjugated via a carbohydrate moiety in the Fc region of the antibody.
  • the carbohydrate group can be used to increase the loading of the same agent that is bound to a thiol group, or the carbohydrate moiety can be used to bind a different therapeutic or diagnostic agent.
  • the general method involves reacting an antibody component having an oxidized carbohydrate portion with a carrier polymer that has at least one free amine function. This reaction results in an initial Schiff base (imine) linkage, which can be stabilized by reduction to a secondary amine to form the final conjugate.
  • the Fc region may be absent if the antibody used as the antibody component of the immunoconjugate is an antibody fragment. However, it is possible to introduce a
  • carbohydrate moiety into the light chain variable region of a full length antibody or antibody fragment. See, for example, Leung et al, J. Immunol. 154: 5919 (1995); Hansen et al, U.S. Patent No. 5,443,953 (1995), Leung et al, U.S. patent No. 6,254,868, incorporated herein by reference in their entirety.
  • the engineered carbohydrate moiety is used to attach the therapeutic or diagnostic agent.
  • a chelating agent may be attached to an antibody, antibody fragment or fusion protein and used to chelate a therapeutic or diagnostic agent, such as a radionuclide.
  • exemplary chelators include but are not limited to DTPA (such as Mx-DTPA), DOTA, TETA, NETA or NOTA.
  • radioactive metals or paramagnetic ions may be attached to proteins or peptides by reaction with a reagent having a long tail, to which may be attached a multiplicity of chelating groups for binding ions.
  • a tail can be a polymer such as a polylysine, polysaccharide, or other derivatized or derivatizable chains having pendant groups to which can be bound chelating groups such as, e.g., ethylenediaminetetraacetic acid (EDTA), diethylenetriaminepentaacetic acid (DTPA), porphyrins, polyamines, crown ethers, bis-thiosemicarbazones, polyoximes, and like groups known to be useful for this purpose.
  • EDTA ethylenediaminetetraacetic acid
  • DTPA diethylenetriaminepentaacetic acid
  • porphyrins polyamines, crown ethers, bis-thiosemicarbazones, polyoximes, and like groups known to be useful for
  • Chelates may be directly linked to antibodies or peptides, for example as disclosed in U.S. Patent 4,824,659, incorporated herein in its entirety by reference.
  • Particularly useful metal-chelate combinations include 2-benzyl-DTPA and its monomethyl and cyclohexyl analogs, used with diagnostic isotopes in the general energy range of 60 to 4,000 keV, such
  • Such metal-chelate complexes can be made very stable by tailoring the ring size to the metal of interest.
  • Other ring-type chelates such as macrocyclic polyethers, which are of interest for stably binding nuclides, such as 223 Ra for RAIT are encompassed.
  • 18 F-labeling of use in PET scanning techniques for example by reaction of F-18 with a metal or other atom, such as aluminum.
  • the 18 F-A1 conjugate may be complexed with chelating groups, such as DOTA, NOTA or NETA that are attached directly to antibodies or used to label targetable constructs in pre- targeting methods.
  • chelating groups such as DOTA, NOTA or NETA that are attached directly to antibodies or used to label targetable constructs in pre- targeting methods.
  • Such F-18 labeling techniques are disclosed in U.S. Patent No. 7,563,433, the Examples section of which is incorporated herein by reference.
  • the immunoconjugate may comprise a
  • camptothecin drug such as SN-38.
  • Camptothecin (CPT) and its derivatives are a class of potent antitumor agents.
  • Irinotecan (also referred to as CPT-11) and topotecan are CPT analogs that are approved cancer therapeutics (Iyer and Ratain, Cancer Chemother.
  • CPTs act by inhibiting topoisomerase I enzyme by stabilizing topoisomerase I-DNA complex (Liu, et al. in The Camptothecins: Unfolding Their Anticancer Potential, Liehr J.G., Giovanella, B.C. and Verschraegen (eds), NY Acad Sci., NY 922: 1-10 (2000)).
  • Preferred optimal dosing of immunoconjugates may include a dosage of between 3 mg/kg and 20 mg/kg, preferably given either weekly, twice weekly or every other week.
  • the optimal dosing schedule may include treatment cycles of two consecutive weeks of therapy followed by one, two, three or four weeks of rest, or alternating weeks of therapy and rest, or one week of therapy followed by two, three or four weeks of rest, or three weeks of therapy followed by one, two, three or four weeks of rest, or four weeks of therapy followed by one, two, three or four weeks of rest, or five weeks of therapy followed by one, two, three, four or five weeks of rest, or administration once every two weeks, once every three weeks or once a month.
  • Treatment may be extended for any number of cycles, preferably at least 2, at least 4, at least 6, at least 8, at least 10, at least 12, at least 14, or at least 16 cycles.
  • the dosage may be up to 24 mg/kg.
  • Exemplary dosages of use may include 1 mg/kg, 2 mg/kg, 3 mg/kg, 4 mg/kg, 5 mg/kg, 6 mg/kg, 7 mg/kg, 8 mg/kg, 9 mg/kg, 10 mg/kg, 11 mg/kg, 12 mg/kg, 13 mg/kg, 14 mg/kg, 15 mg/kg, 16 mg/kg, 17 mg/kg, 18 mg/kg, 19 mg/kg, 20 mg/kg, 22 mg/kg and 24 mg/kg.
  • Preferred dosages are 4, 6, 8, 9, 10, 12, 14, 16 or 18 mg/kg.
  • the person of ordinary skill will realize that a variety of factors, such as age, general health, specific organ function or weight, as well as effects of prior therapy on specific organ systems (e.g., bone marrow) may be considered in selecting an optimal dosage of immunoconjugate, and that the dosage and/or frequency of administration may be increased or decreased during the course of therapy.
  • the dosage may be repeated as needed, with evidence of tumor shrinkage observed after as few as 4 to 8 doses.
  • the optimized dosages and schedules of administration disclosed herein show unexpected superior efficacy and reduced toxicity in human subjects, which could not have been predicted from animal model studies. Surprisingly, the superior efficacy allows treatment of tumors that were previously found to be resistant to one or more standard anti-cancer therapies, including the parental compound, CPT-11, from which SN-38 is derived in vivo.
  • An exemplary preferred embodiment is directed to a conjugate of a drug derivative and an antibody of the general formula 1,
  • MAb is a disease-targeting antibody
  • L2 is a component of the cross-linker comprising an antibody-coupling moiety and one or more of acetylene (or azide) groups
  • LI comprises a defined PEG with azide (or acetylene) at one end, complementary to the acetylene (or azide) moiety in L2, and a reactive group such as carboxylic acid or hydroxyl group at the other end
  • AA is an L-amino acid
  • m is an integer with values of 0, 1, 2, 3, or 4
  • A' is an additional spacer, selected from the group of ethanolamine, 4-hydroxybenzyl alcohol, 4-aminobenzyl alcohol, or substituted or unsubstituted ethylenediamine.
  • the L amino acids of 'AA' are selected from alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine. If the A' group contains hydroxyl, it is linked to the hydroxyl group or amino group of the drug in the form of a carbonate or carbamate, respectively.
  • A' is a substituted ethanolamine derived from an L-amino acid, wherein the carboxylic acid group of the amino acid is replaced by a hydroxymethyl moiety.
  • A' may be derived from any one of the following L-amino acids: alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine.
  • m is
  • A' is L-valinol
  • the drug is exemplified by SN-38. The resultant structure is shown in formula 2.
  • m is 1 and represented by a derivatized L-lysine, A' is L-valinol, and the drug is exemplified by SN- 38.
  • the structure is shown in formula 3.
  • an amide bond is first formed between the carboxylic acid of an amino acid such as lysine and the amino group of valinol, using orthogonal protecting groups for the lysine amino groups.
  • the protecting group on the N-terminus of lysine is removed, keeping the protecting group on the side chain of lysine intact, and the N-terminus is coupled to the carboxyl group on the defined PEG with azide (or acetylene) at the other end.
  • A' of the general formula 2 is A-OH, whereby A- OH is a collapsible moiety such as 4-aminobenzyl alcohol or a substituted 4-aminobenzyl alcohol substituted with a Ci-Ci 0 alkyl group at the benzylic position, and the latter, via its amino group, is attached to an L-amino acid or a polypeptide comprising up to four L-amino acid moieties; wherein the N-terminus is attached to a cross-linker terminating in the antibody-binding group.
  • A- OH is a collapsible moiety such as 4-aminobenzyl alcohol or a substituted 4-aminobenzyl alcohol substituted with a Ci-Ci 0 alkyl group at the benzylic position, and the latter, via its amino group, is attached to an L-amino acid or a polypeptide comprising up to four L-amino acid moieties; wherein the N-terminus is attached to a
  • the structure is represented below (formula 2, referred to as MAb-CLX-SN-38).
  • Single amino acid of AA is selected from any one of the following L- amino acids: alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine.
  • the substituent R on 4-aminobenzyl alcohol moiety is hydrogen or an alkyl group selected from C1-C10 alkyl groups
  • Methods of preparing CL2A-SN-38 and for making and using antibody conjugates thereof are known in the art (see, e.g., U.S. Patent Nos. 7,999,083 and 8,080,250, the Examples sections of each incorporated herein by reference).
  • Pro-2-pyrrolinodoxorubicin may be prepared as disclosed herein and conjugated to antibodies or antibody fragments for use in ADC therapy.
  • Pro-2-P-Dox P2PDox
  • activated Pro-2-P-Dox For coupling to IgG, Pro-2-P-Dox may be activated with SMCC- hydrazide, a procedure that introduces acid-labile hydrazone as well as the maleimide group, the latter for conjugation to thiols of mildly reduced antibody.
  • a DNA-alkylating agent such as 2-PDox, is cell-cycle- p ase-nonspecific and should provide an improved therapeutic index.
  • the conjugate preparation mixed mildly reducing interchain disulfides of IgG with TCEP in PBS, followed by coupling to a 10-fold excess of activated P2PDox.
  • the conjugates were purified on centrifuged SEC on SEPHADEX® equilibrated in 25 mM histidine, pH 7, followed by passage over a hydrophobic column.
  • the products were formulated with trehalose and Tween 80, and lyophilized.
  • the conjugated product with a typical substitution of 6-7 drug/IgG, eluted as a single peak by size-exclusion HPLC, and contained typically ⁇ 1% of unconjugated free drug by reversed-phase HPLC.
  • P2PDox may be conjugated to any known antibody or fragment thereof, for use in ADC treatment of tumors and/or infectious disease, in combination with immunomodulating agents discussed herein.
  • the subject combination therapy may utilize one or more bispecific antibodies (bsAbs), such as a leukocyte redirecting bsAb.
  • bsAbs bispecific antibodies
  • a bispecific antibody as used herein is an antibody that contains binding sites for two different antigens, or two different epitopes on the same antigen. An antibody that can only bind to a single epitope on a single antigen is monospecific, regardless of the number of antigen-binding sites on the antibody molecule.
  • constructs that are capable of producing homogeneous products of single bsAbs, without the need for extensive purification to remove unwanted byproducts.
  • constructs have included tandem scFv, diabodies, tandem diabodies, dual variable domain antibodies and heterodimerization using a motif such as Chl/Ck domain or D L® (Chames & Baty, 2009, Curr Opin Drug Discov Devel 12:276-83; Chames & Baty, mAbs 1 :539-47).
  • Triomabs is a variation on the quadroma approach that use a combination of mouse IgG2a and rat IgG2b antibodies to preferentially produce the recombinant antibody, compared to the random pairing typically seen in rat/rat or mouse/mouse quadromas (Chames & Baty, mAbs 1 :539-47).
  • An anti-CD3 x anti-EpCAM bsAb (catumaxomab) created by this technique was able to efficiently recruit macrophages and NK cells and to activate T cells (Chames & Baty, mAbs 1 :539-47).
  • catumaxomab has been approved in Europe for treatment of malignant ascites in patients with EpCAM positive carcinomas (Chames & Baty, mAbs 1 : 539-47).
  • the recombinant bsAb was reported to induce only moderate anti-mouse and anti-rat responses in humans (Chames & Baty, mAbs 1 :539-47), probably due at least in part to the i.p. route of administration for ascites.
  • Ertumaxomab is another triomab targeting HER2, which may be of use for metastatic breast cancer.
  • Bi20 is another triomab that targets CD20. In vitro, Bi20 exibited efficient lyis of B cells from CLL patients (Chames & Baty, mAbs 1 :539-47).
  • BITE® refers to tandem scFvs that are joined by a short peptide linker (Chames & Baty, mAbs 1 :539-47).
  • Blinatumomab is an anti-CD19 x anti-CD3 BITE® with reported efficacy in hematologic cancers, such as non-Hodgkin's lymphoma and ALL, at very low concentrations (Nagorsen et al., 2009, Leukemia & Lymphoma 50:886-91; Chames & Baty, mAbs 1 :539-47; Topp et al., 2012, Blood 120:5185-87; Bargou et al., 2008, Science 321 :974- 77).
  • Another BITE® with specificity for EpCAM has been used in gastrointestinal, ovarian, colorectal and lung cancer (Amann et al., 2009, J Immunother 32:452-64; Chames & Baty, mAbs 1 :539-47).
  • Another BITE® (MEDI-565) targeted to CEACAM5 has been proposed for use in melanoma, colorectal, lung, pancreatic, stomach, ovarian, uterine, and breast cancers (Sanders et al., 1994, J Pathol 172:343-8).
  • BITE® has been reported to exhibit anti-tumor activity at picomolar or even femtomolar concentrations (Chames & Baty, mAbs 1 :539-47).
  • Another method of bsAb formation involving assembly of two heavy and two light chains derived from two different pre-existing antibodies, is based on a knobs-into-holes approach that facilitates heterodimer formation and prevents homodimer formation (Schaefer et al., 2011, Proc Natl. Acad Sci USA 108: 11187-92).
  • the "CrossMab” technique further involves the exchange of heavy and light chain domains within the Fab of one half of the bispecific antibody, making the two arms so different that light-heavy chain mispairing can not occur (Schaefer et al., 2011).
  • the knobs-into-holes approach introduces amino acids with bulky side chains into the CH3 domain of one heavy chain that fit into appropriately designed cavities in the CH3 domain of the other heavy chain.
  • the combination of approaches prevents mis-match of both heavy chain to heavy chain and heavy chain to light chain interactions, resulting in primarily a single product.
  • the initial CrossMab generated against angiopoietin-2 (Ang-2) and VEGF-A, exhibited binding characteristics comparable to the parent mAbs, with potent anti -angiogenic and anti-tumoral activity (Schaefer et al., 2011, Proc Natl. Acad Sci USA 108: 11187-92; Kienast et al., Clin Cancer Res, Oct. 25, 2013, Epub ahead of print).
  • a bispecific antibody either alone or else complexed to one or more effectors such as cytokines, is formed as a DOCK-AND-LOCK® (DNL®) complex (see, e.g., U.S. Patent Nos.
  • the technique takes advantage of the specific and high-affinity binding interactions that occur between a dimerization and docking domain (DDD) sequence of the regulatory (R) subunits of cAMP-dependent protein kinase (PKA) and an anchor domain (AD) sequence derived from any of a variety of AKAP proteins (Baillie et al., FEBS Letters. 2005; 579: 3264. Wong and Scott, Nat. Rev. Mol. Cell Biol.
  • DDD dimerization and docking domain
  • R regulatory subunits of cAMP-dependent protein kinase
  • AD anchor domain
  • the DDD and AD peptides may be attached to any protein, peptide or other molecule. Because the DDD sequences spontaneously dimerize and bind to the AD sequence, the technique allows the formation of complexes between any selected molecules that may be attached to DDD or AD sequences.
  • the standard DNL® complex comprises a trimer with two DDD-linked molecules attached to one AD-linked molecule
  • variations in complex structure allow the formation of dimers, trimers, tetramers, pentamers, hexamers and other multimers.
  • the DNL® complex may comprise two or more antibodies, antibody fragments or fusion proteins which bind to the same antigenic determinant or to two or more different antigens.
  • the DNL® complex may also comprise one or more other effectors, such as proteins, peptides, immunomodulators, cytokines, interleukins, interferons, binding proteins, peptide ligands, carrier proteins, toxins, ribonucleases such as onconase, inhibitory oligonucleotides such as siRNA, antigens or xenoantigens, polymers such as PEG, enzymes, therapeutic agents, hormones, cytotoxic agents, anti-angiogenic agents, pro-apoptotic agents or any other molecule or aggregate.
  • effectors such as proteins, peptides, immunomodulators, cytokines, interleukins, interferons, binding proteins, peptide ligands, carrier proteins, toxins, ribonucleases such as onconase, inhibitory oligonucleotides such as siRNA, antigens or xenoantigens, polymers such as PEG, enzymes, therapeutic agents, hormones,
  • PKA which plays a central role in one of the best studied signal transduction pathways triggered by the binding of the second messenger cAMP to the R subunits, was first isolated from rabbit skeletal muscle in 1968 (Walsh et al, J. Biol. Chem. 1968;243 :3763).
  • the structure of the holoenzyme consists of two catalytic subunits held in an inactive form by the R subunits (Taylor, J. Biol. Chem. 1989;264:8443). Isozymes of PKA are found with two types of R subunits (RI and RII), and each type has a and ⁇ isoforms (Scott, Pharmacol. Ther. 1991;50: 123).
  • the four isoforms of PKA regulatory subunits are RIa, Ri , Rlla and RIi , each of which comprises a DDD moiety amino acid sequence.
  • the R subunits have been isolated only as stable dimers and the dimerization domain has been shown to consist of the first 44 amino-terminal residues of Rlla (Newlon et al., Nat. Struct. Biol. 1999; 6:222).
  • similar portions of the amino acid sequences of other regulatory subunits are involved in dimerization and docking, each located near the N-terminal end of the regulatory subunit.
  • Binding of cAMP to the R subunits leads to the release of active catalytic subunits for a broad spectrum of serine/threonine kinase activities, which are oriented toward selected substrates through the compartmentalization of PKA via its docking with AKAPs (Scott et al., J. Biol. Chem. 1990;265;21561)
  • AKAP microtubule-associated protein-2
  • the amino acid sequences of the AD are varied among individual AKAPs, with the binding affinities reported for RII dimers ranging from 2 to 90 nM (Alto et al, Proc. Natl. Acad. Sci. USA 2003; 100:4445). AKAPs will only bind to dimeric R subunits.
  • the AD binds to a hydrophobic surface formed by the 23 amino-terminal residues (Colledge and Scott, Trends Cell Biol. 1999; 6:216).
  • the dimerization domain and AKAP binding domain of human Rlla are both located within the same N-terminal 44 amino acid sequence (Newlon et al., Nat. Struct. Biol. 1999;6:222;
  • Entity B is constructed by linking an AD sequence to a precursor of B, resulting in a second component hereafter referred to as b.
  • the dimeric motif of DDD contained in a 2 will create a docking site for binding to the AD sequence contained in b, thus facilitating a ready association of a 2 and b to form a binary, trimeric complex composed of a 2 b.
  • This binding event is stabilized with a subsequent reaction to covalently secure the two entities via disulfide bridges, which occurs very efficiently based on the principle of effective local concentration because the initial binding interactions should bring the reactive thiol groups placed onto both the DDD and AD into proximity (Chmura et al., Proc. Natl. Acad. Sci. USA 2001;98: 8480) to ligate site-specifically.
  • stoichiometry may be produced and used (see, e.g., U.S. Nos. 7,550, 143; 7,521,056;
  • fusion proteins A variety of methods are known for making fusion proteins, including nucleic acid synthesis, hybridization and/or amplification to produce a synthetic double-stranded nucleic acid encoding a fusion protein of interest. Such double-stranded nucleic acids may be inserted into expression vectors for fusion protein production by standard molecular biology techniques (see, e.g. Sambrook et al., MOLECULAR CLONING, A LABORATORY
  • the AD and/or DDD moiety may be attached to either the N-terminal or C-terminal end of an effector protein or peptide.
  • site of attachment of an AD or DDD moiety to an effector moiety may vary, depending on the chemical nature of the effector moiety and the part(s) of the effector moiety involved in its physiological activity. Site-specific attachment of a variety of effector moieties may be performed using techniques known in the art, such as the use of bivalent cross-linking reagents and/or other chemical conjugation techniques.
  • DOCK-AND-LOCK ® (DNL ® ) technology has been used to produce a variety of complexes in assorted formats (Rossi et al., 2012, Bioconjug Chem 23 :309-23).
  • Bispecific hexavalent antibodies based on veltuzumab (anti-CD20) and epratuzumab (anti- CD22) were constructed by combining a stabilized (Fab) 2 fused to a dimerization and docking domain (DDD) with an IgG containing an anchor domain (AD) appended at the C- terminus of each heavy chain (C H 3-AD2-IgG) (Rossi et al., 2009, Blood 113, 6161-71).
  • DDD dimerization and docking domain
  • AD anchor domain
  • Fc-bsHexAbs Compared to mixtures of their parental mAbs, these Fc-based bsHexAbs, referred to henceforth as "Fc-bsHexAbs", induced unique signaling events (Gupta et al., 2010, Blood 116:3258-67), and exhibited potent cytotoxicity in vitro.
  • the Fc-bsHexAbs were cleared from circulation of mice approximately twice as fast as the parental mAbs (Rossi et al., 2009, Blood 113, 6161-71).
  • the Fc-bsHexAbs are highly stable ex vivo, it is possible that some dissociation occurs in vivo, for example by intracellular processing.
  • Fc-bsHexAbs lack CDC activity.
  • Fc-based immunocytokines have also been assembled as DNL® complexes, comprising two or four molecules of interferon-alpha 2b (IFNa2b) fused to the C-terminal end of the C H 3-AD2-IgG Fc (Rossi et al., 2009, Blood 114:3864-71; Rossi et al., 2010, Cancer Res 70:7600-09; Rossi et al., 2011, Blood 118: 1877-84).
  • the Fc-IgG-IFNa maintained high specific activity, approaching that of recombinant IFNa, and were remarkably potent in vitro and in vivo against non-Hodgkin lymphoma (NHL) xenografts.
  • NHL non-Hodgkin lymphoma
  • the Ti/2 of the Fc-IgG-IFNa in mice was longer than PEGylated IFNa, but half as long as the parental mAbs. Similar to the Fc-bsHexAbs, the Fc-IgG-IFNa dissociated in vivo over time and exhibited diminished CDC, but ADCC was enhanced.
  • AD or DDD sequences may be utilized. Exemplary DDD and AD sequences are provided below.
  • SHIQIPPGLTELLQGYTVEVLRQQPPDLVEFAVEYFTRLREARA SEQ ID NO: l
  • DDDl and DDD2 are based on the DDD sequence of the human Rlla isoform of protein kinase A.
  • the DDD and AD moieties may be based on the DDD sequence of the human RIa form of protein kinase A and a corresponding AKAP sequence, as exemplified in DDD3, DDD3C and AD3 below.
  • AD and/or DDD moieties may be utilized in construction of the DNL® complexes.
  • Rlla DDD sequence is the basis of DDD 1 and DDD2 disclosed above.
  • the four human PKA DDD sequences are shown below.
  • the DDD sequence represents residues 1-44 of Rlla, 1-44 of RIip, 12-61 of RIa and 13-66 of Rip. (Note that the sequence of DDD 1 is modified slightly from the human PKA Rlla DDD moiety.)
  • SHIQIPPGLTELLQGYTVEVGQQPPDLVDFAVEYFTRLREARRQ (SEQ ID NO: 10)
  • SHIOIPPGLTELLOGYTVEVLROOPPDLVEFAVEYFTRLREARA SEQ ID NO: l
  • the effect of the amino acid substitutions on AD moiety binding may also be readily determined by standard binding assays, for example as disclosed in Alto et al. (2003, Proc Natl Acad Sci USA 100:4445-50).
  • Alto et al. 2003, Proc Natl Acad Sci USA 100:4445-50
  • the AKAP-IS sequence was designed as a peptide antagonist of AKAP binding to PKA. Residues in the AKAP-IS sequence where substitutions tended to decrease binding to DDD are underlined in SEQ ID NO:3 below.
  • SEQ ID NO:3 The skilled artisan will realize that in designing sequence variants of the AD sequence, one would desirably avoid changing any of the underlined residues, while conservative amino acid substitutions might be made for residues that are less critical for DDD binding. It is noted that Figure 2 of Alto (2003) shows a number of amino acid substitutions that may be made, while retaining binding activity to DDD moieties, based on actual binding experiments.
  • the SuperAKAP-IS sequence may be substituted for the AKAP-IS AD moiety sequence to prepare DNL® constructs. It is anticipated that, as with the AD2 sequence shown in SEQ ID NO:4, the AD moiety may also include the additional N-terminal residues cysteine and glycine and C-terminal residues glycine and cysteine.
  • Figure 2 of Gold et al. disclosed additional DDD-binding sequences from a variety of AKAP proteins.
  • Carr et al. (2001, J Biol Chem 276: 17332-38) examined the degree of sequence homology between different AKAP -binding DDD sequences from human and non-human proteins and identified residues in the DDD sequences that appeared to be the most highly conserved among different DDD moieties. These are indicated below by underlining with reference to the human PKA Rlla DDD sequence of SEQ ID NO: 1. Residues that were particularly conserved are further indicated by italics. The residues overlap with, but are not identical to those suggested by Kinderman et al. (2006) to be important for binding to AKAP proteins.
  • the disclosed methods and compositions may involve production and use of proteins or peptides with one or more substituted amino acid residues.
  • the DDD and/or AD sequences used to make DNL® constructs may be modified as discussed above.
  • amino acid substitutions typically involve the replacement of an amino acid with another amino acid of relatively similar properties (i.e., conservative amino acid substitutions).
  • conservative amino acid substitutions The properties of the various amino acids and effect of amino acid substitution on protein structure and function have been the subject of extensive study and knowledge in the art.
  • the hydropathic index of amino acids may be considered (Kyte & Doolittle, 1982, J. Mol. Biol., 157: 105-132).
  • the relative hydropathic character of the amino acid contributes to the secondary structure of the resultant protein, which in turn defines the interaction of the protein with other molecules.
  • Each amino acid has been assigned a hydropathic index on the basis of its hydrophobicity and charge characteristics (Kyte & Doolittle, 1982), these are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cystine (+2.5); methionine (+1.9); alanine (+1.8); glycine (-0.4); threonine (- 0.7); serine (-0.8); tryptophan (-0.9); tyrosine (-1.3); proline (-1.6); histidine (-3.2); glutamate (-3.5); glutamine (-3.5); aspartate (-3.5); asparagine (-3.5); lysine (-3.9); and arginine (-4.5).
  • the use of amino acids whose hydropathic indices are within ⁇ 2 is preferred, within ⁇ 1 are more preferred, and within ⁇ 0.5 are even more preferred.
  • Amino acid substitution may also take into account the hydrophilicity of the amino acid residue (e.g., U.S. Pat. No. 4,554, 101). Hydrophilicity values have been assigned to amino acid residues: arginine (+3.0); lysine (+3.0); aspartate (+3.0); glutamate (+3.0); serine (+0.3); asparagine (+0.2); glutamine (+0.2); glycine (0); threonine (-0.4); proline (-0.5 .+-.1); alanine (-0.5); histidine (-0.5); cysteine (-1.0); methionine (-1.3); valine (-1.5); leucine (-1.8); isoleucine (-1.8); tyrosine (-2.3); phenylalanine (-2.5); tryptophan (-3.4). Replacement of amino acids with others of similar hydrophilicity is preferred.
  • amino acid side chain For example, it would generally not be preferred to replace an amino acid with a compact side chain, such as glycine or serine, with an amino acid with a bulky side chain, e.g., tryptophan or tyrosine.
  • a compact side chain such as glycine or serine
  • an amino acid with a bulky side chain e.g., tryptophan or tyrosine.
  • tryptophan or tyrosine The effect of various amino acid residues on protein secondary structure is also a
  • arginine and lysine glutamate and aspartate; serine and threonine; glutamine and asparagine; and valine, leucine and isoleucine.
  • amino acid substitutions include whether or not the residue is located in the interior of a protein or is solvent exposed.
  • conservative substitutions would include: Asp and Asn; Ser and Thr; Ser and Ala; Thr and Ala; Ala and Gly; Ile and Val; Val and Leu; Leu and Ile; Leu and Met; Phe and Tyr; Tyr and Trp.
  • conservative substitutions would include: Asp and Asn; Asp and Glu; Glu and Gin; Glu and Ala; Gly and Asn; Ala and Pro; Ala and Gly; Ala and Ser; Ala and Lys; Ser and Thr; Lys and Arg; Val and Leu; Leu and Ile; Ile and Val; Phe and Tyr.
  • amino acid substitutions In determining amino acid substitutions, one may also consider the existence of intermolecular or intramolecular bonds, such as formation of ionic bonds (salt bridges) between positively charged residues (e.g., His, Arg, Lys) and negatively charged residues (e.g., Asp, Glu) or disulfide bonds between nearby cysteine residues.
  • ionic bonds salt bridges
  • positively charged residues e.g., His, Arg, Lys
  • negatively charged residues e.g., Asp, Glu
  • disulfide bonds between nearby cysteine residues.
  • therapeutic agents such as cytotoxic agents, anti- angiogenic agents, pro-apoptotic agents, antibiotics, hormones, hormone antagonists, chemokines, drugs, prodrugs, toxins, enzymes or other agents may be used, either conjugated to the subject bsAbs, ADCs and/or antibodies or separately administered before, simultaneously with, or after the bsAbs, ADCs and/or antibodies.
  • Drugs of use may possess a pharmaceutical property selected from the group consisting of antimitotic, antikinase, alkylating, antimetabolite, antibiotic, alkaloid, anti-angiogenic, pro-apoptotic agents and combinations thereof.
  • Exemplary drugs of use may include, but are not limited to, 5-fluorouracil, afatinib, aplidin, azaribine, anastrozole, anthracyclines, axitinib, AVL-101, AVL-291, bendamustine, bleomycin, bortezomib, bosutinib, biyostatin-1, busulfan, calicheamycin, camptothecin, carboplatin, 10-hydroxycamptothecin, carmustine, Celebrex, chlorambucil, cisplatin (CDDP), Cox-2 inhibitors, irinotecan (CPT-11), SN-38, carboplatin, cladribine, camptothecans, crizotinib, cyclophosphamide, cytarabine, dacarbazine, dasatinib, dinaciclib, docetaxel, dactinomycin, daunorubicin
  • Toxins of use may include ricin, abrin, alpha toxin, saporin, ribonuclease (RNase), e.g., onconase, DNase I, Staphylococcal enterotoxin-A, pokeweed antiviral protein, gelonin, diphtheria toxin, Pseudomonas exotoxin, and Pseudomonas endotoxin.
  • RNase ribonuclease
  • Chemokines of use may include RANTES, MCAF, MlPl-alpha, MIPl-Beta and IP-10.
  • anti-angiogenic agents such as angiostatin, baculostatin, canstatin, maspin, anti-VEGF antibodies, anti-PlGF peptides and antibodies, anti-vascular growth factor antibodies, anti-Flk-1 antibodies, anti-Flt-1 antibodies and peptides, anti-Kras antibodies, anti-cMET antibodies, anti-MIF (macrophage migration-inhibitory factor) antibodies, laminin peptides, fibronectin peptides, plasminogen activator inhibitors, tissue metalloproteinase inhibitors, interferons, interleukin-12, IP-10, Gro-B, thrombospondin, 2- methoxyoestradiol, proliferin-related protein, carboxiamidotriazole, CM101, Marimastat, pentosan polysulphate, angiopoietin-2, interferon-alpha, herbimycin A, PNU145156E, 16K pro
  • Immunomodulators of use may be selected from a cytokine, a stem cell growth factor, a lymphotoxin, a hematopoietic factor, a colony stimulating factor (CSF), an interferon (IFN), erythropoietin, thrombopoietin and a combination thereof. Specifically useful are
  • lymphotoxins such as tumor necrosis factor (TNF), hematopoietic factors, such as interleukin (IL), colony stimulating factor, such as granulocyte-colony stimulating factor (G-CSF) or granulocyte macrophage-colony stimulating factor (GM-CSF), interferon, such as
  • interferons-a, - ⁇ or - ⁇ interferons-a, - ⁇ or - ⁇ , and stem cell growth factor, such as that designated "SI factor”.
  • cytokines include growth hormones such as human growth hormone, N- methionyl human growth hormone, and bovine growth hormone; parathyroid hormone;
  • thyroxine insulin; proinsulin; relaxin; prorelaxin; glycoprotein hormones such as follicle stimulating hormone (FSH), thyroid stimulating hormone (TSH), and luteinizing hormone (LH); hepatic growth factor; prostaglandin, fibroblast growth factor; prolactin; placental lactogen, OB protein; tumor necrosis factor-a and - B; mullerian-inhibiting substance; mouse gonadotropin-associated peptide; inhibin; activin; vascular endothelial growth factor;
  • FSH follicle stimulating hormone
  • TSH thyroid stimulating hormone
  • LH luteinizing hormone
  • thrombopoietin TPO
  • nerve growth factors such as NGF-B; platelet-growth factor; transforming growth factors (TGFs) such as TGF- a and TGF- B; insulin-like growth factor-I and -II; erythropoietin (EPO); osteoinductive factors; interferons such as interferon-a, - ⁇ , and - ⁇ ; colony stimulating factors (CSFs) such as macrophage-CSF (M-CSF); interleukins (ILs) such as IL-1, IL-la, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12; IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IL-21, IL-25, LIF, kit-ligand or FLT-3, angiostatin, thrombospondin, endostatin, tumor
  • Radionuclides of use include, but are not limited to- lu In, 177 Lu, 212 Bi, 213 Bi, 211 At, 62 Cu, 67 Cu, 90 Y, 125 I, 131 1, 32 P, 33 P, 47 Sc, m Ag, 67 Ga, 142 Pr, 153 Sm, 161 Tb, 166 Dy, 166 Ho, 186 Re, 188 Re, 189 Re, 212 Pb, 223 Ra, 225 Ac, 59 Fe, 75 Se, 77 As, 89 Sr, 99 Mo, 105 Rh, 109 Pd, 143 Pr, 149 Pm, 169 Er, 194 Ir, 198 Au, 199 Au, 211 Pb, and 227 Th.
  • the therapeutic radionuclide preferably has a decay-energy in the range of 20 to 6,000 keV, preferably in the ranges 60 to 200 keV for an Auger emitter, 100-2,500 keV for a beta emitter, and 4,000-6,000 keV for an alpha emitter.
  • Maximum decay energies of useful beta-particle-emitting nuclides are preferably 20- 5,000 keV, more preferably 100-4,000 keV, and most preferably 500-2,500 keV. Also preferred are radionuclides that substantially decay with Auger-emitting particles.
  • beta-particle-emitting nuclides are preferably ⁇ 1,000 keV, more preferably ⁇ 100 keV, and most preferably ⁇ 70 keV. Also preferred are radionuclides that substantially decay with generation of alpha-particles.
  • Such radionuclides include, but are not limited to: Dy-152, At-211, Bi-212, Ra-223, Rn-219, Po-215, Bi-211, Ac-225, Fr-221, At-217, Bi-213, Th-227 and Fm-255. Decay energies of useful alpha- particle-emitting radionuclides are preferably 2,000-10,000 keV, more preferably 3,000- 8,000 keV, and most preferably 4,000-7,000 keV.
  • radioisotopes of use include U C, 13 N, 15 0, 75 Br, 198 Au, 224 Ac, 126 I, 133 I, 77 Br, 113m In, 95 Ru, 97 Ru, 103 Ru, 105 Ru, 107 Hg, 203 Hg, 121m Te, 122m Te, 125m Te, 165 Tm, 167 Tm, 168 Tm, 197 Pt, 109 Pd, 105 Rh, 142 Pr, 143 Pr, 161 Tb, 166 Ho, 199 Au, 57 Co, 58 Co, 51 Cr, 59 Fe, 75 Se, 201 T1, 225 Ac, 76 Br, 169 Yb,
  • Some useful diagnostic nuclides may include F, Fe, Cu, Cu, Cu, Ga,
  • Therapeutic agents may include a photoactive agent or dye.
  • compositions such as fluorochrome, and other chromogens, or dyes, such as porphyrins sensitive to visible light, have been used to detect and to treat lesions by directing the suitable light to the lesion. In therapy, this has been termed photoradiation, phototherapy, or photodynamic therapy. See Jori et al. (eds ), PHOTODYNAMIC THERAPY OF TUMORS AND OTHER DISEASES (Libreria Progetto 1985); van den Bergh, Chem. Britain (1986), 22:430. Moreover, monoclonal antibodies have been coupled with photoactivated dyes for achieving phototherapy. See Mew et al., J. Immunol. (1983),130: 1473; idem., Cancer Res. (1985), 45:4380; Oseroff et al., Proc. Natl. Acad. Sci. USA (1986), 83 :8744; idem.,
  • oligonucleotides especially antisense oligonucleotides that preferably are directed against oncogenes and oncogene products, such as bcl-2 or p53.
  • a preferred form of therapeutic oligonucleotide is siRNA.
  • siRNA any siRNA or interference RNA species may be attached to an antibody or fragment thereof for delivery to a targeted tissue.
  • siRNA species against a wide variety of targets are known in the art, and any such known siRNA may be utilized in the claimed methods and compositions.
  • Known siRNA species of potential use include those specific for IKK-gamma (U.S.
  • amyloid beta precursor protein U.S. Patent 7,635,771
  • IGF-1R U.S. Patent 7,638,621
  • ICAM1 U.S. Patent 7,642,349
  • complement factor B U.S. Patent 7,696,344
  • p53 7,781,575)
  • apolipoprotein B 7,795,421
  • siRNA species are available from known commercial sources, such as Sigma-Aldrich (St Louis, MO), Invitrogen (Carlsbad, CA), Santa Cruz Biotechnology (Santa Cruz, CA), Ambion (Austin, TX), Dharmacon (Thermo Scientific, Lafayette, CO), Promega (Madison, WI), Minis Bio (Madison, WI) and Qiagen (Valencia, CA), among many others.
  • Other publicly available sources of siRNA species include the siRNAdb database at the Swedish Bioinformatics Centre, the MIT/ICBP siRNA Database, the RNAi Consortium shRNA Library at the Broad Institute, and the Probe database at NCBI.
  • siRNA species there are 30,852 siRNA species in the NCBI Probe database.
  • the skilled artisan will realize that for any gene of interest, either a siRNA species has already been designed, or one may readily be designed using publicly available software tools. Any such siRNA species may be delivered using the subject DNL® complexes.
  • Various embodiments concern methods of treating a cancer in a subject, such as a mammal, including humans, domestic or companion pets, such as dogs and cats, comprising administering to the subject a therapeutically effective amount of a combination of cytotoxic and/or immunomodulatory agents.
  • cytotoxic bsAbs, ADCs and/or immune checkpoint inhibitor antibodies can be supplemented by administering concurrently or sequentially a therapeutically effective amount of another antibody that binds to or is reactive with another antigen on the surface of the target cell.
  • Preferred additional MAbs comprise at least one humanized, chimeric or human MAb selected from the group consisting of a MAb reactive with CD4, CD5, CD8, CD14, CD15, CD16, CD19, IGF-1R, CD20, CD21, CD22, CD23, CD25, CD30, CD32b, CD33, CD37, CD38, CD40, CD40L, CD45, CD46, CD52, CD54, CD70, CD74, CD79a, CD79b, CD80, CD95, CD126, CD133, CD138, CD154, CEACAM5, CEACAM6, DLL-3, DLL-4, B7, AFP, PSMA, EGP-1, EGP-2, carbonic anhydrase IX, mesothelin, MUC1, MUC2, MUC3, MUC4, MUC5ac, la, MIF, HM1.24, HLA-DR, tenascin, Flt-3, VEGFR, PIGF, ILGF, IL-6, IL-25, tena
  • the combination therapy can be further supplemented with the administration, either concurrently or sequentially, of at least one therapeutic agent.
  • at least one therapeutic agent for example, "CVB" (1.5 g/m 2 cyclophosphamide, 200-400 mg/m 2 etoposide, and 150-200 mg/m 2 carmustine) is a regimen used to treat non-Hodgkin's lymphoma. Patti et al, Eur. J. Haematol. 57: 18 (1993).
  • Other suitable combination chemotherapeutic regimens are well-known to those of skill in the art.
  • first generation chemotherapeutic regimens for treatment of intermediate- grade non-Hodgkin's lymphoma include C-MOPP (cyclophosphamide, vincristine, procarbazine and prednisone) and CHOP (cyclophosphamide, doxorubicin, vincristine, and prednisone).
  • a useful second generation chemotherapeutic regimen is m-BACOD
  • a suitable third generation regimen is MACOP-B (methotrexate, doxorubicin, cyclophosphamide, vincristine, prednisone, bleomycin and leucovorin).
  • Additional useful drugs include phenyl butyrate, bendamustine, and biyostatin-1.
  • the combinations of therapeutic agents can be formulated according to known methods to prepare pharmaceutically useful compositions, whereby the bsAb, ADC, interferon and/or checkpoint inhibitor antibody is combined in a mixture with a
  • Sterile phosphate-buffered saline is one example of a pharmaceutically suitable excipient.
  • Other suitable excipients are well-known to those in the art. See, for example, Ansel et al, PHARMACEUTICAL DOSAGE FORMS AND DRUG DELIVERY SYSTEMS, 5th Edition (Lea & Febiger 1990), and Gennaro (ed.), REMINGTON'S PHARMACEUTICAL SCIENCES, 18th Edition (Mack Publishing Company 1990), and revised editions thereof.
  • the subject bsAbs, ADCs, interferons and/or antibodies can be formulated for intravenous administration via, for example, bolus injection or continuous infusion.
  • the bsAb, ADC and/or antibody is infused over a period of less than about 4 hours, and more preferably, over a period of less than about 3 hours.
  • the first bolus could be infused within 30 minutes, preferably even 15 min, and the remainder infused over the next 2-3 hrs.
  • Formulations for injection can be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions can take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient can be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • a suitable vehicle e.g., sterile pyrogen-free water
  • Control release preparations can be prepared through the use of polymers to complex or adsorb the agents to be administered.
  • biocompatible polymers include matrices of poly(ethylene-co-vinyl acetate) and matrices of a polyanhydride copolymer of a stearic acid dimer and sebacic acid. Sherwood et al,
  • the bsAbs, interferons and/or checkpoint inhibitor antibodies may be administered to a mammal subcutaneously or even by other parenteral routes, such as intravenously, intramuscularly, intraperitoneally or intravascularly.
  • ADCs may be administered
  • the administration may be by continuous infusion or by single or multiple boluses.
  • the bsAb, ADC, interferon and/or checkpoint inhibitor antibody is infused over a period of less than about 4 hours, and more preferably, over a period of less than about 3 hours.
  • the dosage of an administered bsAb, ADC, interferon and/or checkpoint inhibitor antibody for humans will vary depending upon such factors as the patient's age, weight, height, sex, general medical condition and previous medical history and treatments. It may be desirable to provide the recipient with a dosage of bsAb, ADC and/or antibody that is in the range of from about 1 mg/kg to 25 mg/kg as a single intravenous infusion, although a lower or higher dosage also may be administered as circumstances dictate. A dosage of 1-20 mg/kg for a 70 kg patient, for example, is 70-1,400 mg, or 41-824 mg/m 2 for a 1.7-m patient.
  • the dosage may be repeated as needed, for example, once per week for 4-10 weeks, once per week for 8 weeks, or once per week for 4 weeks. It may also be given less frequently, such as every other week for several months, or monthly or quarterly for many months, as needed in a maintenance therapy.
  • a bsAb, ADC, and/or checkpoint inhibitor antibody may be any suitable bsAb, ADC, and/or checkpoint inhibitor antibody.
  • the combination may be administered twice per week for 4-6 weeks. If the dosage is lowered to approximately 200-300 mg/m 2 (340 mg per dosage for a 1.7-m patient, or 4.9 mg/kg for a 70 kg patient), it may be administered once or even twice weekly for 4 to 10 weeks.
  • the dosage schedule may be decreased, namely every 2 or 3 weeks for 2-3 months. It has been determined, however, that even higher doses, such as 20 mg/kg once weekly or once every 2-3 weeks can be administered by slow i.v. infusion, for repeated dosing cycles.
  • the dosing schedule can optionally be repeated at other intervals and dosage may be given through various parenteral routes, with appropriate adjustment of the dose and schedule.
  • interferon agents should be administered at substantially lower dosages to avoid systemic toxicity.
  • Dosages of interferons are more typically in the microgram range, for example 180 ⁇ g s.c. once per week, or 100 to 180 ⁇ g, or 135 ⁇ g, or 135 ⁇ g/1.73 m 2 , or 90 ⁇ g/1.73 m 2 , or 250 ⁇ g s.c. every other day may be of use, depending on the type of interferon.
  • the bsAbs, interferons, ADCs and/or checkpoint inhibitor antibodies may be administered as a periodic bolus injection
  • the bsAbs, ADCs, interferons and/or checkpoint inhibitor antibodies may be administered by continuous infusion.
  • a continuous infusion may be administered for example by indwelling catheter.
  • indwelling catheter Such devices are known in the art, such as HICKMAN®, BROVIAC® or PORT-A-CATH® catheters (see, e.g., Skolnik et al., Ther Drug Monit 32:741-48, 2010) and any such known indwelling catheter may be used.
  • a variety of continuous infusion pumps are also known in the art and any such known infusion pump may be used.
  • the dosage range for continuous infusion may be between 0.1 and 3.0 mg/kg per day. More preferably, the bsAbs, ADCs, interferons and/or checkpoint inhibitor antibodies can be administered by intravenous infusions over relatively short periods of 2 to 5 hours, more preferably 2-3 hours.
  • the combination of agents is of use for therapy of cancer.
  • cancers include, but are not limited to, carcinoma, lymphoma, glioblastoma, melanoma, sarcoma, and leukemia, myeloma, or lymphoid malignancies.
  • squamous cell cancer e.g., epithelial squamous cell cancer
  • Ewing sarcoma e.g., Ewing sarcoma
  • Wilms tumor astrocytomas
  • lung cancer including small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma multiforme, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, hepatocellular carcinoma, neuroendocrine tumors, medullary thyroid cancer, differentiated thyroid carcinoma, breast cancer, ovarian cancer, colon cancer, rectal cancer, endometrial cancer or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulvar cancer, anal carcinoma, penile carcinoma, as well as head-and-neck cancer.
  • cancer includes primary malignant cells or tumors (e.g., those whose cells have not migrated to sites in the subject's body other than the site of the original malignancy or tumor) and secondary malignant cells or tumors (e.g., those arising and spreading as metastasis, the migration of malignant cells or tumor cells to secondary sites that are different from the site of the original tumor).
  • primary malignant cells or tumors e.g., those whose cells have not migrated to sites in the subject's body other than the site of the original malignancy or tumor
  • secondary malignant cells or tumors e.g., those arising and spreading as metastasis, the migration of malignant cells or tumor cells to secondary sites that are different from the site of the original tumor.
  • Cancers conducive to treatment methods of the present invention involves cells which express, over-express, or abnormally express IGF-1R.
  • cancers or malignancies include, but are not limited to: Acute Childhood Lymphoblastic Leukemia, Acute Lymphoblastic Leukemia, Acute Lymphocytic Leukemia, Acute Myeloid Leukemia, Adrenocortical Carcinoma, Adult (Primary)
  • Astrocytoma Childhood Cerebral Astrocytoma, Childhood Extracranial Germ Cell Tumors, Childhood Hodgkin's Disease, Childhood Hodgkin's Lymphoma, Childhood Hypothalamic and Visual Pathway Glioma, Childhood Lymphoblastic Leukemia, Childhood
  • Metastatic Occult Primary Squamous Neck Cancer Metastatic Primary Squamous Neck Cancer, Metastatic Primary Squamous Neck Cancer, Metastatic Squamous Neck Cancer, Multiple Myeloma, Multiple Myeloma/Plasma Cell Neoplasm, Myelodysplastic Syndrome, Myelogenous Leukemia, Myeloid Leukemia, Myeloproliferative Disorders, Nasal Cavity and Paranasal Sinus Cancer, Nasopharyngeal Cancer, Neuroblastoma, Non-Hodgkin's Lymphoma, Nonmelanoma Skin Cancer, Non-Small Cell Lung Cancer, Occult Primary Metastatic Squamous Neck Cancer, Oropharyngeal Cancer, Osteo-/Malignant Fibrous Sarcoma, Osteosarcoma/Malignant Fibrous Histiocytoma, Osteosarcoma/Malignant Fibrous Histiocytoma of Bone, Ovarian Epithelial Cancer, O
  • Pheochromocytoma Pituitary Tumor, Primary Central Nervous System Lymphoma, Primary Liver Cancer, Prostate Cancer, Rectal Cancer, Renal Cell Cancer, Renal Pelvis and Ureter Cancer, Retinoblastoma, Rhabdomyosarcoma, Salivary Gland Cancer, Sarcoidosis Sarcomas, Sezary Syndrome, Skin Cancer, Small Cell Lung Cancer, Small Intestine Cancer, Soft Tissue Sarcoma, Squamous Neck Cancer, Stomach Cancer, Supratentorial Primitive
  • Neuroectodermal and Pineal Tumors T-Cell Lymphoma, Testicular Cancer, Thymoma, Thyroid Cancer, Transitional Cell Cancer of the Renal Pelvis and Ureter, Transitional Renal Pelvis and Ureter Cancer, Trophoblastic Tumors, Ureter and Renal Pelvis Cell Cancer, Urethral Cancer, Uterine Cancer, Uterine Sarcoma, Vaginal Cancer, Visual Pathway and Hypothalamic Glioma, Vulvar Cancer, Waldenstrom's Macroglobulinemia, Wilms' Tumor, and any other hyperproliferative disease, besides neoplasia, located in an organ system listed above.
  • compositions described and claimed herein may be used to treat malignant or premalignant conditions and to prevent progression to a neoplastic or malignant state, including but not limited to those disorders described above.
  • Such uses are indicated in conditions known or suspected of preceding progression to neoplasia or cancer, in particular, where non-neoplastic cell growth consisting of hyperplasia, metaplasia, or most particularly, dysplasia has occurred (for review of such abnormal growth conditions, see Robbins and Angell, BASIC PATHOLOGY, 2d Ed., W. B. Saunders Co., Philadelphia, pp. 68-79 (1976)).
  • Dysplasia is frequently a forerunner of cancer, and is found mainly in the epithelia. It is the most disorderly form of non-neoplastic cell growth, involving a loss in individual cell uniformity and in the architectural orientation of cells. Dysplasia characteristically occurs where there exists chronic irritation or inflammation.
  • Dysplastic disorders which can be treated include, but are not limited to, anhidrotic ectodermal dysplasia, anterofacial dysplasia, asphyxiating thoracic dysplasia, atriodigital dysplasia, bronchopulmonary dysplasia, cerebral dysplasia, cervical dysplasia, chondroectodermal dysplasia, cleidocranial dysplasia, congenital ectodermal dysplasia, craniodiaphysial dysplasia, craniocarpotarsal dysplasia, craniometaphysial dysplasia, dentin dysplasia, diaphysial dysplasia, ectodermal dysplasia, enamel dysplasia, encephalo-ophthalmic dysplasia, dysplasia epiphysialis hemimelia, dysplasia epiphysialis multiplex, dysplasia epiphysialis punctata, epi
  • pseudoachondroplastic spondyloepiphysial dysplasia retinal dysplasia, septo-optic dysplasia, spondyloepiphysial dysplasia, and ventriculoradial dysplasia.
  • Additional pre-neoplastic disorders which can be treated include, but are not limited to, benign dysproliferative disorders (e.g., benign tumors, fibrocystic conditions, tissue hypertrophy, intestinal polyps or adenomas, and esophageal dysplasia), leukoplakia, keratoses, Bowen's disease, Farmer's Skin, solar cheilitis, and solar keratosis.
  • benign dysproliferative disorders e.g., benign tumors, fibrocystic conditions, tissue hypertrophy, intestinal polyps or adenomas, and esophageal dysplasia
  • leukoplakia keratoses
  • Bowen's disease keratoses
  • Farmer's Skin Farmer's Skin
  • solar cheilitis solar keratosis
  • the method of the invention is used to inhibit growth, progression, and/or metastasis of cancers, in particular those listed above.
  • Additional hyperproliferative diseases, disorders, and/or conditions include, but are not limited to, progression, and/or metastases of malignancies and related disorders such as leukemia (including acute leukemias (e.g., acute lymphocytic leukemia, acute myelocytic leukemia (including myeloblastic, promyelocytic, myelomonocytic, monocytic, and erythroleukemia)) and chronic leukemias (e.g., chronic myelocytic (granulocytic) leukemia and chronic lymphocytic leukemia)), polycythemia vera, lymphomas (e.g., Hodgkin's disease and non-Hodgkin's disease), multiple myeloma, Waldenstrom's macroglobulinemia, heavy chain disease, and solid tumors including, but not limited to, sarcomas and carcinomas such as fibrosarcoma, myxosarcoma, lipos
  • lymphangioendotheliosarcoma synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, testicular tumor, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma,
  • Still other embodiments may concern DNA sequences comprising a nucleic acid encoding an antibody, antibody fragment, cytokine or constituent fusion protein of a bsAb, such as a DNL® construct. Fusion proteins may comprise an antibody or fragment or cytokine attached to, for example, an AD or DDD moiety.
  • Various embodiments relate to expression vectors comprising the coding DNA sequences.
  • the vectors may contain sequences encoding the light and heavy chain constant regions and the hinge region of a human immunoglobulin to which may be attached chimeric, humanized or human variable region sequences.
  • the vectors may additionally contain promoters that express the encoded protein(s) in a selected host cell, enhancers and signal or leader sequences. Vectors that are particularly useful are pdHL2 or GS.
  • the light and heavy chain constant regions and hinge region may be from a human EU myeloma immunoglobulin, where optionally at least one of the amino acid in the allotype positions is changed to that found in a different IgGl allotype, and wherein optionally amino acid 253 of the heavy chain of EU based on the EU number system may be replaced with alanine.
  • an IgGl sequence may be converted to an IgG4 sequence.
  • kits containing components suitable for treating or diagnosing diseased tissue in a patient.
  • Exemplary kits may contain one or more bsAbs, ADCs, interferons, and/or checkpoint inhibitor antibodies as described herein.
  • a device capable of delivering the kit components through some other route may be included.
  • One type of device, for applications such as parenteral delivery, is a syringe that is used to inject the composition into the body of a subject. Inhalation devices may also be used.
  • a therapeutic agent may be provided in the form of a prefilled syringe or autoinjection pen containing a sterile, liquid formulation or lyophilized preparation.
  • the kit components may be packaged together or separated into two or more containers.
  • the containers may be vials that contain sterile, lyophilized formulations of a composition that are suitable for reconstitution.
  • a kit may also contain one or more buffers suitable for reconstitution and/or dilution of other reagents.
  • Other containers that may be used include, but are not limited to, a pouch, tray, box, tube, or the like. Kit components may be packaged and maintained sterilely within the containers.
  • Another component that can be included is instructions to a person using a kit for its use.
  • DNL® complexes Several species of exemplary leukocyte redirecting bispecific antibodies were made as DNL® complexes, as described below. The complexes were effective to induce an immune response against appropriate target cells.
  • DOCK-AND-LOCK® DOCK-AND-LOCK®
  • DNL® DOCK-AND-LOCK®
  • An exemplary leukocyte redirecting bispecific antibody with binding sites for CD3 and CD 19 was made as a DNL® complex, referred to as (19)-3s (FIG. 1).
  • An anti-CD 19 F(ab) 2 DNL module was constructed by recombinant fusion of a dimerization and docking domain (DDD2) at the carboxyl terminal end of the Fd chain.
  • DDD2 dimerization and docking domain
  • An anti-CD3-scFv module was designed from Okt3 mAb with addition of an anchor domain (AD2) and assembled in the format V H -L1-V K -L2-6H-L3-AD2 ("6H" disclosed as SEQ ID NO: 13), where the V domains were fused via a flexible peptide linker and the AD2 peptide was preceded by a 6-His linker (SEQ ID NO: 13).
  • the sequences of the anti-CD3 variable regions, linkers and AD2 were as shown below.
  • Expression vectors and D L® modules - D L® complexes were constructed comprising antibody moieties against various disease-associated antigens, linked to an anti- CD3 antibody moiety, generally abbreviated as (X)-3s bsAbs.
  • Independent production cell lines were developed in SpESFX-10 mouse myeloma cells (Rossi et al., 2011, Biotechnol Prog 27:766-75) for each of the DNL® modules used to make the (X)-3s bsAbs.
  • a cDNA sequence encoding the Okt3scFv-AD2 polypeptide (SEQ ID NOs:22-27) was synthesized and cloned into the pdHL2 expression vector via 5' Xba I and 3' Eag I restriction sites.
  • the construct comprised the VH domain fused to the VL in an scFv with the structure VH-L1-VK- L2-6H-L3-AD2 ("6H" disclosed as SEQ ID NO: 13).
  • the expressed protein had two amino acid substitutions from the original Okt3 mAb.
  • a cysteine residue in the CDR-H3 was changed to serine (Kipryanov, 1997, J Immunol Methods 200:69-77).
  • the penultimate residue of the VL was changed from aspartate to lysine.
  • the Okt3scFv-AD2 module was combined with various C H l-DDD2-Fab modules to generate a panel of (X)-3s trivalent bsAbs (Table 2).
  • the C H l-DDD2-Fab-pdHL2 expression vectors were constructed as described previously for similar constructs (Rossi et al., 2008, Cancer Res 68:8384-92).
  • expression vectors encoding Cnl-DDD2-Fab were generated from the corresponding IgG-pdHL2 expression vectors by excising the coding sequence for the C H l-Hinge-C H 2-C H 3 domains with Sac II and Eag I restriction enzymes and replacing it with a 507 bp sequence encoding CH1-DDD2, which was excised from the CHI - DDD2-Fab-hA20-pdHL2 expression vector (Rossi et al., 2008, Cancer Res 68:8384-92) with the same enzymes.
  • C H l-DDD2-Fab modules were derived from the humanized mAbs hA19 (anti-CD 19), labetuzumab (hMN-14, anti-CEACAM5), clivatuzumab (hPAM4, anti-mucin), hMN-15 (anti-CEACAM6), hRS7 (anti-Trop-2), veltuzumab (hA20, anti-CD20), hL243 (anti-HLA-DR) and epratuzumab (hLL2, anti-CD22).
  • the mAb designated hA19 was humanized from the mouse anti-CD19 mAb B43 (Uckun et al., 1988, Blood 71 : 13-29). Each expression vector was linearized by digestion with Sal I restriction enzyme and used to transfect SpESFX-10 cells by electroporation.
  • Clones were selected in media containing 0.2 ⁇ methotrexate (MTX) and screened for protein expression by ELISA.
  • Okt3scFv-AD2 was captured on Ni-NTA HisSorb plates (Qiagen) and detected with an anti-AD2 mAb.
  • C H l-DDD2-Fab modules were captured with goat-anti-human-kappa chain and detected with goat-anti-human-F(ab') 2 -HRP.
  • Productivity of protein-expression was amplified by stepwise increases in MTX concentration up to 3 ⁇ .
  • Okt3scFv-AD2 and C H l-DDD2-Fab modules were purified to homogeneity from the broth of roller bottle cultures by affinity chromatography using Ni-SEPHAROSE® and Kappa-Select resins, respectively.
  • the D L® method was used to assemble (X)-3s bsAbs via the site- specific conjugation of mole equivalents of Okt3scFv-AD2 and C H l-DDD2-Fab modules.
  • (19)-3s were produced by combining 22 mg of Okt3scFv-AD2 with 80 mg of C H l-DDD2-Fab-hA19.
  • the mixture was reduced overnight at room temperature with 1 mM reduced glutathione prior to the addition of 2 mM oxidized glutathione.
  • the (19)-3s was purified from the reaction mixture by sequential affinity chromatography with Kappa-Select and Ni- SEPHAROSE®. Additional (X)-3s constructs were assembled at various scales following a similar process.
  • Cell Lines and Reagents - Raji, Ramos, Daudi, LS174T and Capan-1 cell lines were purchased from the American Type Cell Culture Collection (ATCC, Manassas, MD) and Nalm-6 cells were purchased from Deutsche Sammlung von Mikroorganismen und Zellinien (DSMZ, Braunchweig, Germany). All cell lines, except Capan-1, were maintained in RPMI- 1640 containing 10% FBS, 1% L-glutamine, 1% penicillin-streptomycin and 1% MEM nonessential amino acids. Capan-1 cells were maintained with 20% FBS. All cell culture media and supplements were purchased from Life Technologies (Carlsbad, CA).
  • PBMCs and T cell isolation Human peripheral blood mononuclear cells (PBMC) were purified from whole donor blood (Blood Center of NJ, East Orange, NJ) using UNI- SEP M A XI tubes (Novamed, Ltd, Jerusalem, Israel).
  • CD3-positive T cells were isolated from PBMCs by negative selection using the Pan T Cell Isolation Kit (Miltenyi Biotec, Auburn, CA), according to the manufacturer's protocol. Efficiency of T cell isolation was assessed by FACS after staining the enriched T cells with anti-CD3-PE antibody. In some cases, further staining with CD- 19 and CD- 14 was performed as well to identify contaminating cells.
  • T cell activation - Isolated T cells were plated in 6-well tissue culture plates at a final density of 2.25 x 10 6 cells/well. Daudi cells were added to some wells at a final density of 1.5 x 10 6 cells/well, other wells were left to contain only T cells. Alternatively, PBMCs were added to 6-well tissue culture plates at a final cell density of 6 x 10 6 cells/well. The volume of each well was brought up to 3 mL. To the appropriate wells, 3 ng/mL of (19)-3s, (Ml)-3s or (19)-DDD2 was added. After incubation overnight at 37°C, 1 mL of each sample was removed. The cells were pelleted and labeled on ice with CD69-APC and CD3-PE for 20 minutes. Cells were washed 2 times with 1% BSA in PBS and analyzed using a
  • FACSCALIBERTM flow cytometer (BD Biosciences, San Jose, CA).
  • T-cell proliferation - PBMCs were seeded in T25 flasks at a concentration of 1 x 10 6 cells/mL containing the specified reagents.
  • B cells were removed by negative selection using a B-cell isolation kit from Miltenyi according to manufacturer's protocol.
  • 100 ⁇ of media was removed from each flask, labeled with anti- CD7-APC for 20 minutes on ice, washed once and resuspended in 300 uL of 1% BS A/PBS containing 7-AAD.
  • the entire volume is analyzed using a FACSCALIBERTM flow cytometer. Each sample is counted in duplicate. Analysis is performed using FlowJo Software. For each sample, dead (7-AAD+) cells, and debris (based on forward vs. side scatter) was removed. Finally, live CD7+ cells were selected and plotted using Prism software.
  • Labeled Capan-1 cells were added to 8- well chamber slides (ThermoWaltham, MA) and allowed to attach overnight. The following day, media was removed and PKH26-labeled Jurkat cells were added in media containing 0.1 ⁇ g/mL of (El)-3s, (Ml)-3s or (19)-3s. Following a 1-hour incubation at 37°C, slides were washed with PBS to remove any unbound cells and observed by fluorescence microscopy.
  • Cytotoxicity Assay Hematologic Tumor Cell Lines
  • Target cells were labeled with PKH67 Green Fluorescent Cell Linker Kit (Sigma) according to the manufacturer's protocol. Briefly, 5 x 10 6 target cells were resuspended in 250 ⁇ of diluent C. In a second tube 1 ⁇ L ⁇ of PKH26 dye is added to 250 ⁇ of diluent C. The cell suspension is then added to the dye solution, mixed thoroughly and incubated at RT for 2 minutes. The reaction was quenched by adding an equal volume of FBS. The labeled cells were then washed 3 times with complete RPMI. Unstimulated, isolated T cells were used as effector cells.
  • Effector cells and PKH67- labeled target cells were combined at a 10: 1 ratio and plated in 48-well plates containing serial dilutions of (19)-3s or (14)-3s. Each well contained 5 x 10 4 target cells and 5 x 10 5 effector cells. Jeko-1 assays were performed in 20% RPMI. Plates were incubated for 18 -24 hours in a 37°C incubator containing 5% C0 2 . Following incubation, all cells were removed from 48-well plates into flow cytometer tubes and resuspended in 1% BS A/PBS containing 1 ug/mL of 7AAD, to distinguish live from dead cells, and 30,000 COUNTBRIGHTTM
  • Effector cells used were as follows: For Capan-1 assays, CD8+ enriched T cells were used, following purification from a CD8+ enrichment column (R&D Systems, Minneapolis, MN).
  • For LS174T cells Stimulated T cells were used after incubation of PBMC for 5 days in media containing 25 U/mL IL-2 and 50 ng/mL Okt3 Mab, followed by 2 days incubation in media containing 25 U/mL IL-2 alone.
  • Effector cells and PKH67-labeled target cells were combined at a 3 : 1 ratio (5xl0 4 target cells and 1.5xl0 5 effector cells/well) and plated over 48- well plates containing serial dilutions of (El)-3s, (14)-3s or (19)-3s.
  • Capan-1 assays were performed in 20% RPMI. Plates were incubated for 42 - 48 hours in a 37°C incubator containing 5% C0 2 . Following incubation, suspension cells were combined with trypsinized attached cells from all wells and transferred into flow cytometer tubes.
  • COUNTB RIGHTTM beads were counted as a normalized reference. Data were analyzed using FlowJo software (Treestar, Inc., Ashland, OR). For each sample, dead cells and debris were excluded and total live target cells were counted.
  • mice Female NOD/SCID mice, 8 weeks old, were purchased from Charles River (Wilmington, MA). Mice were injected s.c. with a mixture of Raji (lxlO 6 ) and human PBMCs (5xl0 6 cells) mixed 1 : 1 with matrigel. Therapy began 1 hour later. Treatment regimens, dosages, and number of animals in each experiment are described in the Results. Animals were monitored daily for signs of tumor out-growth. Once tumors appeared, they were measured twice weekly. Tumor volume (TV) was determined by measurements in two dimensions using calipers, with volumes defined as: L x w 2 /2, where L is the longest dimension of the tumor and w the shortest.
  • TTI Tumor volume
  • Efficacy was determined by a log-rank test using Prism GraphPad software (v5; LaJolla, CA) on Kaplan-Meier curves using survival surrogate endpoints as time for tumor progression (TTP) to 1.0 cm 3 . Significance was considered at P ⁇ 0.05.
  • FIG. 3 demonstrates that the target lymphoma cells are in direct contact with the targeted T cells.
  • a dose-response series was performed for (19)-3s mediated association of T cells to an exemplary B-cell lymphoma line (FIG. 4). As shown in FIG. 4, under the conditions of this experiment, saturation of (19)-3s-mediated cell-to-cell association of T cells to target cells was reached at a concentration between 0.037 and 0.111 ⁇ g/ml of the DNL® complex.
  • FIG. 5 shows a comparision of the relative efficacies of BITE® (FIG. 5A), DARTTM (FIG. 5A) and DNL® (FIG. 5B) anti-CD3 x anti-CD 19 complexes for redirecting T cells to targeted CD19 + B cells.
  • BITE® and DARTTM was obtained from Moore et al. (2011, Blood 117:4542-51).
  • the (19)-3s DNL® complex was more effective than BITE® or DARTTM at targeting T cells to B-cell lymphoma (FIG. 5).
  • the (19)-3s DNL® complex also induced a slightly higher maximum level of cell-to-cell association than the comparable BITE® and DARTTM complexes (FIG. 5A). Although difficult to extrapolate from the single data points generated for the (19)-3s DNL® complex, the EC 50 levels appeared to be similar for BITE®, DARTTM and DNL® (FIG. 5)
  • Jurkat T cells were coincubated with target tumor cells containing (X)-3s and evaluated by flow cytometry and fluorescence microscopy.
  • Jurkat T cells are a CD4+ T cell leukemia line, chosen for their ability to demonstrate T cell binding without depletion of the FITC labeled Daudi cells in the presence of various concentrations of (19)-3s and analyzed by flow cytometry for the detection of double positive
  • Capan-1 expresses high levels of TROP2 and moderate levels of MUC5AC.
  • both the TROP2-targeting bsAb, (El)-3s (FIG. 6C), and MUC5AC -targeting bsAb, (Ml)-3s (FIG. 6B) were compared to the non-targeting control bsAb, (19)-3s (FIG. 6A).
  • CFSE-labeled Capan-1 cells were coincubated with PKH26-labeled Jurkat in the presence of these bsAbs. Fluorescent microscopy revealed, as expected, large T-cell/Capan complexes mediated by (El)-3s, followed by smaller, yet substantial complexes mediated by (Ml)-3s and relatively low complex formation following (19)-3s treatment (FIG. 6).
  • (19)-3s specifically induces T cell activation and proliferation.
  • the ability of (19)-3s to activate T cells was evaluated either in PBMCs (FIG. 7A), or T cells coincubated with Daudi B cells (FIG. 7B), by measuring the expression levels of CD69, an early marker of T cell activation.
  • Treatment with 3 ng/mL of (19)-3s induced T cell activation in T cells coincubated with Daudi B cells as indicated by a >50-fold increase in CD69 expression compared with non-targeting control antibodies, (19)-DDD2 and (Ml)-3s, as well as T cells treated with (19)-3s without Daudi target cells (FIG. 7B).
  • T cell proliferation was evaluated after treatment of PBMCs with various CD3-targeting antibodies. (19)-3s at 3 nM or 30 pM induced T cell proliferation similar to that of the positive control IL-2/PHA (FIG. 8A).
  • Non- targeting control antibody, (14)-3s shows some non-specific T cell proliferation at the highest (3 nM) concentration (FIG. 8A).
  • T cell proliferation was not observed in PBMCs depleted of B cells (FIG. 8B), suggesting that target cells are necessary for specific (19)-3s induced T cell proliferation.
  • (X)-3s re-directed T-cell mediated killing of malignant cell lines.
  • the cytotoxicity of each leukocyte targeting molecule was evaluated by its ability to mediate lysis of specific tumor target cells.
  • the bsAbs induced potent T cell-mediated cytotoxicity in various B cell malignancies, including Burkitt lymphoma (Daudi, Ramos, Namalwa) mantle cell lymphoma (Jeko-1) and acute
  • lymphoblastic leukemia (Nalm-6) (Table 3).
  • (19)-3s, (20)-3s, (22)-3s and (C2)-3s bind to T cells and target B cells simultaneously and induce T-cell-mediated killing in vitro.
  • the modular nature of the DNL method allowed the rapid production of several related conjugates for redirected leukocyte killing of various B cell malignancies, without the need for additional recombinant engineering and protein production.
  • the close proximity of the CD20 extracellular epitope to the cell surface resulted in the highest potency for (20)-3s.
  • the in vitro cytotoxic effects of leukocyte redirecting bsAbs were also determined in solid tumor cells (FIG. 11).
  • optimal assay conditions were determined to be a 3 : 1 E:T ratio using stimulated T cells in a 42 - 48 hour assay.
  • Each bsAb induced specific T-cell mediated lysis of the tumor target cells.
  • FIG. 12 A summary of the in vitro cytotoxicity data for various leukocyte redirecting bsAbs in a variety of tumor cell lines is shown in FIG. 12. The various constructs showed a maximal cell lysis of up to 90% or more of the targeted tumor cells, with IC 50 values for cell lines expressing the targeted antiben that were generally in the low picomolar range (FIG. 12).
  • FIG. 13C The untreated group (FIG. 13A), which was inoculated with the same cell mixture but did not receive (19)-3s, had a median survival time (MST) of 31 days.
  • MST median survival time
  • FIG. 14 A follow-up study was begun to determine the efficacy of less frequent dosing (FIG. 14).
  • Groups of 9 NOD/SCID mice were inoculated with Raji and PBMCs in a similar fashion as above. In this study, therapy was extended to two weeks, compared to one week in the first study. Groups received i.v. injections of (19)-3s totaling 360 ⁇ g as 2 x 130 ⁇ g (FIG. 14B), 4 x 65 ⁇ g (FIG. 14D) or 6 x 43 ⁇ g doses over two weeks (FIG. 14E). An additional group was administered 2 x 130 ⁇ g doses SC, instead of i.v. (FIG. 14C). For comparison, control groups of untreated mice (FIG.
  • mice treated with non-targeting (Ml)-3s antibody were prepared.
  • each of the (19)-3s treatment groups had significantly smaller AUC than the untreated control (P ⁇ 0.05).
  • two weekly doses via the SC route was apparently as effective as greater frequency i.v. dosing.
  • FIG. 15 In vivo studies were also performed using solid tumors (FIG. 15). NOD/SCID mouse xenografts were prepared as described above, for the LS174T colon adenocarcinoma (FIG. 15A, FIG. 15B) or Capan-1 pancreatic carcinoma (FIG. 15C, FIG. 15D). In each case, mice administered the targeting (El)-3s (FIG. 15B) or (14)-3s (FIG. 15D) bsAb DNL® constructs showed improved survival compared to controls.
  • the leukocyte-retargeting bsAbs including (19)-3s, (El)-3s and (Ml)- 3s DNL® constructs, mediated synapse formation between T cells and B cells, colon adenocarcinoma or pancreatic carcinoma cells, respectively, via monovalent and bivalent binding to CD3 and CD 19, respectively.
  • T-cell activation, proliferation and target cell killing were induced by the DNL® bsAbs at pM concentrations in an ex vivo setting.
  • Advantageous properties of the DNL® bsAbs, including bivalent tumor binding and slower clearance, would allow for less frequent dosing and possibly SC administration, compared to BITE® or DARTTM constructs, which are administered i.v.
  • DNL® method allows the rapid production of a large number of related conjugates for redirected leukocyte killing of various malignancies, without the need for additional recombinant engineering and protein production.
  • mice Five week-old female NOD/SCID mice were injected s.c. with a mixture of Capan-1 (5xl0 6 ) and human T-cells (2.5xl0 6 cells) mixed 1 : 1 with matrigel (E:T ratio of 1 :2). There were six different treatment groups of 8 mice each. Treatment consisted of one group receiving 47 ⁇ g (El)-3s i.v. every day for five days starting 1 hour after the administration of the Capan-1 /T-cell mixture. Two groups were treated with equimolar amounts of IFN, one received the DNL molecule made from IFN-a2b-DDD2-CK-hRS7 IgGl (El *-2b; 2.5 ⁇ g s.c.
  • mice were monitored daily for signs of tumor out-growth. All animals had their tumors measured twice weekly once tumors began to come up. Mice were euthanized for disease progression if their tumor volumes exceeded 1.0 cm 3 in size.
  • FIG. 16B Mean tumor volumes for the various groups are shown in FIG. 16.
  • the data containing PEGASYS® groups (FIG. 16B) are shown on a separate graph from the El *2b groups (FIG. 16A) for clarity. All treatments were significantly better at controlling tumor growth in terms of area-under-the-curve (AUC) when compared to the untreated mice out to day 29, which was when the first mouse in the untreated group was euthanized for disease progression ( ⁇ 0.0009; AUC 29 da y s ). Combining (El)-3s with PEGASYS® resulted in the best anti-tumor response overall in terms of tumor out-growth (FIG. 16B).
  • AUC area-under-the-curve
  • interferon-a provides a substantial increase in survival and decrease in tumor growth when combined with a leukocyte redirecting bsAb.
  • interferon-a provides a substantial increase in survival and decrease in tumor growth when combined with a leukocyte redirecting bsAb.
  • type I or type III interferons is not limited to the specific (El)-3s bsAb, but will be observed with other leukocyte redirecting bsAbs, made either as D L® complexes or in other forms, such as BITE® or DARTTM.
  • mice (40) were injected with the Capan-l/T-cell mixture, they were randomized into five treatment groups. One hour later, one group of 11 mice received 47 ⁇ g (El)-3s i.v. every day starting 1 h post-tumor cell injection and continued for four more consecutive days (qdx5). One group of 7 animals received interferon in the form of
  • PEGASYS® s.c. on a weekly basis for four weeks.
  • Another group received a combination of (El)-3s i.v. plus PEGASYS® s.c.
  • Untreated control animals receive Capan-l/T cells but no treatment.
  • a further control group received TF12 at amounts equivalent to the (El)-3s in terms of moles (57 ⁇ g qdx5).
  • Group 6 mice (8 animals) received a separate injection of only Capan-1 cells (i.e., no T cells) and was treated with PEGASYS®. All therapy injections were in a volume of 100 ⁇ .. Table 5 summarizes the various groups
  • mice were monitored daily for signs of tumor out-growth. All animals had their tumors measured twice weekly once tumors began to come up. Mice were euthanized for disease progression if their tumor volumes exceeded 1.0 cm 3 in size.
  • mice treated with (El)-3s, PEGASYS®, or PEGASYS® demonstrated significant anti-tumor effects when compared to TF12 and untreated control groups (RO.0102; AUC).
  • the mean tumor volume for the mice treated with the combination of (El)-3s plus PEGASYS® was 0.083 ⁇ 0.048 cm 3 .
  • this treatment group demonstrated a significant anti-tumor effect when compared to all the other treatment groups ( ⁇ 0.0072; AUC).
  • FIG. 20 and FIG. 21 The effects of leukocyte redirecting bsAb (El)-3s alone or in combination with interferon are shown in FIG. 20 and FIG. 21.
  • the (El)-3s bsAb was effective to reduce tumor growth and increase survival in gastric cancer.
  • the combination with interferon-a enhanced the effect of leukocyte redirecting bsAb, even in an interferon resistant tumor.
  • the combination therapy was more effective than either agent added alone. Controls with mice treated with TF12 bsAb alone or in combination with interferon- ⁇ showed little effect on tumor growth or mortality, compared to untreated animals.
  • CL2A-SN-38-antibody conjugates were prepared as previously described (see, e.g., U.S. Patent Nos. 7,999,083 and 8,080,250). Immune-compromised athymic nude mice (female), bearing subcutaneous human pancreatic or colon tumor xenografts were treated with either specific CL2A-SN-38 conjugate or control conjugate or were left untreated. The therapeutic efficacies of the specific conjugates were observed.
  • Example 7 In vivo therapy of lung metastases of GW-39 human colonic tumors in nude mice using ADC hMN-14-[CL2-SN-38], IMMU-130
  • a lung metastatic model of colonic carcinoma was established in nude mice by i.v. injection of GW-39 human colonic tumor suspension, and therapy was initiated 14 days later.
  • Specific anti-CEACAM5 antibody conjugate, hMN14-CL2-SN-38, as well as nontargeting anti-CD22 MAb control conjugate, hLL2-CL2-SN-38 and equidose mixtures of hMN14 and SN-38 were injected at a dose schedule of q4dx8, using different doses. Selective therapeutic effects were observed with the hMN-14 ADC (not shown).
  • the mice treated with hMN14-CL2- SN-38 showed a median survival of greater than 107 days.
  • mice treated with the control conjugated antibody hLL2-CL2-SN-38 which does not specifically target lung cancer cells, showed median survival of 77 days, while mice treated with unconjugated hMN14 IgG and free SN-38 showed a median survival of 45 days, comparable to the untreated saline control of 43.5 days.
  • a significant and surprising increase in effectiveness of the conjugated, cancer cell targeted antibody-SN-38 conjugate which was substantially more effective than unconjugated antibody and free chemotherapeutic agent alone, was clearly seen (not shown).
  • the dose-responsiveness of therapeutic effect of conjugated antibody was also observed (not shown).
  • Example 8 Use of ADC (IMMU-132 or hRS7-SN-38) to Treat Therapy- Refractive Metastatic Colonic Cancer (mCRC)
  • the patient was a 62-year-old woman with mCRC who originally presented with metastatic disease in January 2012. She had laparoscopic ileal transverse colectomy as the first therapy a couple of weeks after diagnosis, and then received 4 cycles of FOLFOX (leucovorin, 5-fluorouracil, oxaliplatin) chemotherapy in a neoadjuvant setting prior to right hepatectomy in March 2012 for removal of metastatic lesions in the right lobe of the liver. This was followed by an adjuvant FOLFOX regimen that resumed in June, 2012, for a total of 12 cycles of FOLFOX. In August, oxaliplatin was dropped from the regimen due to worsening neurotoxicity. Her last cycle of 5-FU was on 09/25/12.
  • CT done in Jan 2013 showed metastases to liver. She was then assessed as a good candidate for enrollment to IMMU-132 (hRS7-SN-38) investigational study.
  • Comorbidities in her medical history include asthma, diabetes mellitus, hypertension, hypercholesteremia, heart murmur, hiatal hernia, hypothyroidism, carpel tunnel syndrome, glaucoma, depression, restless leg syndrome, and neuropathy.
  • Her surgical history includes tubo-ligation (1975), thyroidectomy (1983), cholescystectomy (2001), carpel tunnel release (2008), and glaucoma surgery.
  • her target lesion was a 3.1-cm tumor in the left lobe of the liver.
  • Non-target lesions included several hypo-attenuated masses in the liver.
  • Her baseline CEA was 781 ng/mL.
  • IMMU-132 was given on a once-weekly schedule by infusion for 2 consecutive weeks, then a rest of one week, this constituting a treatment cycle. These cycles were repeated as tolerated.
  • the first infusion of IMMU-132 (8 mg/kg) was started on Feb 15, 2013, and completed without notable events. She experienced nausea (Grade 2) and fatigue (Grade 2) during the course of the first cycle and has been continuing the treatment since then without major adverse events. She reported alopecia and constipation in March 2013.
  • the first response assessment done (after 6 doses) on 04/08/2013 showed a shrinkage of target lesion by 29% by computed tomography (CT). Her CEA level decreased to 230 ng/mL on March 25, 2013.
  • CT computed tomography
  • IMMU-132 targets Trop-2, a type I transmembrane protein expressed in high prevalence and specificity by many carcinomas.
  • This Example reports a Phase I clinical trial of 25 patients with different metastatic cancers (pancreatic, 7; triple-negative breast [TNBC], 4; colorectal [CRC], 3; gastric, 3, esophageal, prostatic, ovarian, non-small-cell lung, small- cell lung [SCLC], renal, tonsillar, urinary bladder, 1 each) after failing a median of 3 prior treatments (some including topoisomerase-I and -II inhibiting drugs).
  • IMMU-132 was administered in repeated 21-day cycles, with each treatment given on days 1 and 8. Dosing started at 8 mg/kg/dose (i.e., 16 mg/kg/cycle), and escalated to 18 mg/kg before encountering dose-limiting neutropenia, in a 3+3 trial design. Fatigue, alopecia, and occasional mild to moderate diarrhea were some of the more common non- hematological toxicities, with 2 patients also reporting a rash. Over 80% of 24 assessable patients had stable disease or tumor shrinkage (SD and PR) among the various metastatic cancers as best response by CT. Three patients (CRC, TNBC, SCLC) have PRs by RECIST 1.1; median TTP for all patients, excluding those with pancreatic cancer, is >18 weeks.
  • SD and PR tumor shrinkage
  • Neutropenia has been controlled by dose reduction to 8-10 mg/kg/dose (16-20 mg/kg/cycle).
  • IMMU-130 an ADC of SN-38 conjugated by a pH-sensitive linker (7.6 average drug- antibody ratio) to the humanized anti-CEACAM5 antibody (labetuzumab), is completing two Phase I trials. In both, eligible patients with advanced mCRC were required to have failed/relapsed standard treatments, one being the topoisomerase-I inhibiting drug, CPT-11 (irinotecan), and an elevated plasma CEA (>5 ng/mL).
  • IMMU-130 was administered every 14 days (EOW) at doses starting from 2.0 mg/kg in the first protocol (IMMU-130-01). Febrile neutropenia occurred in 2 of 3 patients at 24 mg/kg; otherwise at ⁇ 16 mg/kg, neutropenia (> Grade 2) was observed in 7 patients, with one also experiencing thrombocytopenia.
  • CEA blood titers correlated with tumor response, and high levels did not interfere with therapy. There have been no anti-antibody or anti-SN-38 antibody reactions, based on ELISA tests. In each study, the ADC was cleared by 50% within the first 24 h, which is much longer exposure than with typical doses of the parental molecule, CPT-11. These results indicate that this novel ADC, given in different regimens averaging -16-24 mg/kg/cycle, shows a high therapeutic index in advanced mCRC patients. Since CEACAM5 has elevated expression in breast and lung cancers, as well as other epithelial tumors, it may be a useful target in other cancers as well.
  • CTLA4 mAb is evaluated alone or in combination with the exemplary T-cell redirecting bsAb (El)-3s, with interferon-a (PEGINTERFERON®), or with the exemplary ADC hRS7-SN-38 (IMMU-132) in murine tumor models.
  • El T-cell redirecting bsAb
  • PEGINTERFERON® interferon-a
  • IMMU-132 exemplary ADC hRS7-SN-38
  • CTLA4 mAb is initiated one day after the first dose of IMMU-132, (El)-3s or interferon-a. Percent tumor growth inhibition and number of days to reach target tumor size are used to evaluate efficacy. Antitumor activity is scored as: complete regression (CR; nonpalpable tumor) or partial regression (PR; 50% reduction in tumor volume). Synergy is defined as antitumor activity significantly superior (p ⁇ 0.05) to the activity of monotherapy with each agent.
  • the patient is a 60-year-old man diagnosed with non-small cell lung cancer.
  • the patient is given chemotherapy regimens of carboplatin, bevacizumab for 6 months and shows a response, and then after progressing, receives further courses of chemotherapy with carboplatin, etoposide, TAXOTERE®, gemcitabine over the next 2 years, with occasional responses lasting no more than 2 months.
  • the patient then presents with a left mediastinal mass measuring 6.5 x 4 cm and pleural effusion.
  • the patient After signing informed consent, the patient is given IMMU-132 at a dose of 12 mg/kg every other week. After the first week of treatment, the patient is given combination therapy with IMMU-132 and PEGINTERFERON®. During the first two injections, brief periods of neutropenia and diarrhea are experienced, with 4 bowel movements within 4 hours, but these resolve or respond to symptomatic medications within 2 days. After a total of 6 infusions of IMMU-132 and 5 infusions of PEGINTERFERON®, CT evaluation of the index lesion shows a 22% reduction, just below a partial response but definite tumor shrinkage.
  • the patient continues with this therapy for another two months, when a partial response of 45% tumor shrinkage of the sum of the diameters of the index lesion is noted by CT, thus constituting a partial response by RECIST criteria.
  • the combination therapy appears to provide a synergistic response, compared to the two agents administered separately.
  • the patient is a 75-year-old woman initially diagnosed with metastatic colonic cancer (Stage IV). She has a right partial hemicolectomy and resection of her small intestine and then receives FOLFOX, FOLFOX + bevacizumab, FOLFIRI + ramucirumab, and FOLFIRI + cetuximab therapies for a year and a half, when she shows progression of disease, with spread of disease to the posterior cul-de-sac, omentum, with ascites in her pelvis and a pleural effusion on the right side of her chest cavity. Her baseline CEA titer just before this therapy is 15 ng/mL.
  • IMMU-130 anti-CEACAM5-SN-38
  • F MU-130 anti-CEACAM5-SN-38
  • MT110 leukocyte redirecting bsAb MT110
  • her plasma CEA titer shrinks modestly to 1.3 ng/mL, but at the 8-week evaluation she shows a 21% shrinkage of the index tumor lesions, which increases to a 27% shrinkage at 13 weeks.
  • the patient's ascites and pleural effusion both decrease (with the latter disappearing) at this time, thus improving the patient's overall status remarkably.
  • the combination therapy appears to provide a synergistic response, compared to the two agents administered separately.
  • Example 14 Combination Therapy With ADC (IMMU-130), T-Cell Redirecting bsAb ((El)-3s) and Interferon-a to Treat Gastric Cancer Patient with Stage IV Metastatic Disease
  • the patient is a 52-year-old male who sought medical attention because of gastric discomfort and pain related to eating for about 6 years, and with marked weight loss during the past 12 months. Palpation of the stomach area reveals a firm lump which is then gastroscoped, revealing an ulcerous mass at the lower part of his stomach. This is biopsied and diagnosed as a gastric adenocarcinoma, with copious CEA staining by
  • IMMU-130 anti-CEACAM5- SN-38
  • combination therapy with IMMU-130, (El)-3s and interferon-a is initiated.
  • the patient exhibits no evidence of diarrhea or neutropenia over the following 4 weeks.
  • the patient then undergoes a CT study to measure his metastatic tumor sizes and to view the original area of gastric resection.
  • the radiologist measures, according to RECIST 1.1 criteria, a decrease of the sum of the metastatic lesions, compared to baseline prior to therapy, of 23%. There does not seem to be any clear lesion in the area of the original gastric resection.
  • the patient's CEA titer at this time is 7.2 ng/mL, which is much reduced from the baseline value of 14.5 ng/mL.
  • the patient continues on weekly combination therapy, with reduction of the IMMU-130 dose to 6 mg/kg, and after a total of 13 infusions, his CT studies show that one liver metastasis has disappeared and the sum of all metastatic lesions is decreased by 41%, constituting a partial response by RECIST.
  • the patient's general condition improves and he resumes his usual activities while continuing to receive maintenance therapy every third week.
  • the value is 4.8 ng/mL, which is within the normal range for a smoker, which is the case for this patient.
  • the plasmid vector pdHL2 has been used to produce a number of antibodies and antibody-based constructs. See Gillies et al., J Immunol Methods (1989), 125: 191-202; Losman et al., Cancer (Phila) (1997), 80:2660-6.
  • the di-cistronic mammalian expression vector directs the synthesis of the heavy and light chains of IgG.
  • the vector sequences are mostly identical for many different IgG-pdHL2 constructs, with the only differences existing in the variable domain (V H and V L ) sequences. Using molecular biology tools known to those skilled in the art, these IgG expression vectors can be converted into Fab-DDD or Fab- AD expression vectors.
  • Fab-DDD expression vectors To generate Fab-DDD expression vectors, the coding sequences for the hinge, CH2 and CH3 domains of the heavy chain were replaced with a sequence encoding the first 4 residues of the hinge, a 14 residue linker and a DDD moiety, such as the first 44 residues of human Rlla (referred to as DDDl, SEQ ID NO: 1).
  • Fab-AD expression vectors To generate Fab-AD expression vectors, the sequences for the hinge, CH2 and CH3 domains of IgG were replaced with a sequence encoding the first 4 residues of the hinge, a 15 residue linker and an AD moiety, such as a 17 residue synthetic AD called AKAP-IS (referred to as ADl, SEQ ID NO:3), which was generated using bioinformatics and peptide array technology and shown to bind Rlla dimers with a very high affinity (0.4 nM). See Alto, et al. Proc. Natl. Acad. Sci., U.S.A (2003), 100:4445-50. Two shuttle vectors were designed to facilitate the conversion of IgG-pdHL2 vectors to either Fab-DDDl or Fab-AD 1 expression vectors, as described below.
  • ADl 17 residue synthetic AD
  • the CHI domain was amplified by PCR using the pdHL2 plasmid vector as a template.
  • the left PCR primer consisted of the upstream (5') end of the CHI domain and a SacII restriction endonuclease site, which is 5' of the CHI coding sequence.
  • the right primer consisted of the sequence coding for the first 4 residues of the hinge (PKSC, SEQ ID NO:28) followed by four glycines and a serine, with the final two codons (GS) comprising a Bam HI restriction site.
  • the 410 bp PCR amplimer was cloned into the PGEMT® PCR cloning vector (PROMEGA®, Inc.) and clones were screened for inserts in the T7 (5') orientation.
  • a duplex oligonucleotide was synthesized to code for the amino acid sequence of DDDl preceded by 11 residues of the linker peptide, with the first two codons comprising a BamHI restriction site. A stop codon and an Eagl restriction site are appended to the 3 'end.
  • the encoded polypeptide sequence is shown below.
  • oligonucleotides designated RIIA1-44 top and RIIA1-44 bottom, which overlap by 30 base pairs on their 3' ends, were synthesized and combined to comprise the central 154 base pairs of the 174 bp DDDl sequence.
  • the oligonucleotides were annealed and subjected to a primer extension reaction with Taq polymerase. Following primer extension, the duplex was amplified by PCR. The amplimer was cloned into PGEMT® and screened for inserts in the T7 (5') orientation.
  • a duplex oligonucleotide was synthesized to code for the amino acid sequence of ADl preceded by 11 residues of the linker peptide with the first two codons comprising a BamHI restriction site. A stop codon and an Eagl restriction site are appended to the 3 'end. The encoded polypeptide sequence is shown below.
  • GS GGGGS GGGGS QIE YL AKQI VPN AIQQ A (SEQ ID NO: 30)
  • a 190 bp fragment encoding the DDD1 sequence was excised from PGEMT® with BamHI and Notl restriction enzymes and then ligated into the same sites in CHI -PGEMT® to generate the shuttle vector CHI -DDD1 -PGEMT®.
  • a 110 bp fragment containing the AD1 sequence was excised from PGEMT® with BamHI and Notl and then ligated into the same sites in CHI -PGEMT® to generate the shuttle vector CHI -AD 1 -PGEMT®.
  • CH1-DDD1 or CHI -AD 1 can be incorporated into any IgG construct in the pdHL2 vector.
  • the entire heavy chain constant domain is replaced with one of the above constructs by removing the SacII/EagI restriction fragment (CH1-CH3) from pdHL2 and replacing it with the SacII/EagI fragment of CH1-DDD1 or CHI -AD 1, which is excised from the respective PGEMT® shuttle vector.
  • C-DDD2-Fd-hMN-14-pdHL2 is an expression vector for production of C-DDD2-Fab- hMN-14, which possesses a dimerization and docking domain sequence of DDD2 (SEQ ID NO:2) appended to the carboxyl terminus of the Fd of hMN-14 via a 14 amino acid residue Gly/Ser peptide linker.
  • the fusion protein secreted is composed of two identical copies of hMN-14 Fab held together by non-covalent interaction of the DDD2 domains.
  • the expression vector was engineered as follows. Two overlapping, complimentary oligonucleotides, which comprise the coding sequence for part of the linker peptide and residues 1-13 of DDD2, were made synthetically. The oligonucleotides were annealed and phosphorylated with T4 PNK, resulting in overhangs on the 5' and 3' ends that are compatible for ligation with DNA digested with the restriction endonucleases BamHI and Pstl, respectively.
  • the duplex DNA was ligated with the shuttle vector CH1-DDD1 -PGEMT®, which was prepared by digestion with BamHI and Pstl, to generate the shuttle vector CH1-DDD2- PGEMT®.
  • a 507 bp fragment was excised from CHI -DDD2-PGEMT® with SacII and EagI and ligated with the IgG expression vector hMN-14(I)-pdHL2, which was prepared by digestion with SacII and EagI.
  • the final expression construct was designated C-DDD2-Fd- hMN-14-pdHL2. Similar techniques have been utilized to generated DDD2-fusion proteins of the Fab fragments of a number of different humanized antibodies.
  • h679-Fab-AD2 was designed to pair to C-DDD2-Fab-hMN-14.
  • h679-Fd-AD2- pdHL2 is an expression vector for the production of h679-Fab-AD2, which possesses an anchoring domain sequence of AD2 (SEQ ID NO:4) appended to the carboxyl terminal end of the CHI domain via a 14 amino acid residue Gly/Ser peptide linker.
  • AD2 has one cysteine residue preceding and another one following the anchor domain sequence of AD1.
  • the expression vector was engineered as follows. Two overlapping, complimentary oligonucleotides (AD2 Top and AD2 Bottom), which comprise the coding sequence for AD2 and part of the linker sequence, were made synthetically. The oligonucleotides were annealed and phosphorylated with T4 P K, resulting in overhangs on the 5' and 3' ends that are compatible for ligation with DNA digested with the restriction endonucleases BamHI and Spel, respectively.
  • duplex DNA was ligated into the shuttle vector CHl-ADl-PGEMT®, which was prepared by digestion with BamHI and Spel, to generate the shuttle vector CH1-AD2- PGEMT®.
  • a 429 base pair fragment containing CHI and AD2 coding sequences was excised from the shuttle vector with SacII and Eagl restriction enzymes and ligated into h679-pdHL2 vector that prepared by digestion with those same enzymes.
  • the final expression vector is h679-Fd-AD2-pdHL2.
  • a trimeric DNL® construct designated TF2 was obtained by reacting C-DDD2-Fab- hMN-14 with h679-Fab-AD2.
  • a pilot batch of TF2 was generated with >90% yield as follows.
  • Protein L-purified C-DDD2-Fab-hMN-14 200 mg was mixed with h679-Fab-AD2 (60 mg) at a 1.4: 1 molar ratio.
  • the total protein concentration was 1.5 mg/ml in PBS containing 1 mM EDTA.
  • Subsequent steps involved TCEP reduction, HIC chromatography, DMSO oxidation, and IMP 291 affinity chromatography. Before the addition of TCEP, SE- HPLC did not show any evidence of a 2 b formation.
  • TF2 was purified to near homogeneity by FMP 291 affinity chromatography (not shown).
  • IMP 291 is a synthetic peptide containing the HSG hapten to which the 679 Fab binds (Rossi et al., 2005, Clin Cancer Res l l :7122s-29s).
  • SE-HPLC analysis of the IMP 291 unbound fraction demonstrated the removal of a 4 , a 2 and free kappa chains from the product (not shown).
  • the functionality of TF2 was determined by BIACORE® assay.
  • TF2 C-DDD1- hMN-14+h679-ADl (used as a control sample of noncovalent a 2 b complex), or C-DDD2- hMN-14+h679-AD2 (used as a control sample of unreduced a 2 and b components) were diluted to 1 ⁇ g/ml (total protein) and passed over a sensorchip immobilized with HSG.
  • the response for TF2 was approximately two-fold that of the two control samples, indicating that only the h679-Fab-AD component in the control samples would bind to and remain on the sensorchip.
  • TF10 DNL® construct comprising two copies of a C-DDD2-Fab-hPAM4 and one copy of C-AD2-Fab-679.
  • the TF10 bispecific ([hPAM4] 2 x h679) antibody was produced using the method disclosed for production of the (anti CEA) 2 x anti HSG bsAb TF2, as described above.
  • the TF10 construct bears two humanized PAM4 Fabs and one humanized 679 Fab.
  • tissue culture supernatant fluids were combined, resulting in a two-fold molar excess of hPAM4-DDD2.
  • the reaction mixture was incubated at room temperature for 24 hours under mild reducing conditions using 1 mM reduced glutathione. Following reduction, the reaction was completed by mild oxidation using 2 mM oxidized glutathione.
  • TF10 was isolated by affinity chromatography using IMP291-affigel resin, which binds with high specificity to the h679 Fab.
  • the IgG and Fab fusion proteins shown in Table 7 were constructed and incorporated into DNL® constructs.
  • the fusion proteins retained the antigen-binding characteristics of the parent antibodies and the DNL® constructs exhibited the antigen-binding activities of the incorporated antibodies or antibody fragments.
  • trimeric D L® constructs may comprise three different effector moieties, for example two different antibody moieties and a cytokine moiety.
  • a bispecific MAb-IFNa designated 20-C2- 2b, which comprises two copies of IFN-a2b and a stabilized F(ab) 2 of hL243 (humanized anti-HLA-DR; IMMU-114) site-specifically linked to veltuzumab (humanized anti-CD20).
  • 20-C2-2b inhibited each of four lymphoma and eight myeloma cell lines, and was more effective than monospecific CD20-targeted MAb-IFNa or a mixture comprising the parental antibodies and IFNa in all but one (HLA-DR7CD20 " ) myeloma line (not shown), suggesting that 20-C2-2b is useful for the treatment of various hematopoietic disorders.
  • the 20-C2-2b displayed greater cytotoxicity against KMS 12-BM (CD20 + /HLA-DR + myeloma) than monospecific MAb-IFNa that targets only HLA-DR or CD20 (not shown), indicating that all three components in 20-C2-2b can contribute to toxicity.
  • Immunomedics, Inc. veltuzumab or v-mab (anti-CD20 IgGi), hL243y4p (Immu-1 14, anti- HLA-DR IgG 4 ), a murine anti-IFNa MAb, and rat anti-idiotype MAbs to v-mab (WR2) and hL243 (WT).
  • Monospecific MAb-IFNa (20-2b-2b, 734-2b-2b and C2-2b-2b) and the bispecific HexAb (20-C2-C2) were generated by combination of an IgG-AD2-module with DDD2- modules using the DNL® method, as described in the preceding Examples.
  • the construction of the mammalian expression vector as well as the subsequent generation of the production clones and the purification of C H 3-AD2-IgG-v-mab are disclosed in the preceding Examples.
  • the expressed recombinant fusion protein has the AD2 peptide linked to the carboxyl terminus of the CH3 domain of v-mab via a 15 amino acid long flexible linker peptide.
  • Co-expression of the heavy chain- AD2 and light chain polypeptides results in the formation of an IgG structure equipped with two AD2 peptides.
  • the expression vector was transfected into Sp/ESF cells (an engineered cell line of Sp2/0) by electroporation.
  • the pdHL2 vector contains the gene for dihydrofolate reductase, thus allowing clonal selection, as well as gene amplification with methotrexate (MTX).
  • Stable clones were isolated from 96-well plates selected with media containing 0.2 ⁇ MTX. Clones were screened for C H 3-AD2-IgG-vmab productivity via a sandwich ELISA. The module was produced in roller bottle culture with serum-free media.
  • the DDD-module, IFNa2b-DDD2 was generated as discussed above by recombinant fusion of the DDD2 peptide to the carboxyl terminus of human IFNa2b via an 18 amino acid long flexible linker peptide. As is the case for all DDD-modules, the expressed fusion protein spontaneously forms a stable homodimer.
  • the C H l-DDD2-Fab-hL243 expression vector was generated from hL243-IgG-pdHL2 vector by excising the sequence for the 3 ⁇ 41- ⁇ -3 ⁇ 42-3 ⁇ 43 domains with SacII and Eagl restriction enzymes and replacing it with a 507 bp sequence encoding CH1-DDD2, which was excised from the C-DDD2-hMN-14-pdHL2 expression vector with the same enzymes.
  • the culture broth containing the C H l-DDD2-Fab-hL243 module was applied directly to KAPPASELECT® affinity gel (GE-Healthcare), which was washed to baseline with PBS and eluted with 0.1 M Glycine, pH 2.5.
  • the DNL® mixture was purified with Protein A (MAB SELECTTM), which binds the C H 3-AD2-IgG-v-MAb group and eliminates un-reacted IFNa2b-DDD2 or C H l-DDD2-Fab-hL243.
  • the Protein A-bound material was further purified by IMAC using HIS-SELECT® HF Nickel Affinity Gel, which binds specifically to the IFNa2b-DDD2 moiety and eliminates any constructs lacking this group.
  • the final process step, using an hL243-anti-idiotype affinity gel removed any molecules lacking C H l-DDD2-Fab-hL243.
  • affinity chromatography may be used to purify DNL® complexes comprising any combination of effector moieties, so long as ligands for each of the three effector moieties can be obtained and attached to the column material.
  • the selected DNL® construct is the one that binds to each of three columns containing the ligand for each of the three effector moieties and can be eluted after washing to remove unbound complexes.
  • the eluate which contained -20 mg protein, was neutralized with 3 M Tris-HCl, pH 8.6 and dialyzed into HIS- SELECT® binding buffer (10 mM imidazole, 300 mM NaCl, 50 mM NaH 2 P0 4 , pH 8.0) prior to application to a 5-mL HIS-SELECT® IMAC column.
  • HIS- SELECT® binding buffer 10 mM imidazole, 300 mM NaCl, 50 mM NaH 2 P0 4 , pH 8.0
  • the column was washed to baseline with binding buffer and eluted with 250 mM imidazole, 150 mM NaCl, 50 mM NaH 2 P0 4 , pH 8.0.
  • the IMAC eluate which contained -1 1.5 mg of protein, was applied directly to a WP (anti-hL243) affinity column, which was washed to baseline with PBS and eluted with 0.1 M glycine, pH 2.5.
  • the process resulted in 7 mg of highly purified 20-C2-2b. This was approximately 44% of the theoretical yield of 20-C2-2b, which is 50% of the total starting material (16 mg in this example) with 25% each of 20-2b-2b and 20-C2-C2 produced as side products.
  • the bispecific MAb-IFNa was generated by combining the IgG-AD2 module, CH3- AD2-IgG-v-mab, with two different dimeric DDD-modules, C H l-DDD2-Fab-hL243 and IFNa2b-DDD2. Due to the random association of either DDD-module with the two AD2 groups, two side-products, 20-C2-C2 and 20-2b-2b are expected to form, in addition to 20- C2-2b.
  • Non-reducing SDS-PAGE resolved 20-C2-2b (-305 kDa) as a cluster of bands positioned between those of 20-C2-C2 (-365 kDa) and 20-2b-2b (255 kDa).
  • Reducing SDS-PAGE resolved the five polypeptides (v-mab HC-AD2, hL243 Fd-DDD2, IFNa2b- DDD2 and co-migrating v-mab and hL243 kappa light chains) comprising 20-C2-2b (not shown).
  • IFNa2b-DDD2 and hL243 Fd-DDD2 are absent in 20-C2-C2 and 20-2b-2b.
  • MAB SELECTTM binds to all three of the major species produced in the DNL® reaction, but removes any excess IFNa2b-DDD2 and C H l-DDD2-Fab-hL243.
  • the HIS-SELECT® unbound fraction contained mostly 20-C2-C2 (not shown).
  • the unbound fraction from WT affinity chromatography comprised 20-2b-2b (not shown).
  • Each of the samples was subjected to SE-HPLC and immunoreactivity analyses, which corroborated the results and conclusions of the SDS-PAGE analysis.
  • LC/MS analysis of 20-C2-2b identified both the O-glycosylated and non-glycosylated species of IFNa2b-DDD2 with mass accuracies of 15 ppm and 2 ppm, respectively (not shown).
  • the observed mass of the O-glycosylated form indicates an O-linked glycan having the structure NeuGc-NeuGc-Gal-GalNAc, which was also predicted ( ⁇ 1 ppm) for 20-2b-2b (not shown).
  • LC/MS identified both v-mab and hL243 kappa chains as well as hL243-Fd-DDD2 (not shown) as single, unmodified species, with observed masses matching the calculated ones ( ⁇ 35 ppm).
  • v-mab HC-AD2 Two major glycoforms of v-mab HC-AD2 were identified as having masses of 53,714.73 (70%) and 53,877.33 (30%), indicating G0F and GIF N-glycans, respectively, which are typically associated with IgG (not shown). The analysis also confirmed that the amino terminus of the HC-AD2 is modified to pyroglutamate, as predicted for polypeptides having an amino terminal glutamine.
  • Immunoreactivity assays demonstrated the homogeneity of 20-C2-2b with each molecule containing the three functional groups (not shown). Incubation of 20-C2-2b with an excess of antibodies to any of the three constituent modules resulted in quantitative formation of high molecular weight immune complexes and the disappearance of the 20-C2-2b peak (not shown). The HIS-SELECT® and WT affinity unbound fractions were not
  • the 20-C2-2b DNL® construct depleted lymphoma cells more effectively than normal B cells and had no effect on T cells (not shown). However, it did efficiently eliminate monocytes (not shown). Where v-mab had no effect on monocytes, depletion was observed following treatment with hL243a4p and MAb-IFNa, with 20-2b-2b and 734-2b-2b exhibiting similar toxicity (not shown). Therefore, the predictably higher potency of 20-C2-2b is attributed to the combined actions of anti-HLA-DR and IFNa, which may be augmented by HLA-DR targeting.
  • monocyte depletion may be a pharmacodynamic effect associated anti-HLA-DR as well as IFNa therapy; however, this side effect would likely be transient because the monocyte population should be repopulated from hematopoietic stem cells.
  • Example 18 Use of NK- Targeted Leukocyte-Redirecting bsAbs
  • bsAbs to retarget leukocytes is not limited to antibodies against T cells.
  • bsAbs that bind to monocytes, NK cells or neutrophils may also be used for retargeting purposes.
  • CD 16 is an activating low-affinity Fc- ⁇ receptor for IgG, which is highly expressed by the CD56 dim subset of NK cells (Gleason et al., 2012, Mol Cancer Ther 11 :2674-84).
  • bsAbs comprising an anti-CD 16 antibody component have the ability to activate NK-mediated cytotoxicity through direct signaling of CD 16, inducing directed secretion of lytic granules and target cell death (Gleason et al., 2012).
  • a CD16/CD19 bispecific killer cell engager (BiKE) and a CD16/CD19/CD22 trispecific killer cell engager (TriKe) are prepared according to (Gleason et al., 2012, Mol Cancer Ther 11 :2674-84), using DNA shuffling and ligation techniques as previously reported (Vallera et al., 2005, Clin Cancer Res 11 :3879-88).
  • the expressed BiKE and TriKE are purified by sequential ion exchange and size-exclusion column chromatography. Resting PBMCs are exposed to primary ALL and CLL tumor cells in the presence of CD16/CD19 BiKE or CD16/CD19/CD22 TriKE (10 ⁇ g/mL).
  • a CD16/CD33 BiKE is prepared as disclosed in Wiernik et al. (2013, Clin Cancer Res 19:3844-55.
  • the BiKE is administered to nude mice injected with human HL60 promyelocytic leukemia xenograft cells, co-administered with human PBMCs.
  • the BiKE treated mice show a decreased mortality and tumor growth rate compared to mice treated with control bsAbs. Addition of an anti-CD33-SN-38 ADC further enhances the cytotoxic effect of the BiKE.
  • a trivalent, trispecific cell targeting construct is made as described in patent
  • EP1309795B1 comprising: (i) chimerizing or humanizing a mouse anti-CD16 mab as described in patent US 618728 from which the Fab of Claim 1 of EP1309795 is derived; (ii) constructing a single chain antibody comprised of the Fv of the humanized anti-HLA-DR antibody described in US7512189, and joining the scFv by a linker to the carboxyl terminal of the light chain of the anti-CD 16 Fab of (i); and (iii) constructing a single chain of the Fv of the humanized anti-CD19 described in US8486395 and joining the scFv by a linker to the carboxyl terminal of the CHI of the anti-CD 16 Fab of (ii).
  • the trivalent construct is administered to a subject with non-Hodgkin's lymphoma, in combination with hLL2-SN38. A partial response is observed and the tumor shows a regression in size that lasts for 12 months.
  • a bispecific antibody was produced as a tandem single-chain variable fragment (scFv) for redirecting T cells via CD3 binding to tumor cells, particularly carcinomas, via Trop-2 targeting.
  • Trop-2 is a tumor-associated antigen (TAA) that could be highly effective for targeting various epithelial cancers.
  • TAA tumor-associated antigen
  • Trop-2 is a 35 kDa transmembrane glycoprotein that is overexpressed relative to normal tissues in a variety of human cancers, including pancreatic and gastric carcinomas, where increased expression correlates with poor prognosis (Fong et al., 2008, Br J Cancer 99: 1290-5; Iacobuzio-Donahue et al., 2002, Am J Pathol 160: 1239-49; Kapoor, 2013, Tumour Biol 34: 1967-8; Muhlmann et al., 2009, J Clin Pathol 62: 152-8; Stein et al., 1993, Int J Cancer 55:938-46; Stein et al., 1993, Int J Cancer 55:938-46).
  • VH and VK Variable domains derived from hRS7, the humanized version of the original murine anti-Trop-2 mAb, RS7, were combined with the variable domains of the murine anti-CD3 mAb, Okt3, to generate the El-3 bsAb.
  • SEQ ID NO:31 A double stranded DNA sequence (SEQ ID NO:31) was synthesized and assembled into the pUC57 plasmid vector. SEQ ID NO:31 was excised from pUC57 by digestion with Xba I and Eag I restriction endonucleases, and ligated into the pdHL2 mammalian expression vector, which was prepared by digestion with the same enzymes.
  • the coding sequence directs the synthesis of a single polypeptide (SEQ ID NO:32) comprising a leader peptide, hRS7VK (SEQ ID NO:33), LI (SEQ ID NO:34), hRS7VH (SEQ ID NO:35), L2 (SEQ ID NO:36), Okt3VH (SEQ ID NO:37), L3 (SEQ ID NO:38), Okt3VK (SEQ ID NO:39), and 6-His (SEQ ID NO: 13).
  • a schematic representation of the tandem scFv El-3 is shown in FIG. 22.
  • VEGGS GGSGGS GGS GGVD (SEQ ID NO:38)
  • the El-3 protein was purified from the culture broth of roller bottle cultures by immobilized metal affinity chromatography (IMAC) using Nickel -SEPHAROSE® resin, followed by size exclusion high performance liquid chromatography (SE-HPLC).
  • IMAC immobilized metal affinity chromatography
  • SE-HPLC size exclusion high performance liquid chromatography
  • the purified product resolved as a single SE-HPLC peak (not shown) and a single polypeptide band by SDS-PAGE (not shown), with relative mobilities consistent with its calculated molecular size of 53,423 Da.
  • PBMCs Peripheral blood mononuclear cells
  • Trop-2/cell pancreatic cancer, 500,000 Trop-2/cell
  • NCI-N87 gastric cancer, 247,000 Trop-2/cell cell lines
  • BxPC3 and NCI-N87 were maintained in RPMI1640 media supplemented with 10% FBS, while Capan-1 cells were maintained in 20% FBS/RPMI1640.
  • CD8 + T cells (1.2 x 10 5 cells/well) were combined with target cells (2 x 10 4 cells/well) at a 6: 1 ratio in 96-well tissue culture plates. Titrations of El-3 and (El)-3s were added to the assay plates.
  • IC 50 values pM concentration resulting in 50% killing.
  • Donors 1 and 2 were the same for each donor.
  • Donors 3, 4 and 5 were independent donors.
  • mice Female 4-8-week old NOD/SCID mice were administered subcutaneous injections of a mixture of PBMCs and NCI-N87 (2: 1) mixed with an equal volume of MATRIGEL®. Therapy consisted of i.v. injections of 50 ⁇ g of El-3 on days 1 and 4, or daily injections with 47 ⁇ g of (El)-3s on days 1 through 5. The untreated group received the mixture of NCI-N87 and PBMCs without bsAb.
  • Tumor volume (TV) was determined twice weekly by measurements in two dimensions using calipers, with volumes defined as: L x W 2 /2, where L is the longest dimension of the tumor and W the shortest (FIG. 24).
  • Statistical analysis of tumor growth was based on area under the curve (AUC).
  • Profiles of individual tumor growth were obtained through linear-curve modeling.
  • An F-test was employed to determine equality of variance between groups prior to statistical analysis of growth curves.
  • a Critical Z test on the survival data identified any outliers within a given treatment group with P ⁇ 0.05 censored from the final data analysis.
  • a two-tailed t-test was used to assess statistical significance between the various treatment groups and controls, except for the untreated control, where a one-tailed t-test was used.
  • efficacy was determined by log-rank using Prism software on Kaplan-Meier curves using survival surrogate endpoints as time for tumor progression (TTP) to 1.0 cm3. Significance was considered at P ⁇ 0.05 for all comparisons.
  • Example 23 Combination Therapy with T-Cell Redirecting bsAb and Anti-PDl
  • bispecific antibodies for redirecting T cells to cancers have shown promise in both pre-clinical and clinical studies. However, clinical success has been minimal for solid cancers to date.
  • bsAbs trivalent bsAb
  • Trop-2 is highly expressed in diverse epithelial cancers, including breast, lung, gastric, colorectal, pancreatic, bladder, ovarian, uterine and prostate carcinomas, with limited presence on normal human tissues.
  • first generation bsAbs e.g. BiTE
  • efficient T-cell killing is mediated by (El)-3s with minimal cytokine release.
  • IMMU-cPDl is a chimeric mAb that binds with high affinity to human PD1 and efficiently blocks binding to its ligand, PD-L1.
  • the MDA-MB-231 human TNBC cell line has relatively low levels of surface Trop-2 (36,000/cell) and expresses PD-L1 constitutively.
  • (El)-3s mediated potent T-cell redirected killing (IC 50 ⁇ 10 pM).
  • (El)-3s is an attractive candidate for T-cell redirected therapy of breast cancer due to its potent activity with potentially reduced side effects and the prevalence of Trop-2 expression associated with this disease.
  • Tumor micro arrays representing 117 breast cancer patients showed >85% positivity for Trop-2.
  • Our immunohistochemical analysis of more than 50 individual TNBC patient specimens demonstrated 92% positivity with 80% having moderate to strong Trop-2 staining.
  • Combining checkpoint inhibitors with redirected T cell therapy may represent a new paradigm for the management of solid cancers, including breast, and is worthy of further investigation.
  • the blocking (antagonistic) anti-PDl monoclonal antibody 5G9.G1.B11 and its chimeric counterpart 2G9 were generated as follows. BALB/c mice were immunized with recombinant human PDl-Fc fusion protein (AB Biosciences), resulting in the isolation of a positive clone (5G9) by hybridoma technology. To ensure monoclonality, 5G9 was subcloned twice, yielding 5G9.G1.B11 (5G9 for short). The 5G9 mAb was purified to homogeneity, as shown by SE-HPLC and SDS-PAGE analyses (data not shown).
  • the reactivity of 5G9 for PD1 was confirmed by SE-HPLC with its binding to recombinant PD1- His (not shown), and by flow cytometry with its binding to PDl-expressed on activated Jurkat T cells (FIG. 25). Importantly, the blocking activity of 5G9 was demonstrated by a dose-dependent, notable increase of IL-2 secreted by T cells in a mixed lymphocyte assay (FIG. 26).
  • the amino acid sequences pertaining to the V K and V H of 5G9 were provided in FIG. 27A and FIG. 27B, respectively.
  • the CDRs of 5G9 are further delineated in FIG. 27C.
  • a chimeric version of 5G9 comprising the V H and V K of 5G9 and human Fc of IgGl, was generated, and the mAb from the lead clone (2G9) was purified and shown to be homogeneous by SDS-PAGE (not shown).
  • the binding of 2G9 to recombinant PDl-Fc with a high affinity of 70 pM was demonstrated by ELISA (FIG. 28A), and further confirmed by flow cytometry (FIG. 28B) using a subline of SpESFX transfected to overexpress PDl (SpESFX-2Dl).
  • the blocking activity of 2G9 was similar to that of 5G9 or EH12, as demonstrated by inhibiting the binding of biotinylated PDl to the endogenous PD-L1 expressed on MDA-MB-231 (FIG. 29).
  • a humanized anti-PDl antibody was constructed as follows.
  • the V genes of the anti- PD1 antibody from Example 24 above were identified by PCR amplification and DNA sequencing. To confirm their authenticity, the cloned Vk and V H genes were expressed in cell culture as a chimeric Ab as described by Orlandi et al., (Proc. Natl. Acad. Sci., USA, 86: 3833 (1989)) and shown in Example 24. Based on the V gene sequences, a humanized Anti- PDl antibody was designed and constructed as described by Leung et al. (Mol. Immunol., 32: 1413 (1995)).
  • cDNA was prepared from a transfected cell line producing a murine anti-PDl antibody by general molecular cloning techniques (Sambrook et al., Molecular Cloning, A laboratory manual, 2nd Ed (1989)).
  • the Vk sequence for the MAb was amplified using the extended primer set described by Leung et al. (BioTechniques, 15: 286 (1993)).
  • the V H sequence was amplified using the primers annealing to the constant region of murine IgG described by Leung et al. (Hybridoma, 13 :469 (1994)).
  • the humanized V genes are constructed by a combination of long oligonucleotide template syntheses and PCR amplification as described by Leung et al. ⁇ Mol. Immunol, 32: 1413 (1995)).
  • PCR products for Vk were subcloned into a staging vector that contains an Ig promoter, a signal peptide sequence and convenient restriction sites to facilitate in-frame ligation of the Vk. PCR products. PCR products for V H were subcloned into a similar staging vector.
  • Vk and V H expression cassettes were assembled in the modified staging vectors, VKpBR2 and VHpBS2, excised as Xbal/BamHI and Xhol/Hindlll fragments, respectively, and subcloned into a single expression vector, pdHL2, as described by Gilles et al. (J.
  • hygromycin selection medium Calbiochem, San Diego, Calif.
  • Colonies typically emerged 2-3 weeks post-electroporation.
  • the cultures were then expanded for further analysis.
  • Transfectoma clones that are positive for the secretion of chimeric, humanized or human heavy chain were identified by ELISA assay.
  • FIG. 30 shows the amino acid sequences of the light (SEQ ID NO:48) and heavy (SEQ ID NO:49) chains of the V5 hPDl .
  • the CDR sequences are underlined.
  • Framework region (FR) residues where the human FR residue was replaced with the corresponding murine residue from the parent antibody are indicated in bold.
  • For the light chain the substitutions were concentrated in FR3, while for the heavy chain they were distributed throughout the humanized antibody.
  • the corresponding DNA sequences encoding the light (SEQ ID NO:50) and heavy (SEQ ID NO:51) chains of the hPDl antibody (V5) are shown in FIG. 31.
  • the humanized anti-PDl were compared to chimeric anti-PDl in vitro, by binding to recombinant human PDl-His.
  • the humanized PDl antibodies exhibited somewhat lower affinities for recombinant PDl-His, with EC 50 values of 149 and 167 pM vs. 72 pM for cPDl .
  • the slightly lower affinity of hPDl for the target antigen was also observed in 2D1 cells transfected with human PD1 antigen, with EC 50 values of 0.1682 ⁇ g/mL for cPDl vs. 0.5679 ⁇ g/mL for hPDl v5.
  • both cPDl and hPDl enhanced the efficacy of the (El)-3s bsAb, targeting Trop-2 and CD3.
  • Anti -tumor activity was assessed by MTS assay against MDA-MB-231 human breast cancer cells.
  • the anti-PDl antibody resulted in a decrease in EC 50 from 4.7 pM with (El)-3s alone to 1.1 pM with (El)-3s plus cPDl .
  • the addition of hPDl (v5) resulted in a decrease from 4.7 pM to 2.1 pM, i.e. over a two-fold decrease in EC 50 .

Abstract

La présente invention concerne des compositions et des méthodes d'utilisation d'anticorps bispécifiques comprenant au moins un site de liaison pour un antigène associé à une tumeur (TAA) et au moins un site de liaison pour un antigène exprimé sur un lymphocyte T effecteur, une cellule NK, un monocyte ou un neutrophile. Les anticorps bispécifiques sont utiles pour induire une réponse immunitaire contre une tumeur exprimant TAA. Les méthodes peuvent consister à administrer l'anticorps bispécifique en combinaison avec un ou plusieurs agents thérapeutiques tels que des conjugués anticorps-médicaments (ADC), des interférons (de préférence l'interféron-α), et/ou des anticorps inhibiteurs de point de contrôle. L'anticorps bispécifique est capable de cibler des lymphocytes T effecteurs, des cellules NK, des monocytes ou des neutrophiles pour induire une cytotoxicité, médiée par les leucocytes, de cellules cancéreuses. La réponse immunitaire cytotoxique est amplifiée par co-administration de l'interféron, de l'anticorps inhibiteur de point de contrôle et/ou d'ADC. Dans des modes de réalisation préférés, l'inhibiteur de point de contrôle est un anticorps anti-PD1 chimérique ou humanisé tel que décrit ici.
PCT/US2017/034250 2016-06-01 2017-05-24 Polythérapie avec des anticorps bispécifiques de redirection des lymphocytes t et des inhibiteurs de point de contrôle WO2017210058A1 (fr)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US15/169,903 US9670286B2 (en) 2012-08-14 2016-06-01 Disease therapy by inducing immune response to Trop-2 expressing cells
US15/169,903 2016-06-01
US201662351646P 2016-06-17 2016-06-17
US62/351,646 2016-06-17
US15/497,931 US9879088B2 (en) 2012-08-14 2017-04-26 Disease therapy by inducing immune response to Trop-2 expressing cells
US15/497,931 2017-04-26

Publications (1)

Publication Number Publication Date
WO2017210058A1 true WO2017210058A1 (fr) 2017-12-07

Family

ID=60478919

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/034250 WO2017210058A1 (fr) 2016-06-01 2017-05-24 Polythérapie avec des anticorps bispécifiques de redirection des lymphocytes t et des inhibiteurs de point de contrôle

Country Status (1)

Country Link
WO (1) WO2017210058A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10513558B2 (en) 2015-07-13 2019-12-24 Cytomx Therapeutics, Inc. Anti-PD1 antibodies, activatable anti-PD1 antibodies, and methods of use thereof
WO2019224385A3 (fr) * 2018-05-24 2020-01-16 Glenmark Pharmaceuticals S.A. Anticorps bispécifiques combinés et thérapies immuno-oncologiques
WO2020051248A1 (fr) * 2018-09-05 2020-03-12 Arizona Board Of Regents On Behalf Of Arizona State University Plate-forme de virus oncolytique permettant de traiter le cancer hématologique
CN113164778A (zh) * 2018-12-19 2021-07-23 拜耳公司 抗ceacam6和tim3抗体的药物组合
US11667723B2 (en) 2020-08-17 2023-06-06 Utc Therapeutics (Shanghai) Co., Ltd. Lymphocytes-antigen presenting cells co-stimulators and uses thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100330080A1 (en) * 2008-07-02 2010-12-30 Torsten Dreier Antigen binding polypeptides
US20110008369A1 (en) * 2008-03-12 2011-01-13 Finnefrock Adam C Pd-1 binding proteins
US20140099254A1 (en) * 2012-08-14 2014-04-10 Ibc Pharmaceuticals, Inc. Combination therapy for inducing immune response to disease
US20150132217A1 (en) * 2012-08-14 2015-05-14 Ibc Pharmaceuticals, Inc. Disease therapy by inducing immune response to trop-2 expressing cells
US20160145355A1 (en) * 2013-06-24 2016-05-26 Biomed Valley Discoveries, Inc. Bispecific antibodies

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110008369A1 (en) * 2008-03-12 2011-01-13 Finnefrock Adam C Pd-1 binding proteins
US20100330080A1 (en) * 2008-07-02 2010-12-30 Torsten Dreier Antigen binding polypeptides
US20140099254A1 (en) * 2012-08-14 2014-04-10 Ibc Pharmaceuticals, Inc. Combination therapy for inducing immune response to disease
US20150132217A1 (en) * 2012-08-14 2015-05-14 Ibc Pharmaceuticals, Inc. Disease therapy by inducing immune response to trop-2 expressing cells
US20160145355A1 (en) * 2013-06-24 2016-05-26 Biomed Valley Discoveries, Inc. Bispecific antibodies

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10513558B2 (en) 2015-07-13 2019-12-24 Cytomx Therapeutics, Inc. Anti-PD1 antibodies, activatable anti-PD1 antibodies, and methods of use thereof
WO2019224385A3 (fr) * 2018-05-24 2020-01-16 Glenmark Pharmaceuticals S.A. Anticorps bispécifiques combinés et thérapies immuno-oncologiques
WO2020051248A1 (fr) * 2018-09-05 2020-03-12 Arizona Board Of Regents On Behalf Of Arizona State University Plate-forme de virus oncolytique permettant de traiter le cancer hématologique
CN113164778A (zh) * 2018-12-19 2021-07-23 拜耳公司 抗ceacam6和tim3抗体的药物组合
US11667723B2 (en) 2020-08-17 2023-06-06 Utc Therapeutics (Shanghai) Co., Ltd. Lymphocytes-antigen presenting cells co-stimulators and uses thereof

Similar Documents

Publication Publication Date Title
JP6767029B2 (ja) 疾患に対する免疫反応を誘導するための併用療法
US10662252B2 (en) Disease therapy by inducing immune response to Trop-2 expressing cells
US10245321B2 (en) Combination therapy for inducing immune response to disease
US10111954B2 (en) Combination therapy for inducing immune response to disease
AU2018236844B2 (en) T-cell redirecting bispecific antibodies for treatment of disease
US10131712B2 (en) Combination therapy with T-cell redirecting bispecific antibodies and checkpoint inhibitors
CA2937236C (fr) Traitement d'une maladie par induction d'une reponse immune a des cellules exprimant le trop-2
WO2017210058A1 (fr) Polythérapie avec des anticorps bispécifiques de redirection des lymphocytes t et des inhibiteurs de point de contrôle

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: 17807256

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17807256

Country of ref document: EP

Kind code of ref document: A1