WO2014051433A1 - BISPECIFIC IgG ANTIBODIES AS T CELL ENGAGERS - Google Patents

BISPECIFIC IgG ANTIBODIES AS T CELL ENGAGERS Download PDF

Info

Publication number
WO2014051433A1
WO2014051433A1 PCT/NL2013/050693 NL2013050693W WO2014051433A1 WO 2014051433 A1 WO2014051433 A1 WO 2014051433A1 NL 2013050693 W NL2013050693 W NL 2013050693W WO 2014051433 A1 WO2014051433 A1 WO 2014051433A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
sequence
bispecific
igg antibody
clec12a
Prior art date
Application number
PCT/NL2013/050693
Other languages
French (fr)
Inventor
Alexander Berthold Hendrik Bakker
Pieter Fokko VAN LOO
Ton Logtenberg
Original Assignee
Merus B.V.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to EA201590640A priority Critical patent/EA201590640A1/en
Priority to DK13777349.5T priority patent/DK2900694T3/en
Application filed by Merus B.V. filed Critical Merus B.V.
Priority to EP13777349.5A priority patent/EP2900694B1/en
Priority to MX2015003885A priority patent/MX2015003885A/en
Priority to PL13777349T priority patent/PL2900694T3/en
Priority to KR1020157010993A priority patent/KR102390177B1/en
Priority to BR112015006824A priority patent/BR112015006824A2/en
Priority to SG11201502451QA priority patent/SG11201502451QA/en
Priority to ES13777349.5T priority patent/ES2692951T3/en
Priority to LTEP13777349.5T priority patent/LT2900694T/en
Priority to CA2889681A priority patent/CA2889681C/en
Priority to NZ630563A priority patent/NZ630563A/en
Priority to AU2013324527A priority patent/AU2013324527B9/en
Priority to EP18192737.7A priority patent/EP3470431A1/en
Priority to CN201811462337.9A priority patent/CN110066338B/en
Priority to CN201380061615.9A priority patent/CN105051066B/en
Priority to SI201331229T priority patent/SI2900694T1/en
Priority to JP2015534421A priority patent/JP6471095B2/en
Priority to RS20181217A priority patent/RS57910B1/en
Publication of WO2014051433A1 publication Critical patent/WO2014051433A1/en
Priority to IL237945A priority patent/IL237945B/en
Priority to ZA2015/02835A priority patent/ZA201502835B/en
Priority to HK16100035.9A priority patent/HK1211966A1/en
Priority to AU2018204173A priority patent/AU2018204173B2/en
Priority to HRP20181717TT priority patent/HRP20181717T1/en
Priority to CY20181101099T priority patent/CY1120976T1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • C07K16/2851Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the lectin superfamily, e.g. CD23, CD72
    • 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
    • C07K16/3061Blood cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • 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
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/526CH3 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/53Hinge
    • 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
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/71Decreased effector function due to an Fc-modification
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the invention relates to the field of antibody engineering.
  • it relates to the field of therapeutic (human) antibodies for the treatment of diseases involving aberrant cells.
  • bispecific antibodies have been widely used for the retargeting of immune effector cells to tumor cells.
  • one binding site is directed against a tumor-associated antigen (TAA) and the second antigen against a trigger molecule on the effector cells, such as for example CD3 on T cells (Kontermann, MABS 2012 (4) 182-197; Chames and Baty, MABS 2009 (1) 539-547; Moore et al. Blood 2011 (117) 4542-4551).
  • TAA tumor-associated antigen
  • the first bispecific antibodies targeting CD3 and a tumor cell associated antigen were of rodent nature and were produced using hybrid hybridomas (Liu et al. 1985 PNAS 82: 8648, Staerz et al.
  • CD3xTAA bispecific IgG mediated induction of tumor cell lysis by polyclonal resting human T cells could not be achieved unless co-stimulation was provided by added exogenous IL-2 or anti-CD28 mAb.
  • This is exemplified by the hybrid rat IgG2b/mouse IgGl CD3xCD19 bispecific molecule that was able to induce lysis of the CD 19 positive REH B-ALL tumor cell line by resting human T lymphocytes only upon co-administration of IL-2 (Haagen et al. 1995 Blood 85:3208). Zeidler et al.
  • the strong activation potential was correlated to the hybrid subclass combination mouse IgG2a/rat IgG2b that, in contrast to other reported combinations (e.g., mouse IgG2a/mouse IgGl or rat IgG2b/mouse IgGl), not only binds but also activates Fey receptor-positive accessory cells.
  • This so-called triomAb CD3xEpcam bispecific antibody also known as catumaxomab, has been developed clinically and has been registered in Europe for palliative treatment of abdominal tumors of epithelial origin. While this bispecific antibody has clearly demonstrated clinical efficacy, its rodent nature induces anti-product immune responses upon repetitive dosing and therefore prevents a widespread application of this format.
  • CD3xTAA formats have been explored to solve both the manufacturing issues and the immunogenicity problems associated with the hybrid rodent triomAb format.
  • Such formats are often immunoglobulin-like molecules that deviate from full length human IgG molecules, and include molecules such as Dual-Affinity Re-Targeting (DARTTM) molecules that are developed by Macrogenics worldwide web at macrogenics.com/Platforms-DART.html, Bispecific T cell Engager (BiTE®) molecules that were developed by Micromet, now Amgen (Sheridan C, Nat Biotechnol.
  • DARTTM Dual-Affinity Re-Targeting
  • BiTE® Bispecific T cell Engager
  • BiTE® displays very efficient tumor cell lysis at low levels in vitro
  • administration of this bispecific format to patients is associated with significant challenges. Due to their small size, BiTE®s are rapidly cleared from the circulation and dosing of patients thus requires continuous infusion. As the dosing regimen has an overall duration of more than 2 months, this treatment has a significant impact on the quality of life of the patients.
  • IgG molecules in eradicating aberrant cells that combine a long circulatory half-life upon intravenous administration without the need for continuous infusion without being immunogenic and with only limited side effects.
  • FIG. 1 CLEC 12A and related sequences.
  • FIG. 2 T-cell activation by various antibodies: monoclonal bivalent CD3 IgG, bispecific CD3XCLEC12 IgG, bispecific CD3xisotype control IgG, monoclonal bivalent CLEC12A IgG, monoclonal bivalent isotype control IgG.
  • FIG. 3 Specific lysis of HL60 cells by CD3XCLEC12A bispecific IgG and control antibodies.
  • FIG. 4 Specific lysis of HL60 cells by CD3XCLEC12A bispecific IgG and control antibodies (E:T ratios).
  • FIG. 5 Specific lysis of HL60 cells with several CD3XCLEC12A bispecific
  • IgG molecules consisting of various CLEC12A arms & fixed CD3 arm, and control antibodies.
  • FIG. 7 Fc silencing does not affect FcRn binding.
  • FIG. 8 CD3xCLEC 12A bsAb target specific induction of T cell proliferation.
  • FIG. 9 CD8+ T cell compartment of AML patients compared to healthy donors.
  • FIG. 10 Specific CD3xCLEC12A DM-Fc induced T cell activation and HL60 tumor cell lysis by AML patient T cells.
  • FIG. 11 Specific lysis of AML blasts by autologous AML patient T cells.
  • FIG. 12 Specific monocyte lysis by patient T cells.
  • FIG. 13 Fc silencing significantly eliminates bystander cell cytokine release.
  • FIG. 14A FACS staining anti-CD3 antibodies on HPB-ALL cells
  • FIG. 14B Plate bound IgG, T cells labeled with CFSE, read out at day 5 by FACS
  • FIG 15 HL60 cytotoxicity assay
  • FIG 16 FACS staining anti-CLEC12A antibodies on HL60 cells
  • FIG. 17 HL60 cytotoxicity assay
  • FIG 18 FACS analysis
  • FIG 19 HL60 cytotoxicity assay
  • FIG 20 VH sequences of CD3-specific and CLEC12A-specific Fab arms. VL sequence of 012 common light chain. CDR sequences are bold and underlined.
  • the present invention describes a fully human IgG bispecific full length antibody for the treatment of AML.
  • One arm of the antibody binds an epitope on immune effector cells, preferably CD3, whilst the other arm targets CLEC12A, a myeloid cell specific surface target that is expressed in 90-95% of de novo and relapsed AML patients.
  • CLEC12A is expressed on AML leukemic stem cells, but not on normal haematopoietic cells.
  • CD3xCLEC12A bispecific IgGl antibody of the present invention should not induce platelet or red blood cell depletion.
  • a CD3xCLEC12A bispecific IgG antibody according to the present invention in a preferred embodiment contains a modified Fc region so as to reduce non-specific immune activation resulting from engagement of T cells and FcyR expressing cells within PBMC.
  • a bispecific human IgGl T cell engager antibody has advantages over current IgG that make use of the hybrid subclass combination mouse IgG2a/rat IgG2b, since a human IgGl will be less immunogenic and can thus be applied for repeated therapy.
  • a full length bispecific human IgGl T cell engager antibody has advantages over immunoglobulin/like molecules such as DARTTM, TandAb® or BiTE® as the full length human IgGl is not rapidly cleared from the circulation and dosing of patients will thus not require continuous infusion, which is more beneficial to patients.
  • the invention provides a bispecific IgG antibody, wherein said bispecific IgG antibody comprises one arm that specifically recognizes CLEC12A or a functional equivalent thereof, and a second arm that specifically recognizes an antigen on immune effector cells capable of recruiting such cells to an aberrant cell expressing CLEC12A or said functional equivalent.
  • the term "specifically recognizes CLEC12A or a functional equivalent thereof means that said arm has the capability of specifically recognizing CLEC12A or said functional equivalent, in the situation that CLEC12A or said functional equivalent is present in the vicinity of said antibody.
  • the term "specifically recognizes an antigen on immune effector cells” means that said arm has the capability of specifically recognizing said antigen when said antigen is present in the vicinity of said antibody.
  • antigen recognition by an antibody is typically mediated through the complementarity regions of the antibody and the specific three-dimensional structure of both the antigen and the antibody arm allowing these two structures to bind together with precision (an interaction similar to a lock and key), as opposed to random, non-specific sticking of antibodies.
  • antibodies according to the present invention that "specifically recognize CLEC12A or a functional equivalent thereof, and “specifically recognize an antigen on immune effector cells” may recognize other compounds as well, if such other compounds contain the same kind of epitope.
  • the terms “specifically recognizes CLEC12A or a functional equivalent thereof, “specifically recognizes an antigen on immune effector cells” and “specifically recognizes CD3” do not exclude binding of the antibodies to other compounds that contain the same (kind of) epitope. Instead, cross- reactivity is allowed.
  • An antibody according to the present invention is typically capable of binding CLEC12A (or a functional equivalent thereof) and an antigen on immune effector cells, preferably CD3, with a binding affinity of at least 1x10-5 M, as outlined in more detail below.
  • antibody as used herein means a proteinaceous molecule belonging to the immunoglobulin class of proteins, containing one or more domains that bind an epitope on an antigen, where such domains are derived from or share sequence homology with the variable region of an antibody.
  • Antibodies for therapeutic use are preferably as close to natural antibodies of the subject to be treated as possible (for instance human antibodies for human subjects).
  • Antibody binding can be expressed in terms of specificity and affinity. The specificity determines which antigen or epitope thereof is specifically bound by the binding domain.
  • the affinity is a measure for the strength of binding to a particular antigen or epitope.
  • Specific binding, or “specifically recognizing” is defined as binding with affinities (KD) of at least 1x10-5 M, more preferably 1x10-7 M, more preferably higher than Ixl0-9M.
  • affinities KD
  • antibodies for therapeutic applications have affinities of up to 1x10-10 M or even higher.
  • Antibodies of the present invention are typically bispecific full length antibodies of the human IgG subclass.
  • the antibodies of the present invention are of the human IgGl subclass.
  • full length IgG' is defined as comprising an essentially complete IgG, which however does not necessarily have all functions of an intact IgG.
  • a full length IgG contains two heavy and two light chains. Each chain contains constant (C) and variable (V) regions, which can be broken down into domains designated CHI, CH2, CH3, VH, and CL, VL.
  • C constant
  • V variable
  • An IgG antibody binds to antigen via the variable region domains contained in the Fab portion, and after binding can interact with molecules and cells of the immune system through the constant domains, mostly through the Fc portion.
  • Full length antibodies according to the invention encompass IgG molecules wherein mutations may be present that provide desired characteristics. Such mutations should not be deletions of substantial portions of any of the regions. However, IgG molecules wherein one or several amino acid residues are deleted, without essentially altering the binding characteristics of the resulting IgG molecule, are embraced within the term "full length IgG". For instance, such IgG molecules can have one or more deletions of between 1 and 10 amino acid residues, preferably in non-CDR regions, wherein the deleted amino acids are not essential for the binding specificity of the IgG.
  • full length IgG antibodies are preferred because of their favourable half life and the need to stay as close to fully autologous (human) molecules for reasons of immunogenicity.
  • bispecific IgG antibodies are used.
  • bispecific full length IgGl antibodies are used. IgGl is favoured based on its long circulatory half life in man.
  • the bispecific IgG antibody according to the invention is a human IgGl.
  • the term 'bispecific' (bs) means that one arm of the antibody binds to a first antigen whereas the second arm binds to a second antigen, wherein said first and second antigens are not identical.
  • said first and second antigens are in fact two different molecules that are located on two different cell types.
  • the term 'one arm [of the antibody]' preferably means one Fab portion of the full length IgG antibody.
  • Bispecific antibodies that mediate cytotoxicity by recruiting and activating endogenous immune cells are an emerging class of next-generation antibody therapeutics. This can be achieved by combining antigen binding specificities for target cells (i.e., tumor cells) and effector cells (i.e., T cells, NK cells, and macrophages) in one molecule (Cui et al.
  • bispecific antibodies are provided wherein one arm binds the CLEC12A antigen on aberrant (tumor) cells whereas the second arm binds an antigen on immune effector cells.
  • the resulting VH/VL pair will bind either antigen 1 or antigen 2.
  • Such so called “two-in-one antibodies”, described in for instance WO 2008/027236, WO 2010/108127 and Schaefer et al (Cancer Cell 20, 472-486, October 2011), are also encompassed by the term "bispecific antibody” because they also have the capability of binding two different antigens.
  • a VH is used that specifically recognizes CLEC12A, or a functional equivalent thereof
  • a VL is used that specifically recognizes an antigen on immune effector cells
  • an antibody according to the present invention comprises a VH that specifically recognizes an antigen on immune effector cells, and a VL that specifically recognizes CLEC12A, or a functional equivalent thereof. Either way, the resulting antibody typically contains two VH/VL pairs, wherein each VH/VL pair will bind either CLEC12A (or a functional equivalent thereof), or an antigen on immune effector cells. Two-in-one antibodies will typically either bind two similar antigens (AA or BB; monospecific bivalent) or bind two different antigens (AB; bispecific).
  • two-in-one antibodies are used for therapeutic applications according to the present invention, a portion of these antibodies will not exert the desired effect due to their binding to either two CLEC12A molecules (or functional equivalents thereof) or two antigens on immune effector cells, such as CD3. Since the therapeutic goal can still be achieved with a portion of the administered antibodies, two-in-one antibodies are nevertheless suitable.
  • C-type lectin domain family 12 member A also known as C-type lectin-like molecule- 1 (CLL-1), an antigen that is expressed on leukemic blast cells and on leukemic stem cells in acute myeloid leukemia (AML), including the CD34 negative or CD34 low expressing leukemic stem cells (side population)
  • AML acute myeloid leukemia
  • CD34 negative or CD34 low expressing leukemic stem cells side population
  • CLEC12A is otherwise restricted to the hematopoietic lineage, particularly to myeloid cells in peripheral blood and bone marrow, i.e., granulocytes, monocytes and dendritic cell precursors. More importantly, CLEC12A is absent on hematopoietic stem cells. This expression profile makes CLEC12A a particularly favorable target in AML.
  • Alternative names for CLEC12A include dendritic cell-associated C-type lectin-2 (DCAL-2), myeloid inhibitory C-type lectin-like receptor (MICL) and killer cell lectin-like receptor subfamily L, member 1 (KLRL1) (Zhang W. et al. GenBankTM access.no: AF247788; A.S.
  • 'CLEC12A or functional equivalent thereof means all variants that are referenced above and isoforms thereof that retain the strict myeloid expression profile (both at surface expression level and mRNA level) as described in Bakker et al. Cancer Res 2004, 64, p8443-50.
  • the invention includes bispecific IgG antibodies wherein one arm specifically recognizes functional equivalents of CLEC12A, including those functional equivalents that lack the above mentioned additional intracellular stretch of 10 amino acids.
  • Bispecific IgG antibodies according to the invention wherein one arm specifically recognizes the full length form of CLEC12A are, however, preferred.
  • the term 'aberrant cells' as used herein includes tumor cells, more specifically tumor cells of hematological origin including also pre-leukemic cells such as cells that cause myelodysplastic syndromes (MDS) and leukemic cells such as acute myeloid leukemia (AML) tumor cells or chronic myelogenous leukemia (CML) cells.
  • pre-leukemic cells such as cells that cause myelodysplastic syndromes (MDS) and leukemic cells such as acute myeloid leukemia (AML) tumor cells or chronic myelogenous leukemia (CML) cells.
  • AML acute myeloid leukemia
  • CML chronic myelogenous leukemia
  • the term 'immune effector cell' or 'effector cell' as used herein refers to a cell within the natural repertoire of cells in the mammalian immune system which can be activated to affect the viability of a target cell.
  • Immune effector cells include cells of the lymphoid lineage such as natural killer (NK) cells, T cells including cytotoxic T cells, or B cells, but also cells of the myeloid lineage can be regarded as immune effector cells, such as monocytes or macrophages, dendritic cells and neutrophilic granulocytes.
  • said effector cell is preferably an NK cell, a T cell, a B cell, a monocyte, a macrophage, a dendritic cell or a neutrophilic granulocyte.
  • recruitment of effector cells to aberrant cells means that immune effector cells are brought in close vicinity to the aberrant target cells cells such that the effector cells can directly kill, or indirectly initiate the killing of the aberrant cells that they are recruited to.
  • the bispecific antibodies of the invention specifically recognize antigens on immune effector cells that are at least over-expressed by these immune effector cells compared to other cells in the body.
  • Target antigens present on immune effector cells may include CD3, CD 16, CD25, CD28, CD64, CD89, NKG2D and NKp46,
  • the antigen on immune effector cells is CD3 expressed on T cells, or a functional equivalent thereof (a functional equivalent would be a CD3-like molecule with a similar distribution on T-cells and a similar function (in kind, not necessarily in amount)).
  • CD3 also encompasses functional equivalents of CD3.
  • the most preferred antigen on an immune effector cell is the CD3s chain. This antigen has been shown to be very effective in recruiting T cells to aberrant cells.
  • a bispecific IgG antibody according to the present invention preferably contains one arm that specifically recognizes CD3s.
  • the invention provides a bispecific full length IgG antibody, wherein said bispecific antibody comprises one arm that specifically recognizes CLEC12A or a functional equivalent thereof, and a second arm that specifically recognizes an antigen on immune effector cells capable of recruiting such cells to an aberrant cell expressing CLEC12A or said functional equivalent, wherein said immune effector cells comprise T cells.
  • the invention provides a bispecific IgG antibody according to the invention wherein said antigen on said immune effector cells is CD3 or functional equivalent thereof, preferably human CD3s.
  • the invention provides F(ab)'2 fragments of such bispecific IgG CLEC 12AxCD3 antibody.
  • bispecific IgG antibody it is an aspect of the invention to provide a bispecific IgG antibody according to the invention wherein both arms comprise a common light chain.
  • the term 'common light chain' according to the invention refers to light chains which may be identical or have some amino acid sequence differences while retaining the binding specificity of the antibody. It is for instance possible within the scope of the definition of common light chains as used herein, to prepare or find light chains that are not identical but still functionally equivalent, e.g., by introducing and testing conservative amino acid changes, changes of amino acids in regions that do not or only partly contribute to binding specificity when paired with the heavy chain, and the like.
  • the terms 'common light chain', 'common VL', 'single light chain', 'single VL', with or without the addition of the term 'rearranged' are all used herein interchangeably. It is an aspect of the present invention to use as common light chain a human light chain that can combine with different heavy chains to form antibodies with functional antigen binding domains (WO2004/009618, WO2009/ 157771, Merchant et al. 1998, Nissim et al. 1994).
  • the common light chain has a germline sequence.
  • a preferred germline sequence is a light chain variable region that is frequently used in the human repertoire and has superior ability to pair with many different VH regions, and has good thermodynamic stability, yield and solubility.
  • a most preferred germline light chain is 012, preferably the rearranged germline human kappa light chain IgVKl-39*01/IGJKl*01 (nomenclature according to the IMGT database worldwide web at imgt.org or fragment or a functional derivative thereof.
  • the terms rearranged germline human kappa light chain IgVKl-39*01/IGJKl*01, IGKV1-39/IGKJ1, huVKl-39 light chain or in short huVKl-39 are used interchangeably throughout the application.
  • common also refers to functional equivalents of the light chain of which the amino acid sequence is not identical. Many variants of said light chain exist wherein mutations (deletions, substitutions, additions) are present that do not materially influence the formation of functional binding regions.
  • a bispecific IgG antibody according to the invention wherein the arm that specifically recognizes CLEC12A or a functional equivalent thereof comprises a heavy chain CDR1 sequence consisting of a sequence that is at least 90% identical to SGYTFTSY and a heavy chain CDR2 sequence consisting of a sequence that is at least 90% identical to IINPSGGS and a heavy chain CDR3 sequence consisting of a sequence that is at least 90% identical to GTTGDWFD.
  • said heavy chain CDR 1, 2 and 3 sequences consist of a sequence that is at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99% identical to the recited CDR sequences.
  • said heavy chain CDR 1 , 2 and 3 sequences preferably contain sequences that deviate in no more than three, preferably no more than two, more preferably no more than one amino acid from the recited CDR sequences.
  • said heavy chain CDR 1, 2 and 3 sequences are identical to the recited CDR sequences.
  • the recited CDR sequences are the CDR sequences of Fab arm 4327 which, as shown in the Examples, has good CLEC12A binding properties.
  • a bispecific IgG antibody according to the invention comprises a variable heavy chain (VH) sequence that is at least 90% identical to this VH of antibody 4327.
  • a bispecific IgG antibody wherein the arm that specifically recognizes CLEC12A or a functional equivalent thereof comprises a VH sequence consisting of a sequence that is at least 90%, preferably at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99% or even 100%, identical to the sequence QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLEWMGIINPSGGS TSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCAKGTTGDWFDYWGQGTLV TVS.
  • bispecific antibodies according to the invention containing the above mentioned VH sequence, together with a VH sequence of a Fab arm recognizing CD3 (and together with a common light chain), have an excellent capacity of inducing T cell mediated lysis of CLEC12A-positive AML tumor cells.
  • a bispecific IgG antibody according to the invention wherein the arm that specifically recognizes CLEC12A or a functional equivalent thereof comprises a heavy chain CDR1 sequence consisting of a sequence that is at least 90% identical to SGYTFTSY and a heavy chain CDR2 sequence consisting of a sequence that is at least 90% identical to IINPSGGS and a heavy chain CDR3 sequence consisting of a sequence that is at least 90% identical to GNYGDEFDY.
  • said heavy chain CDR 1 , 2 and 3 sequences consist of a sequence that is at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99% identical to the recited CDR sequences.
  • said heavy chain CDR 1, 2 and 3 sequences preferably contain sequences that deviate in no more than three, preferably no more than two, more preferably no more than one amino acid from the recited CDR sequences.
  • said heavy chain CDR 1, 2 and 3 sequences are identical to the recited CDR sequences.
  • the recited CDR sequences are the CDR sequences of the VH region of antibody 4331 which, as shown in the Examples, has good CLEC12A binding properties.
  • the VH sequence of Fab arm 4331 is also shown in Figure 20.
  • a bispecific IgG antibody according to the invention comprises a VH sequence that is at least 90% identical to this VH of Fab arm 4331. Further provided is therefore a bispecific IgG antibody according to the invention, wherein the arm that specifically recognizes CLEC12A or a functional equivalent thereof comprises a VH sequence consisting of a sequence that is at least 90%, preferably at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99% or even 100%, identical to the sequence EVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLEWMGIINPSGGS TSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCARGNYGDEFDYWGQGTLV TVSS.
  • bispecific antibodies according to the invention containing the VH sequence of Fab arm 4331, together with a VH sequence of a Fab arm recognizing CD3, (together with a common light chain) have an excellent capacity of inducing T cell mediated lysis of CLEC12A-positive AML tumor cells.
  • Yet another preferred embodiment provides a bispecific IgG antibody according to the invention wherein the arm that specifically recognizes CLEC12A or a functional equivalent thereof comprises a heavy chain CDR1 sequence consisting of a sequence that is at least 90% identical to SGYTFTGY and a heavy chain CDR2 sequence consisting of a sequence that is at least 90% identical to WINPNSGG and a heavy chain CDR3 sequence consisting of a sequence that is at least 90% identical to DGYFADAFDY.
  • said heavy chain CDR 1, 2 and 3 sequences consist of a sequence that is at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99% identical to the recited CDR sequences.
  • said heavy chain CDR 1 , 2 and 3 sequences preferably contain sequences that deviate in no more than three, preferably no more than two, more preferably no more than one amino acid from the recited CDR sequences.
  • said heavy chain CDR 1, 2 and 3 sequences are identical to the recited CDR sequences.
  • the recited CDR sequences are the CDR sequences of the VH of antibody 3918 which, as shown in the Examples, also has good CLEC12A binding properties.
  • the VH sequence of the antibody 3918 is also shown in Figure 20.
  • a bispecific IgG antibody according to the invention comprises a VH sequence that is at least 90% identical to this VH of antibody 3918. Further provided is therefore a bispecific IgG antibody according to the invention, wherein the arm that specifically recognizes CLEC12A or a functional equivalent thereof comprises a VH sequence consisting of a sequence that is at least 90%, preferably at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99% or even 100%, identical to the sequence QVQLVQSGAEVKKPGASVKVSCKASGYTFTGYYMHWVRQAPGQGLEWMGWINPNSG GTNYAQKFQGRVTMTRDTSISTAYMELSRLRSDDTAVYYCARDGYFADAFDYWGQGT LVTVSS.
  • bispecific antibodies according to the invention containing the VH sequence of Fab arm 3918, together with the VH sequence of a Fab arm recognizing CD3, (and together with a common light chain) also have a good capacity of inducing T cell mediated lysis of CLEC12A-positive AML tumor cells.
  • a bispecific IgG antibody according to the invention wherein the second arm specifically recognizes CD3 and comprises a heavy chain CDR1 sequence consisting of the sequence SYGMH and a heavy chain CDR2 sequence consisting of the sequence IIWYSGSKKNYADSVKG and a heavy chain CDR3 sequence consisting of the sequence GTGYNWFDP.
  • said CD3-specific arm comprises a VH sequence consisting of the sequence
  • the recited CDR sequences and VH sequence are the sequences of antibody 3896. These sequences are also depicted in Figure 20. A heavy chain comprising these CD3-specific CDR sequences and/or the recited VH sequence of Fab arm 3896 is preferred for a bispecific IgG antibody according to the invention, because these sequences provide the bispecific antibody with an optimal affinity for CD3-expressing immune cells, while simultaneously allowing sufficient binding to CLEC12A-positive AML tumor cells.
  • the overall effect of a bispecific antibody is determined by the combined effect of the affinity of one arm for antigen 1 and the affinity of the other arm for antigen 2.
  • an antibody of the present invention having a specificity for CLEC12A (or a functional equivalent thereof) and an antigen on immune effector cells (preferably CD3), an optimized timing of binding to CD3-positive immune cells and CLEC12A-expressing tumor cells is preferred in order to efficiently induce T cell mediated lysis of CLEC12A-positive tumor cells. It is hypothesized that the balance between affinities of a CLEC12A/CD3 bispecific antibody is of utmost importance.
  • CLEC12A -positive tumor cells are first bound by one or more bispecific antibodies according to the invention, where after T cells are attracted by the free CD3 arm of the bispecific antibody and subsequent T cell activation takes place.
  • CD3 positive T cells and CLEC12A-positve tumor cells are bound essentially simultaneously by the bispecific antibody.
  • the affinities for both CLEC12A (or a functional equivalent thereof) and for an antigen on immune effector cells (preferably CD3) are preferably chosen or modulated such that the right balance is achieved, i.e.
  • such excellent balance between the binding affinities for CD3 and CLEC12A is preferably achieved by combining a VH having the CDR sequences (or whole VH sequence) of Fab arm 3896 (which is specific for CD3) with a VH having the CDR sequences (or whole VH sequence) of either Fab arms 4327 or 4331 or 3918 or 3116 (which are specific for CLEC12A).
  • Such resulting bispecific antibodies display a favorable balance between the binding affinities for CD3 and CLEC12A, so that T cells and CLEC12A-positive AML tumor cells are efficiently brought together, and T cell mediated lysis of CLEC12A-positive AML tumor cells is optimally induced.
  • a bispecific IgG antibody according to the present invention is preferably provided wherein both arms comprise a common light chain variable domain.
  • a particularly preferred common light chain is the human rearranged kappa light chain IgVKl-39*01/IgJKl*01, also named 012.
  • the nucleotide and amino acid sequence of the 012 VL are also depicted in Figure 20.
  • the CDR sequences are bold and underlined.
  • a bispecific antibody according to the invention containing a common light chain that at least comprises the CDR sequences of 012 is therefore preferred.
  • One aspect of the invention therefore provides a bispecific IgG antibody according to the invention, wherein the first and the second arms further comprise a light chain CDR1 sequence consisting of a sequence that is at least 90% identical to RASQSISSYLN and a light chain CDR2 sequence consisting of a sequence that is at least 90% identical to AASSLQS and a light chain CDR3 sequence consisting of a sequence that is at least 90% identical to QQS YSTPPT.
  • said light chain CDR 1 , 2 and 3 sequences consist of a sequence that is at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99% identical to the recited CDR sequences.
  • said light chain CDR 1, 2 and 3 sequences preferably contain sequences that deviate in no more than three, preferably no more than two, more preferably no more than one amino acid from the recited CDR sequences.
  • said light chain CDR 1, 2 and 3 sequences are identical to the recited CDR sequences.
  • a bispecific IgG antibody according to the invention comprises a VL sequence that is at least 90% identical to the 012 VL chain.
  • first and the second arms comprise a VL sequence consisting of a sequence that is at least 90%, preferably at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99% or even 100%, identical to the sequence DIQMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQKPGKAPKLLIYAASSLQSGVPSR FSGSGTDFTLTISSLQPEDFATYYCQQSYSTPPTFGQGTKVEIK.
  • % identical to is defined herein as the percentage of residues in a candidate amino acid sequence that is identical with the residues in a reference sequence after aligning the two sequences and introducing gaps, if necessary, to achieve the maximum percent identity.
  • Methods and computer programs for the alignment are well known in the art.
  • One computer program which may be used or adapted for purposes of determining whether a candidate sequence falls within this definition is "Align 2", authored by Genentech, Inc., which was filed with user documentation in the United States Copyright Office, Washington, D.C. 20559, on Dec. 10, 1991.
  • a bispecific full length IgG antibody according to the invention by definition has two different antigen binding sites but the Fc region of the IgG also comprises a third binding site for an Fc receptor. If a cell carries both an Fc receptor and one of the targets of the bispecific antibody, cross-linking of the Fc receptor and said target on the surface of said cell may occur, which may lead to undesired effects.
  • the invention provides a bispecific full length IgG antibody according to the invention, wherein said bispecific IgG antibody has mutated lower hinge and/or CH2 domains such that interaction of said bispecific IgG antibody with Fc gamma (Fey) receptors is significantly reduced.
  • the term "such that interaction of said bispecific IgG antibody with Fc gamma receptors is significantly reduced” means that the capability of said bispecific IgG antibody of interacting with Fc gamma receptors, if such Fc gamma receptors are present in the vicinity of said antibody, is reduced.
  • a region of the antibody, preferably the lower hinge and/or the CH2 domain of the antibody is mutated (typically by expressing a mutated nucleic acid sequence encoding it) whereby the ability to interact with an Fc receptor is diminished. It is preferred that the interaction with the Fc receptor is essentially abolished.
  • said mutated lower hinge and/or CH2 domains comprise at least one substitution at amino acids positions 235 and/or 236 (numbering according to Kabat). Preferably, both amino acids positions 235 and 236 are substituted.
  • substitutions at these sites are capable of essentially preventing the interaction between the bispecific antibody and the Fc receptor present on the tumor cells.
  • substitutions L235G and/or G236R are very suitable for that purpose.
  • a bispecific IgG antibody according to the invention, wherein said mutated CH2 and/or lower hinge domains comprise substitution L235G and/or G236R is therefore also provided herein.
  • both L235G and G236R are substituted.
  • a person skilled in the art may introduce lower hinge and/or the CH2 domain mutations that comprise the substitutions 234F, 235E and/or 33 IS (Oganesyan et al. Biol. Crystall. 2008(D64)700).
  • all three substitutions are introduced in this alternative.
  • the invention provides a method for producing a bispecific full length IgG antibody according to the invention from a single cell, wherein said bispecific full length IgG antibody comprises two CH3 domains that are capable of forming an interface, said method comprising providing in said cell a) a first nucleic acid molecule encoding a 1st CH3 domain-comprising polypeptide chain, b) a second nucleic acid molecule encoding a 2nd CH3 domain-comprising polypeptide chain, wherein said nucleic acid molecules are provided with means for preferential pairing of said 1st and 2nd CH3 domain-comprising polypeptides, said method further comprising the step of culturing said host cell and allowing for expression of said two nucleic acid molecules and harvesting said bispecific full length IgG antibody from the culture.
  • Said first and second nucleic acid molecules may be part of the same vector or gene delivery vehicle and may be integrated at the same site of the host cell's genome. Alternatively, said first and second
  • a preferred embodiment provides a method for producing a full length bispecific IgG antibody according to the invention from a single cell, wherein said bispecific IgG antibody comprises two CH3 domains that are capable of forming an interface, said method comprising providing: - a cell having a) a first nucleic acid sequence encoding a IgG heavy chain that specifically recognizes CLEC12A and that contains a 1st CH3 domain, and b) a second nucleic acid sequence encoding a IgG heavy chain that specifically recognizes an antigen on immune effector cells, preferably CD3, and that contains a 2nd CH3 domain, wherein said nucleic acid sequences are provided with means for preferential pairing of said 1st and 2nd CH3 domains, said method further comprising the step of culturing said cell and allowing for expression of said two nucleic acid sequences and harvesting said bispecific IgG antibody from the culture.
  • said cell also has a third nucleic acid sequence encoding a common light chain.
  • a preferred common light chain is 012, preferably the rearranged germline human kappa light chain IgVKl-39*01/IGJKl*01, as described above.
  • the preferred mutations to produce essentially only bispecific full length IgG molecules are the amino acid substitutions L351K and T366K (numbering according to Kabat) in the first CH3 domain and the amino acid substitutions L351D and L368E in the second CH3 domain, or vice versa.
  • a method according to the invention for producing a bispecific IgGl antibody wherein said first CH3 domain comprises the amino acid substitutions L351K and T366K (numbering according to Kabat) and wherein said second CH3 domain comprises the amino acid substitutions L351D and L368E, said method further comprising the step of culturing said cell and allowing for expression of said nucleic acid sequences and harvesting said bispecific antibody from the culture.
  • Antibodies that can be produced by these methods are also part of the present invention.
  • the invention further provides a pharmaceutical composition
  • a pharmaceutical composition comprising a bispecific IgG antibody according to the invention and a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier includes any and all solvents, salts, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • the active compound may be coated in a material to protect the compound from the action of acids and other natural conditions that may inactivate the compound.
  • the antibodies and pharmaceutical compositions according to the invention find their use in the treatment of various leukemias and pre-leukemic diseases of myeloid origin but also B cell lymphomas.
  • Diseases that can be treated according to the invention include myeloid leukemias or pre-leukemic diseases such as AML, MDS and CML and Hodgkin's lymphomas and most non-Hodgkin's lymphomas.
  • myeloid leukemias or pre-leukemic diseases such as AML, MDS and CML and Hodgkin's lymphomas and most non-Hodgkin's lymphomas.
  • MDS myelodysplastic syndrome
  • CML chronic myelogenous leukemia
  • AML acute myeloid leukemia
  • a bispecific IgG antibody according to the invention in the preparation of a medicament for the treatment or prevention of myelodysplastic syndrome (MDS), chronic myelogenous leukemia (CML) or preferably acute myeloid leukemia (AML).
  • MDS myelodysplastic syndrome
  • CML chronic myelogenous leukemia
  • AML acute myeloid leukemia
  • the amount of antibody according to the invention to be administered to a patient is typically in the therapeutic window, meaning that a sufficient quantity is used for obtaining a therapeutic effect, while the amount does not exceed a threshold value leading to an unacceptable extent of side-effects.
  • An antibody according to the invention exerting sufficient therapeutic effects at low dosage is, therefore, preferred.
  • AML acute myeloid leukemia
  • CD3xCLEC12A bispecific IgG antibodies that efficiently induce AML tumour cell lysis.
  • CD3xCLEC12A bispecific antibodies thus are a targeted therapy with fewer side effects that specifically eradicates leukemic stem cells in order to improve the prognosis of AML patients. Because CLEC12A is expressed on leukemic stem cells (LSC) and not on normal haematopoietic stem cells, therapy directed against this antigen (as has been shown in vitro) will eradicate the LSC while sparing the normal stem cell.
  • LSC leukemic stem cells
  • IgG bispecific antibodies are clinically evaluated in relapsed AML patients.
  • the clinical efficacy is analyzed using AML blast reduction in the bone marrow as an objective response criterion.
  • An efficacious bispecific IgG for AML provides a novel therapeutic option for a large patient segment for which there is currently no treatment available. In addition to providing a means to achieve durable remissions, this treatment option also has a curative potential for AML when applied during remission.
  • Example 1 Generation and functional characterization of a candidate
  • a candidate CD3XCLEC12A bispecific IgGl was generated for which the CD3 and CLEC12A Fab arms were derived from antibodies previously described.
  • the VH region from anti-CD3 antibody 15C3 one of the CD3-specific antibodies as disclosed in WO2005/118635, was used and this VH is referred to as '3056'.
  • VH regions were cloned into expression vectors using methods known in the art for production of bispecific IgGl (Gunasekaran et al. JBC 2010 (285) 19637-19646; WO2009/089004), in conjunction with the rearranged human IGKV1-39/IGKJ1 (huVKl-39) light chain.
  • the huVKl-39 was previously shown to be able to pair with more than one heavy chain thereby giving rise to antibodies with diverse specificities, which facilitates the generation of bispecific molecules (De Wildt RM et al. J. Mol. Biol. 1999 (285) 895-901 ; De Kruif et al. J. Mol. Biol. 2009 (387) 548-58; WO2009/157771).
  • Table 1 Binding to cell-expressed CD3 and CLEC12A by flow cytometry.
  • results are given as the mean fluorescent intensity.
  • Affinity measurements of the candidate 3056x3116 bispecific IgGl for CD35/8 and the extracellular domain of CLEC12A are determined by surface plasmon resonance (BIAcore). Briefly, purified recombinant antigens are covalently coupled to the surface of a CM5 sensor chip using free amine chemistry: antigens are diluted in a kAc buffer to 10 ⁇ g/ml and coupled to a surface that is activated with NHS/EDC (according to the manufacturer's recommendations).
  • Fab arms present in bispecific antibodies are serially diluted to 100, 50, 20, 10, 1 and 0.1 nM in Hepes buffered saline (HBS) and flowed over the antigen-coupled surface of the CM5 sensor chip at a high (30 ⁇ 1/ ⁇ ) flow rate (to prevent re-binding).
  • HBS Hepes buffered saline
  • FC1 Flow cell 1
  • FC2 and FC3 are used for the two different antigens recognized by the bispecific antibody, to be able to measure the affinities of both Fab arms in a single kinetic run over all three surfaces.
  • the on-rates (that are concentration-dependent) of bispecific antibodies are simultaneously measured on the two different antigens they recognize. Sensorgrams of the association and dissociation phases of the different bispecific proteins are thus obtained.
  • the affinities of the Fab arms are determined.
  • the setup of the experiment is reversed: the bispecific antibody is covalently coupled to the surface of the sensor chip using free amine chemistry and recombinant purified antigen is flowed over the surface at a high (30 ⁇ 1/ ⁇ ) flow rate to measure the affinity of the Fab arm directed to that antigen.
  • T-cell stimulatory capacity was investigated with healthy donor resting T-cells. Briefly, peripheral blood was obtained from healthy donors after informed consent. T-cells were isolated by standard density gradient isolation to enrich for peripheral blood mononuclear cells (PBMC), followed by negative selection using magnetic beads (pan T-cell kit, Miltenyi Biotec, cat.no.130-091-155). Using this purification strategy, T-cells were so-called 'untouched' (i.e., not stained by antibodies, so-called 'resting T cells') to limit the possibility of pre-activation.
  • PBMC peripheral blood mononuclear cells
  • Both the bivalent CD3 IgG and the CD3XCLEC12A bispecific IgG efficiently induced upregulation of the T-cell activation markers CD69 and CD25 on CD4-positive and CD8-positive T-cells (FIG. 2).
  • FBS which did not block Fc receptors present on HL60 cells
  • CD3Xisotype control IgG was shown to induce T-cell activation. This effect was diminished in the presence of HS, suggesting that the observed T-cell activation by monovalent CD3 binding of the CD3Xisotype control IgG was dependent on Fc cross-linking.
  • T-cell activation induced by the candidate 3056x3116 CD3xCLEC12A bispecific IgG was only partially dependent on Fc-inter actions, as the potency to upregulate CD69 and CD25 was largely maintained in the presence of HS (FIG. 2). This indicated that the intrinsic potency of monovalent CD3 binding was sufficient to activate T-cells when the binding molecule bridged to the CLEC12A antigen on the HL60 target cells following binding with the other Fab arm.
  • the HL60 cells in this assay were labeled with carboxyfluorescein diacetate succimidyl ester (CFSE) and cocultured with T-cells at various effector: target cell ratios. After one, two or three days, surviving CFSE-positive HL60 cells were quantified by flow cytometry. Results were expressed as the percentage of specific lysis related to PBS.
  • CFSE carboxyfluorescein diacetate succimidyl ester
  • CD3 monospecific bivalent IgG induced resting T-cell mediated killing of HL60 cells (FIG. 3).
  • CD3XCLEC12A bispecific monovalent IgG and the control CD3Xisotype control also induced resting T-cell mediated killing of HL60 cells.
  • this example demonstrates that a CD3xCLEC12A bispecific molecule is a potent inducer of T-cell mediated tumor cell lysis and confirms our hypothesis that T cell engagement for effective killing of aberrant cells can be mediated by a CD3xCLEC12A full length IgGl bispecific antibody.
  • the activity induced by the CD3XCLEC12A bispecific IgG is not dependent on Fey receptor interactions.
  • panels of CD3 Fab arms and CLEC12A Fab arms are generated.
  • Example 1 showed that CD3xCLEC12A bispecific molecules can be potent inducers of T-cell mediated tumor cell lysis. Therefore, to generate more extensive panels of such bispecific molecules separate panels of CD3 binders as well as CLEC12A binders were generated.
  • CD3s-specific VH regions are generated by immunization of mice transgenic for the huVKl-39 light chain (WO2009/ 157771) and for a human heavy chain (HC) minilocus with CD3s in various formats: (1) isolated CD35/8 or CD3y/s that may be fused of coupled to a carrier molecule (such as human IgG-Fc or a His-tag) as known in the art with or without adjuvant, (2) cells expressing CD35/8 or CD3y/s, or (3) DNA construct encoding CD35/8 or CD3y/8, or a combination of these strategies.
  • a carrier molecule such as human IgG-Fc or a His-tag
  • Antigen-specific Fab arms are selected from phage libraries from immunized mice or from synthetic phage display libraries which contain the VL region of the huVKl-39 light chain and a collection of human VH regions. For generation of synthetic libraries, randomized CDR3 primers were used as described in De Kruif et al.
  • Bacteriophages from these libraries are selected in multiple rounds for binding to isolated CD35/8 protein that may be coupled to a carrier molecule (see above) or to cells expressing CD3s such as HPB-ALL or cells transfected to express CD35/8 or CD3y/8, or a combination of these strategies.
  • Non-binding phages are removed and binding phages are eluted with an acidic buffer or, to direct the selected Fab repertoire to a desired specificity, with antibodies against a specific epitope, for example with antibodies that are cross-reactive to cynomolgous CD3s.
  • These phages are then transfected into competent bacteria which were grown under selection pressure for phage-containing bacteria. After picking a number of surviving bacterial colonies, phages are rescued and submitted to the next selection round.
  • phages are screened for binding to cell-expressed antigen by flow cytometry and to isolated antigen by ELISA.
  • benchmark CD3 antibodies are used such are known in the art, e.g., OKT-3.
  • Nucleotide material from essentially all phages that showed specific binding to antigen-expressing cells is submitted to colony PCR to amplify the VH regions and sequence PCR to determine the VH region sequence. The resulting sequences are clustered based on uniqueness of their HCDR3.
  • VH sequences are further grouped based on the likelihood of a unique VDJ (i.e., if HCDR3 in different clusters contain ⁇ 2 amino acids difference, they are considered part of the same cluster and are grouped together). From each cluster, one or a few VH regions per cluster are selected for cloning into vectors for expression in a IgG monospecific bivalent format in conjunction with the huVKl-39 light chain. VH regions for which specific binding to isolated antigen and cell-expressed antigen is confirmed are subsequently cloned in vectors for expression in a CD3XCLEC12A bispecific format. These are then characterized to select a candidate with therapeutic potential (see following examples). [0067] Example 3: Generation and characterization of CLEC12 Fab arms for
  • CD3xCLEC12 bsAb As it was demonstrated in Example 1 that CD3xCLEC12A bispecific molecules have the potency to induce T-cell mediated tumor cell lysis, we next wished to establish more extensive panels of such bispecific molecules. In addition to the panel of CD3 binders as described in Example 2 we also generated a panel of CLEC12A binders.
  • CLEC 12 A- specific Fab arms were selected from Fab synthetic phage display libraries which contained the rearranged human IGKV1-39/IGKJ1 VL region and a collection of human VH regions (De Kruif et al. Biotechnol Bioeng. 2010 (106)741-50). Bacteriophages from these banks were selected in two rounds for binding to CLEC12A. This was done by incubation with the extracellular domain of CLEC12A (amino acids 75 to 275) coupled to a His-tag (Sino Biological, cat.no. 11896-H07H) which was coated to a surface.
  • Non-binding phages were removed, binding phages were chemically eluted, and used to infect bacteria which were grown under selection pressure for phage-containing bacteria. After picking a number of surviving bacterial colonies, phages were rescued and submitted to the next round of selection and propagation.
  • CLEC12A expressed on the tumor cell line HL60 by flow cytometry.
  • the CLEC12A benchmark antibody was used as a positive control for binding.
  • Nucleotide material from essentially all phages that showed specific binding to CLEC12A-expressing cells was submitted to colony PCR to amplify the VH regions and sequence PCR to determine the VH region sequence. The resulting sequences were clustered based on uniqueness of their HCDR3.
  • the VH regions from each unique HCDR3 cluster were cloned into vectors for expression in IgG monospecific or bispecific formats in conjunction with the rearranged human IGKV1-39/IGKJ1 LC.
  • Example 4 Selection of functional CLEC 12 Fab arm for CD3xCLEC 12 bsAb
  • CD3XCLEC12A bispecific molecules were then functionally tested in a target cell lysis assay as described in Example 1. Results were expressed as the percentage of specific lysis related to the isotype control. All candidate CLEC12A Fab arms showed a dose-dependent specific lysis of HL60 target cells in the bispecific format, with kinetics that were similar to or better than when the CLEC12A benchmark Fab arm used (FIG. 5). [0078] Also, the CD3xisotype control bsAb showed a dose-dependent target cell lysis, although 1 log higher concentrations were required for the same extent of specific lysis.
  • Example 5 efficacy of CD3xCLEC12 product candidates using AML T cells and/or AML tumor cells
  • Examples 1 and 4 demonstrated the potency of CD3XCLEC12A bispecific IgG using either CD3 Fab arm 3056 or 3896 and using the CLEC12A Fab arm candidates 4327, 4331 or 3918 or the CLEC12A benchmark Fab arm 3116 in inducing HL60 target cell lysis mediated by healthy donor resting T-cells.
  • T-cells derived from patients with AML one of the primary indications for therapeutic application of a CD3XCLEC12A bispecific drug, can be stimulated to kill tumor targets upon stimulation with a CD3XCLEC12A bispecific full length IgG.
  • patient-derived T-cells can kill autologous AML tumor cell blasts upon stimulation with a CD3XCLEC12A bispecific full length IgG.
  • T-cells are isolated from peripheral blood of AML patients according to procedures as described in Example 1. Purified patient-derived T-cells are then incubated with
  • the T-cell mediated target cell lysis assay is performed with AML tumor blasts isolated from the same patient (Norde et al. Blood 2009 (113)2312). Isolated blasts are then labeled with CFSE and cocultured with autologous patient-derived T-cells in the presence of the cytokine mixture as described in Norde et al. and in the presence of the
  • Target cell lysis is monitored as described in
  • Example 6 Cytokine release by T cells after contact with CD3XCLEC12A bispecific IgG
  • T-cell stimulatory biologicals overstimulation of T-cells is a serious risk as this may lead to cytokine release syndrome (Suntharalingam et al. 2006, New England J Med 355(10), pages 1018-1028; Chatenoud et al. 1990, Transplantation 49(4), pages 697-702).
  • T-cell stimulation induced by CD3XCLEC12A bispecific IgG the induction of T-cell cytokines was studied in a coculture of T-cells and Fc receptor-expressing target cells.
  • IFNy, TNFa and IL-2 (Table 3), which are considered to mainly drive cytokine release syndrome.
  • production of IL-4, IL-6, IL-8 and IL-10 was increased by incubation with CD3 IgG.
  • CD3XCLEC12A bispecific IgG only induced IL-8 production to a similar level as CD3 IgG; the other cytokines were not significantly induced by the bispecific IgG.
  • GM-CSF was below the detection limit in any condition.
  • Table 3 antibody induced cytokine release by T cells.
  • Results are given as the average concentration of cytokine in pg/ml of two donor ⁇ standard deviation. [0088] The data shown here suggest a favorable therapeutic profile for the different
  • CD3XCLEC12A bispecific IgG molecules, as they potently induce target cell lysis (Examples 1 and 4) without triggering T-cells to secrete potentially harmful amounts of pro-inflammatory cytokines as observed with CD3 IgG.
  • Example 7 Effect of Fc silencing on in vitro efficacy of CD3XCLEC12A bsAb
  • Example 4 was suggested to be due to interaction of the bsAb Fc part with Fc receptors on HL60 target cells. As such target non-specific cell lysis may als occur in vivo, either by interaction with Fc receptors on target cells or on bystander cells such as NK cells, engineering of the CH2/lower hinge region was employed to induce silencing of Fc-mediated activity of the bsAb.
  • CD3XCLEC12A bsAbs (3056x3116) with either a DM-Fc or a TM-Fc were generated and confirmed to bind CLEC12A-expressing cells by flow cytometry with the same intensity as the bsAb with wild type Fc (data not shown).
  • these bsAbs and the wild type, DM-Fc and TM-Fc versions of the CD3Xisotype control bsAb were tested in the HL60 target cell lysis assay (see Examples 1 and 4). Results were expressed as the percentage of specific lysis related to the isotype control.
  • Fc silencing either by the DM or by the TM had no or only a minor influence on the extent of HL60 cell specific lysis induced by CD3XCLEC12A bsAb (FIG. 6).
  • CD3Xisotype control bsAb the potency to induce lysis of HL60 cells was significantly reduced with the TM and even further with the DM.
  • Binding of the candidate 3056x3116 CD3XCLEC12A bsAb with WT Fc or with silenced DM-Fc or a TM-Fc to human FcRn was determined by Bio-Layer Interferometry (BLI, Octet QK, ForteBio).
  • Binding of CD3XCLEC12A bsAb with silenced Fc to CD 16, CD32 and CD64 is determined by Bio-Layer Interferometry (BLI, Octet QK, ForteBio). Protocol in short: purified CD3XCLEC12A WT Fc IgGl, DM-Fc IgGl or TM-Fc IgGl is captured to Protein L biosensors (ForteBio, Cat no 18-5085) at a concentration of 50 ⁇ g/ml in lx Kinetics Buffer (ForteBio 18-5032) at RT.
  • CD 16 (Sino Biological Inc, 10389-H08H1)
  • CD32 (Sino Biological Inc, 10374-H08H)
  • CD64 (Sino Biological Inc, 10256-H08H) protein
  • Binding of CD3XCLEC 12A bs Ab with silenced Fc to human C 1 q is determined by capture ELISA.
  • purified CD3XCLEC12A WT Fc IgGl, DM-Fc IgGl or TM-Fc IgGl is coated in a concentration range of 25-0.012 ⁇ g/ml in PBS on Nunc-Immuno maxisorp F96 plate (Nunc, 439454) O/N at 4C.
  • human Clq (Quidel, A400) is added at 2.0 ⁇ g/ml in ELISA buffer (2%MILK/PBST).
  • the complex is then visualized using sheep-anti-human Clq polyclonal IgG (Meridian, K90020C) and rabbit-anti-sheep HRP conjugated polyclonal IgG (Southern Biotech, 6150-05). Finally, using TMB substrate (BD 51-2606KC/51-2607KC) binding is developed and OD450 is quantified using a Micro plate reader (Multiskan EX, Thermo Electron Corporation).
  • Example 9 Evaluation of in vivo efficacy of CD3xCLEC12A bispecific IgG.
  • Animal xenograft studies using luciferase expressing HL60 cells (HL60(-Luc) cells) are performed to confirm and extend the in vitro findings using the CD3xCLEC12A bispecific IgGl. More specifically these studies are performed to determine the steady state plasma concentrations at effective doses, which will be taken into account in setting the starting dose for the Phase 1 clinical evaluation.
  • NOD/SCID mice or comparable immune-compromised mice
  • tumor dimensions are scored 1 week after the initial HL60(Luc) inoculation. The arithmetic average of tumor dimensions (either denoted as tumor volumes or as total bioluminescence) from each group is plotted against time.
  • Example 10 Use of a bispecific full length IgGl antibody CD3xCLEC12A in a phase Ia/Ib study.
  • the final lead CD3xCLEC12A bispecific full length IgGl candidate is used to manufacture GMP grade material and is clinically evaluated in AML patients.
  • a formal non-clinical safety analysis of the product candidate is performed to establish a safe starting dose for first in man studies.
  • an open-label, multi-centre dose escalation Phase Ia/b trial is performed in relapsed and/or refractory AML and in patients unfit for intensive treatment, to explore the safety and tolerability of the CD3xCLEC12A bispecific IgG upon i.v. administration.
  • Secondary endpoints include pharmacokinetic and pharmacodynamic characterization and preliminary efficacy analysis.
  • Example 11 Capacity of CD3xCLEC 12A bsAb to induce T cell proliferation.
  • T cell numbers are low compared to the amount of AML blasts at diagnosis. It is well known that T cells undergo proliferation upon activation resulting in an increased number of T cells.
  • a CD3xCLEC12A bsAb can activate T cells and has the potency to induce T-cell mediated tumor cell lysis.
  • AML patients treated with CD3xCLEC12A bsAb benefit from expansion of T cell subsets upon CD3xCLEC12A bispecific molecule mediated T cell activation as T cell proliferation will result in an increased number of effector T cells.
  • CD3xCLEC12A bsAb induces in vitro T cell proliferation
  • resting T cells were purified, labeled with carboxyfluorescein diacetate succimidyl ester (CFSE) and co-cultured with autologous CLEC12A+ monocytes in the presence of CD3xCLEC12A bsAb or control Abs.
  • CFSE carboxyfluorescein diacetate succimidyl ester
  • CD3xCLEC12A bsAbs with the DM-Fc tail was used.
  • CD3xisotype control WT-Fc bsAb As controls, a CD3xisotype control WT-Fc bsAb, a CD3xisotype control DM-Fc bsAb, a monoclonal CD3 with WT-Fc and an irrelevant isotype control (IgG with WT-FC) were included.
  • Monocytes and T cells from healthy donor peripheral blood were isolated by standard density gradient isolation to enrich for peripheral blood mononuclear cells (PBMC), followed by a CD 14 positive selection for monocytes using CD 14 microbeads (human CD 14 microbeads, Miltenyi Biotec, cat.no.
  • pan T-cell isolation kit Miltenyi Biotec, cat.no. 130- 096-535
  • the pan T-cell isolation kit allows isolation of resting (untouched) T cells (i.e. not stained with antibodies) avoiding the possibility of pre- activation of T cells.
  • CFSE-labeled purified resting T cells were subsequently incubated with purified monocytes and bsAbs in medium with 10% normal human serum (HS) at an effector: target cell ratio of 5: 1 for seven days. At day 7 decrease of CFSE signal as read-out for T cell proliferation was measured by flow cytometry. Results were expressed CFSE signal per CD3+, CD3+CD4+ or CD3+CD8+ T cells in histograms.
  • HS normal human serum
  • CD3xCLEC12A bsAb is not only capable of target specific induction of T cell mediated tumor lysis as demonstrated previously, but can also potently induce target specific T cell proliferation resulting in an increased number of T cells. Moreover this further demonstrates that Fc silencing by CH2/lower hinge engineering not only contributes to target-specific killing of aberrant cells but also to target-specific induction of T cell proliferation by the CD3xCLEC12A DM-Fc bsAb IgG.
  • Example 12 Evaluation of CD3xCLEC12A induced expansion of TEMRA subset from AML patients.
  • CD3xCLEC12 DM-Fc bsAb As activation of T cell proliferation was demonstrated for CD3xCLEC12 DM-Fc bsAb, we next wished to investigate whether CD3xCLEC12A DM-Fc bsAb is capable of inducing proliferation of the CD8+ cytotoxic T cell compartment in AML patients. CD8 + cytotoxic T cells have been recognized as the main effectors mediating tumor regression (Sluijter et al., 2010).
  • CD8+ T cells can be divided into four subsets: naive (CCR7+CD45RA+), central memory (T CM , CCR7+CD45RA-), effector memory (T EM , CCR7-CD45RA-), and CD45RA+ effector memory (TEMRA, CCR7-CD45RA+) cells.
  • naive and memory CD8+ T-cell subsets have different capacities to proliferate and differentiate in response to TCR stimulation (Geginat et al., 2003).
  • PBMC peripheral blood samples from AML patients and healthy donors by standard density gradient isolation.
  • PBMCs were stained with CCR7, CD3, CD4, CD8, CD45RA and CD45RO antibodies to analyze for the CD8+ T cell subsets by flow cytometry. Results were expressed as percentage of a subset in the total CD8+ T cell compartment.
  • resting T cells from AML patients in clinical remission are purified according to example 1 1.
  • resting T cells are either labeled with CFSE or not labeled (CFSE labeling as described in example 11) and co-cultured with HL60 leukemia cells at an E:T ratio 5: 1 with control or test antibodies for 7 days.
  • CFSE labeled T cells are used for quantification of T cell proliferation, whereas unlabeled T cells are used to determine the percentage of proliferated T cell subsets.
  • CFSE-labeled and unlabeled T cells are incubated with PBS, isotype control WT-Fc Ab, CD3xCLEC12A DM-Fc bsAb, CD3xisotype control DM-Fc bsAb and CD3 monoclonal Ab with WT-Fc at 1 ⁇ g/ml.
  • CFSE labeled T cell are stained with CD3, CD4 and CD8 antibodies and subjected to FACS analysis to determine the absolute T cells numbers and number of cell divisions
  • unlabelled CFSE T cells are stained with CCR7, CD3, CD4, CD8, CD45RA and CD45RO antibodies to determine composition of the proliferated CD8+ T cell subsets by flow cytometry.
  • T cell proliferation is expressed as CFSE signal per T cell subset in histograms and the size of the four CD8+ T cells subsets is expressed as percentage within the total CD8+ T cell compartment.
  • Example 13 Efficacy of CD3xCLEC12A bsAb to induce AML patient T cell mediated tumour cell lysis.
  • example 1 it was demonstrated that a CD3xCLEC12A bsAb can induce killing of CLEC12A-positive HL60 cells by resting T cells from healthy donors.
  • T cells were isolated from frozen peripheral blood of AML patients (AML FAB classification AML-M1/M2, M4 or M5) in clinical remission using pan T-cell isolation kit as described in example 1 1.
  • Purified AML patient derived resting T-cells were subsequently incubated with CSFE-labeled HL60 cells in medium supplemented with 10% normal HS at an effector: target cell ratio of 5: 1 for two days, in the presence of PBS, isotype control WT-Fc Ab, CD3xCLEC12A DM-Fc, CD3xisotype DM-Fc, and positive control CD 3 WT-Fc Ab (all antibodies at concentration of 1 ⁇ g/ml).
  • T cell activation was determined by flow cytometric analysis for CD3, CD4, and CD25. These results were expressed as percentage CD25+ cells per CD4+ T cells. Moreover, surviving CFSE-positive HL60 cells were quantified by flow cytometry. Results were expressed as the percentage of specific lysis relative to IgG.
  • the CD3xisotype control DM-Fc bsAb did not induce killing of HL60 cells, neither by AML patient T cells nor by healthy donor T cells.
  • the CD3xCLEC12A bispecific molecule is a potent inducer of T cell mediated tumor cell lysis, regardless of whether these T cells are AML patient derived or from healthy donors.
  • the CD3xCLEC12A bsAb has the capacity to induce potent lysis of HL60 tumor cells by AML patient T cells, subsequently the capacity of the CD3xCLEC12A bsAb to target specific activation of AML T cells was evaluated. In addition, the capacity of the CD3xCLEC12A bsAb to induce lysis of primary CLEC12A-positive AML blasts by AML patient derived autologous T cells was determined.
  • frozen stored bone marrow samples from AML patients at diagnosis samples containing >70% of primary AML blasts as determined by flow cytometric analysis were thawed, cultured overnight (O/N) in IMDM medium supplemented with 10% FCS, lOOng/ml GM-CSF, lOOng/ml G-CSF, 50ng/ml IL-3, 25ng/ml SCF and 20ng/ml Flt3L as previously described (Norde et al., 2009).
  • T cell activation was determined by flow cytometric analysis for CD3, CD4, CD8, and CD25. These results were expressed as percentage CD25+ cells per CD4+ or CD8+ AML T cells.
  • AML blast lysis was determined by quantification of the surviving CFSE7CD45 10W double positive AML blasts by flow cytometry. Results were expressed as the percentage of specific blast lysis relative to IgG.
  • CD3xCLEC12A bsAb can efficiently induce killing of CLEC12A positive tumor cells by AML patient T cells.
  • Example 14 Effect of Fc-silencing on non-specific cytokine release
  • CD3xCLEC12A bsAb IgGl format with Fc silencing by CH2/lower hinge engineering (DM-Fc) resulted in reduced affinity for Fcgamma receptors and abrogated non-specific Fc receptor mediated cytotoxicity of the leukemia-derived HL60 cell line.
  • DM-Fc CH2/lower hinge engineering
  • PBMC peripheral blood from healthy donors by density gradient centrifugation and were plated at a density of 1*10 A 6 cells/ml.
  • PBMC peripheral blood monoclonal Ab with WT-Fc.
  • isotype control Ab CD3xCLEC12A WT-Fc bsAb, CD3xCLEC12A DM-Fc bsAb, CD3xisotype control WT-Fc bsAb, CD3xisotype control DM-Fc bsAb or CD3 monoclonal Ab with WT-Fc.
  • surviving monocytes were quantified by flow cytometry based on CD14-expression. Results were expressed as the percentage of specific lysis related to IgG.
  • GM-CSF GM-CSF
  • IFN- ⁇ IL- ⁇
  • IL-2 IL-4
  • IL-5 IL-6
  • IL-8 IL-10
  • TNF-a TNF-a
  • Results shown are of cytokine concentration measured in pg/ml.
  • the levels of GM-CSF, IL-4 and IL-5 cytokines were below detection limit of this assay (data not shown).
  • CD3xCLEC12A and CD3xisotype control bsAb both with WT-Fc tail induced release of IL- ⁇ , IL-6, TNF-a, IL-10, IL-2 and IFN- ⁇ (Figue 13).
  • IL-8 no or only very low levels of those cytokines were found in CD3xCLEC12A and CD3xisotype control bsAb when carrying the DM-Fc tail, with an exception for IL-8.
  • monocytes are the main source of IL-8
  • the high IL-8 levels are assumed to be released from the lysed monocytes and are not a result from a-specific FcR mediated release.
  • Fc silencing through the DM mutations in the bsAb IgG format significantly eliminates the Fc receptor mediated release of IL-lb, IL-6, TNF-a, IL-2 and IFN- ⁇ cytokines associated with CRS.
  • Fc silencing by the DM mutation in the CH2/lower hinge region contributes to the enhancement of the efficiency and specific recruitment of effector cells by CD3xCLEC12A DM-bsAb by diminishing the potential non-specific immune activation mediated by normal Fey receptor expressing accessory cells and associated release of proinflammatory cytokines.
  • candidate 3896 as full length bivalent monoclonal anti-CD3 IgG to membrane bound CD3 was compared with candidate 3056 as full length bivalent monoclonal anti-CD3 IgG by FACS analysis using CD3 expressing HPB-ALL cells.
  • An irrelevant human IgGl served as an isotype control IgG.
  • Flow cytometry was performed according to standard procedures known in the art. As shown in Figure 14A, the 3896 CD3 IgG dose-dependently bound to CD3 on HPB-ALL cells, as did the 3056 CD3 IgG.
  • a panel of CLEC12A-specific Fab arms was selected from phage display libraries. All CLEC12A binding molecules contained the huVkl-39 light chain. Three CLEC12A binding molecules were selected: Fabs 3918, 4327 and 4331. These Fabs were expressed as full length human IgGl: 3918 CLEC12A IgG, 4327 CLEC12A IgG and 4331 CLEC12A IgG.
  • the nucleotide and amino acid sequences of the VH of 3918 CLEC12A IgG, the VH of 4327 CLEC12A IgG, the VH of 4331 CLEC12A IgG and the common VL (IGKV1-39; 012) are provided in Figure 20.
  • the full length CLEC12A antibodies were tested for binding to CLEC12A expressed by HL60 cells.
  • Fabs 3918, 4327 and 4331 are good CLEC12A binding arms.
  • Example 17 It was tested whether bispecific molecules containing the 3896 CD3 Fab arm and the CLEC12A Fab arm 3981, 4327 or 4331 were functional.
  • VH sequence of the 3896 CD3 Fab arm and the VH region of either the CLEC12A benchmark antibody, the 3918 CLEC12A Fab, the 4327 CLEC12A Fab or the 4331 CLEC12A Fab were cloned into expression vectors using methods known in the art for production of bispecific IgGl (Gunasekaran et al., WO2009/089004) in conjunction with the rearranged huVKl-39 light chain to result in bispecific antibodies; 3896xCLEC12A benchmark, 3896x3918, 3896x4327 and 3896x4331.
  • the observed differences between the 3896x4327 and 3896x4331 versus the 3896xCLEC12A benchmark bispecific IgG may reflect a difference in binding affinity of these novel anti-CLEC12A Fab arms or they might be targeting a different CLEC12A epitope that allows a more efficient crosslinking of the tumor cells with CD3 expressing T cells.
  • PCT/NL2013/050294 we have disclosed methods and means for producing bispecific antibodies from a single cell, whereby means are provided that favor the formation of bispecific antibodies over the formation of monospecific antibodies. These methods can also be favorably employed in the present invention.
  • preferred mutations to produce essentially only bispecific full length IgG molecules are the amino acid substitutions L351K and T366K (numbering according to Kabat) in the first CH3 domain (the 'KK-variant' heavy chain) and the amino acid substitutions L351D and L368E in the second CH3 domain (the 'DE- variant' heavy chain), or vice versa. It was previously demonstrated in our co-pending US 13/866,747 and PCT/NL2013/050294 applications that the DE-variant and KK- variant preferentially pair to form heterodimers (so-called 'DEKK' bispecific molecules).
  • DE-variant heavy chains DE-variant heavy chains
  • KK-variant heavy chains KKKK homodimers
  • CD3xCLEC12A bispecific molecules were used to drive heterodimerization of the different heavy chains for making CD3xCLEC12A bispecifics.
  • the CH2 / lower hinge double mutations L235G and G236R; DM
  • the Fc tail of these resulting bispecific molecules is referred to as 'DM DEKK'.
  • VH regions of either the 3116, 4327 or 4331 CLEC12A Fab arms were cloned into expression vectors containing the DE-variant + DM heavy chain whereas the VH region of the 3056 CD3 antibody was cloned into an expression vector containing the KK- variant + DM heavy chain (US regular application NO: 13/866,747 and PCT/NL2013/050294) and these expression vectors, together with a nucleic acid molecule encoding the rearranged human IGKV1-39/IGKJ1 (huVKl-39) light chain, were provided to a host cell such that the host cell expressed and produced bispecific antibodies.

Landscapes

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

Abstract

The present invention provides human IgG bispecific antibodies, wherein one arm of the antibody binds an epitope on immune effector cells, whilst the other arm targets CLEC12A or a functional equivalent thereof. Pharmaceutical uses of these antibodies against malignancies such as MDS, CML or AML are also provided.

Description

BISPECIFIC IgG ANTIBODIES AS T CELL ENGAGERS
TECHNICAL FIELD
[0001] The invention relates to the field of antibody engineering. In particular it relates to the field of therapeutic (human) antibodies for the treatment of diseases involving aberrant cells. More in particular it relates to bispecific antibodies for the treatment of tumors.
BACKGROUND OF THE INVENTION
[0002] In laboratories, bispecific antibodies have been widely used for the retargeting of immune effector cells to tumor cells. In this case, one binding site is directed against a tumor-associated antigen (TAA) and the second antigen against a trigger molecule on the effector cells, such as for example CD3 on T cells (Kontermann, MABS 2012 (4) 182-197; Chames and Baty, MABS 2009 (1) 539-547; Moore et al. Blood 2011 (117) 4542-4551). The first bispecific antibodies targeting CD3 and a tumor cell associated antigen were of rodent nature and were produced using hybrid hybridomas (Liu et al. 1985 PNAS 82: 8648, Staerz et al. 1986 PNAS 83: 1453, Lanzavecchia et al. 1987, Eur.J.Imm. 17: 105). In these hybrid hybridomas the reassortment of Ig heavy and light chains resulted in the production of bispecific functional antibody molecules within a much larger pool of monospecific and non-functional bispecific antibodies resulting from heavy and light chain mispairing. Because of their double specificity, these functional bispecific antibodies were able to bridge murine and human cytotoxic T lymphocytes (CTL) to target cells and trigger cytotoxic function resulting in the lysis of tumor cells displaying the relevant antigen. However, the CD3xTAA bispecific IgG mediated induction of tumor cell lysis by polyclonal resting human T cells could not be achieved unless co-stimulation was provided by added exogenous IL-2 or anti-CD28 mAb. This is exemplified by the hybrid rat IgG2b/mouse IgGl CD3xCD19 bispecific molecule that was able to induce lysis of the CD 19 positive REH B-ALL tumor cell line by resting human T lymphocytes only upon co-administration of IL-2 (Haagen et al. 1995 Blood 85:3208). Zeidler et al. demonstrated using a similar rat IgG2b/mouse IgG2a CD3xEpcam molecule that bispecific IgG-induced lysis of Epcam-positive tumor cells could be achieved in mixed cell cultures comprising both peripheral blood mononuclear cells (PBMC) and tumor cells without addition of exogenous IL2 (Zeidler et al. 1999 J. Immunol. 163: 1246). The authors claimed that the 'third' arm of the antibody, the Fc region, is causing this effect through interaction with Fey receptor-positive accessory cells present within the PBMC fraction. In particular, the strong activation potential was correlated to the hybrid subclass combination mouse IgG2a/rat IgG2b that, in contrast to other reported combinations (e.g., mouse IgG2a/mouse IgGl or rat IgG2b/mouse IgGl), not only binds but also activates Fey receptor-positive accessory cells. This so-called triomAb CD3xEpcam bispecific antibody, also known as catumaxomab, has been developed clinically and has been registered in Europe for palliative treatment of abdominal tumors of epithelial origin. While this bispecific antibody has clearly demonstrated clinical efficacy, its rodent nature induces anti-product immune responses upon repetitive dosing and therefore prevents a widespread application of this format.
[0003] Alternative CD3xTAA formats have been explored to solve both the manufacturing issues and the immunogenicity problems associated with the hybrid rodent triomAb format. Such formats are often immunoglobulin-like molecules that deviate from full length human IgG molecules, and include molecules such as Dual-Affinity Re-Targeting (DART™) molecules that are developed by Macrogenics worldwide web at macrogenics.com/Platforms-DART.html, Bispecific T cell Engager (BiTE®) molecules that were developed by Micromet, now Amgen (Sheridan C, Nat Biotechnol. 2012 (30):300-l), Dual Variable Domain -immunoglobulin (DVD-Ig™) molecules that are developed by Abbott, and TandAb® RECRUIT molecules that are developed by Affimed world wide web at affimed.com/tandab-recruit. It was demonstrated for one of these formats that successful retargeting of peripheral blood lymphocytes to lyse CD19-positive tumor cells using a CD3xCD19 diabody required pre-activation the of the peripheral blood T lymphocytes, now using anti-CD3 antibody plus human IL-2 (Kipriyanov et al. 1998 Int.J.Can. 77:763). Other formats, such as the bivalent single chain Fv CD3xTAA BiTE® format (Loffler et al. 2000 Blood 95:2098) do not require pre-activation of resting T cells and is able to induce antigen positive tumor cell lysis in vitro in an extremely efficient manner (Dreier et al. 2002 IntJ.Canc. 100:690). Additional studies using BiTE®s targeting different TAAs revealed that the potent efficacy of the BiTE® format was correlated to the antigen size and particularly to the distance of the epitope on the TAA to the tumor cell membrane (Bluemel et al. 2010 Cancer Immunol. Immunother. 59: 1197). The effective formation of cytolytic T cell synapses was demonstrated for BiTE® molecules which is explained to form the structural basis for their potency (Offner et al. Molecular Immunology 2006 (43) 763-771) which is also believed to be linked to the small size of the BiTE® format. If size matters, this would suggest that larger molecules such as intact IgG would be too large to form effective cytolytic synapses. The CD3xCD19 BiTE®, blinatumomab, has demonstrated remarkable clinical efficacy in refractory non-Hodgkin lymphoma and acute lymphatic leukemia patients (Bargou et al. 2008 Science 321:974). Although the CD3xCD19 BiTE® displays very efficient tumor cell lysis at low levels in vitro, administration of this bispecific format to patients is associated with significant challenges. Due to their small size, BiTE®s are rapidly cleared from the circulation and dosing of patients thus requires continuous infusion. As the dosing regimen has an overall duration of more than 2 months, this treatment has a significant impact on the quality of life of the patients.
[0004] There thus remains a need for effective full length bispecific T cell engaging
IgG molecules in eradicating aberrant cells that combine a long circulatory half-life upon intravenous administration without the need for continuous infusion without being immunogenic and with only limited side effects.
BRIEF DESCRIPTION OF THE FIGURES
[0005] FIG. 1 : CLEC 12A and related sequences.
[0006] FIG. 2: T-cell activation by various antibodies: monoclonal bivalent CD3 IgG, bispecific CD3XCLEC12 IgG, bispecific CD3xisotype control IgG, monoclonal bivalent CLEC12A IgG, monoclonal bivalent isotype control IgG.
[0007] FIG. 3: Specific lysis of HL60 cells by CD3XCLEC12A bispecific IgG and control antibodies.
[0008] FIG. 4: Specific lysis of HL60 cells by CD3XCLEC12A bispecific IgG and control antibodies (E:T ratios).
[0009] FIG. 5: Specific lysis of HL60 cells with several CD3XCLEC12A bispecific
IgG molecules consisting of various CLEC12A arms & fixed CD3 arm, and control antibodies.
[0010] FIG. 6: Specific lysis of HL60 cells with CD3XCLEC12A bispecific IgG in combination with Fc silencing (DM=Double mutant; TM= triple mutant; WT=wildtype, no Fc silencing).
[0011] FIG. 7: Fc silencing does not affect FcRn binding.
[0012] FIG. 8: CD3xCLEC 12A bsAb target specific induction of T cell proliferation. [0013] FIG. 9: CD8+ T cell compartment of AML patients compared to healthy donors.
[0014] FIG. 10: Specific CD3xCLEC12A DM-Fc induced T cell activation and HL60 tumor cell lysis by AML patient T cells.
[0015] FIG. 11 : Specific lysis of AML blasts by autologous AML patient T cells.
[0016] FIG. 12: Specific monocyte lysis by patient T cells.
[0017] FIG. 13: Fc silencing significantly eliminates bystander cell cytokine release.
[0018] FIG. 14A: FACS staining anti-CD3 antibodies on HPB-ALL cells
[0019] FIG. 14B: Plate bound IgG, T cells labeled with CFSE, read out at day 5 by FACS
[0020] FIG 15:HL60 cytotoxicity assay
[0021] FIG 16: FACS staining anti-CLEC12A antibodies on HL60 cells
[0022] FIG. 17: HL60 cytotoxicity assay
[0023] FIG 18: FACS analysis
[0024] FIG 19: HL60 cytotoxicity assay
[0025] FIG 20: VH sequences of CD3-specific and CLEC12A-specific Fab arms. VL sequence of 012 common light chain. CDR sequences are bold and underlined.
SUMMARY OF THE INVENTION
[0026] The present invention describes a fully human IgG bispecific full length antibody for the treatment of AML. One arm of the antibody binds an epitope on immune effector cells, preferably CD3, whilst the other arm targets CLEC12A, a myeloid cell specific surface target that is expressed in 90-95% of de novo and relapsed AML patients. CLEC12A is expressed on AML leukemic stem cells, but not on normal haematopoietic cells. Unlike CD33, CLEC12A is not expressed on erythroid precursors or megakaryocytes, so the CD3xCLEC12A bispecific IgGl antibody of the present invention should not induce platelet or red blood cell depletion.Expenments with bone marrow cell colonies have shown that depletion of CLEC12A+ cells in normal bone marrow does not affect the myeloid lineages that give rise to platelets and red blood cells. A CD3xCLEC12A bispecific IgG antibody according to the present invention in a preferred embodiment contains a modified Fc region so as to reduce non-specific immune activation resulting from engagement of T cells and FcyR expressing cells within PBMC. Based on data described for the triomAb bispecific antibody in the prior art it was highly doubted that a CD3xTAA bispecific IgG of a fully human IgGl format would be able to induce lytic anti-tumor activity in resting peripheral blood lymphocytes without the need for pre- activation of T cells. In addition, the available data for the BiTE® format suggested that a full length IgG molecule would be too large to create effective cytolytic synapses between tumor cells and effector cells. Surprisingly, we demonstrated that a fully human CD3xCLEC12A bispecific full length IgGl was able to induce very efficient T cell mediated lysis of CLEC12A-positive HL60 AML tumor cells in vitro. In fact, effective lysis was mediated by resting T lymphocytes purified from PBMC without the need of prior activation of the T cells. Furthermore, we demonstrated that this lytic activity is not necessarily dependent on interactions with FcyR present on HL60 cells as this lytic activity was not affected by the presence of excess human IgG when the assay was performed in human serum containing media. This is the first time that a full length human IgGl bispecific T cell engager antibody exerts efficient tumor cell lysis without the need of pre-activation of T cells or the need of active FcyR interactions. Effective lysis is achieved despite the relatively large size of the IgGl when compared to BiTE® molecules. Remarkably, when CH2/lower hinge mutations were introduced in the CD3xCLEC12A bispecific IgGl molecule to further decrease Fc receptor interactions, this still resulted in efficient tumor cell lysis by immune effector cells. A bispecific human IgGl T cell engager antibody has advantages over current IgG that make use of the hybrid subclass combination mouse IgG2a/rat IgG2b, since a human IgGl will be less immunogenic and can thus be applied for repeated therapy. In addition, a full length bispecific human IgGl T cell engager antibody has advantages over immunoglobulin/like molecules such as DART™, TandAb® or BiTE® as the full length human IgGl is not rapidly cleared from the circulation and dosing of patients will thus not require continuous infusion, which is more beneficial to patients.
EMBODIMENTS
[0027] The invention provides a bispecific IgG antibody, wherein said bispecific IgG antibody comprises one arm that specifically recognizes CLEC12A or a functional equivalent thereof, and a second arm that specifically recognizes an antigen on immune effector cells capable of recruiting such cells to an aberrant cell expressing CLEC12A or said functional equivalent. [0028] As used herein, the term "specifically recognizes CLEC12A or a functional equivalent thereof means that said arm has the capability of specifically recognizing CLEC12A or said functional equivalent, in the situation that CLEC12A or said functional equivalent is present in the vicinity of said antibody. Likewise, the term "specifically recognizes an antigen on immune effector cells" means that said arm has the capability of specifically recognizing said antigen when said antigen is present in the vicinity of said antibody. Such antigen recognition by an antibody is typically mediated through the complementarity regions of the antibody and the specific three-dimensional structure of both the antigen and the antibody arm allowing these two structures to bind together with precision (an interaction similar to a lock and key), as opposed to random, non-specific sticking of antibodies. As an antibody typically recognizes an epitope of an antigen, and as such epitope may be present in other compounds as well, antibodies according to the present invention that "specifically recognize CLEC12A or a functional equivalent thereof, and "specifically recognize an antigen on immune effector cells" may recognize other compounds as well, if such other compounds contain the same kind of epitope. Hence, the terms "specifically recognizes CLEC12A or a functional equivalent thereof, "specifically recognizes an antigen on immune effector cells" and "specifically recognizes CD3" do not exclude binding of the antibodies to other compounds that contain the same (kind of) epitope. Instead, cross- reactivity is allowed. An antibody according to the present invention is typically capable of binding CLEC12A (or a functional equivalent thereof) and an antigen on immune effector cells, preferably CD3, with a binding affinity of at least 1x10-5 M, as outlined in more detail below.
[0029] The term "antibody" as used herein means a proteinaceous molecule belonging to the immunoglobulin class of proteins, containing one or more domains that bind an epitope on an antigen, where such domains are derived from or share sequence homology with the variable region of an antibody. Antibodies for therapeutic use are preferably as close to natural antibodies of the subject to be treated as possible (for instance human antibodies for human subjects). Antibody binding can be expressed in terms of specificity and affinity. The specificity determines which antigen or epitope thereof is specifically bound by the binding domain. The affinity is a measure for the strength of binding to a particular antigen or epitope. Specific binding, or "specifically recognizing" is defined as binding with affinities (KD) of at least 1x10-5 M, more preferably 1x10-7 M, more preferably higher than Ixl0-9M. Typically, antibodies for therapeutic applications have affinities of up to 1x10-10 M or even higher. Antibodies of the present invention are typically bispecific full length antibodies of the human IgG subclass. Preferably, the antibodies of the present invention are of the human IgGl subclass.
[0030] The term 'full length IgG' according to the invention is defined as comprising an essentially complete IgG, which however does not necessarily have all functions of an intact IgG. For the avoidance of doubt, a full length IgG contains two heavy and two light chains. Each chain contains constant (C) and variable (V) regions, which can be broken down into domains designated CHI, CH2, CH3, VH, and CL, VL. An IgG antibody binds to antigen via the variable region domains contained in the Fab portion, and after binding can interact with molecules and cells of the immune system through the constant domains, mostly through the Fc portion. The terms 'variable region domain', 'variable region', 'variable domain', 'VH/VL pair', 'VH/VL', 'Fab portion', 'Fab arm', 'Fab' or 'arm' are used herein interchangeably. Full length antibodies according to the invention encompass IgG molecules wherein mutations may be present that provide desired characteristics. Such mutations should not be deletions of substantial portions of any of the regions. However, IgG molecules wherein one or several amino acid residues are deleted, without essentially altering the binding characteristics of the resulting IgG molecule, are embraced within the term "full length IgG". For instance, such IgG molecules can have one or more deletions of between 1 and 10 amino acid residues, preferably in non-CDR regions, wherein the deleted amino acids are not essential for the binding specificity of the IgG.
[0031] Full length IgG antibodies are preferred because of their favourable half life and the need to stay as close to fully autologous (human) molecules for reasons of immunogenicity. According to the invention, bispecific IgG antibodies are used. In a preferred embodiment, bispecific full length IgGl antibodies are used. IgGl is favoured based on its long circulatory half life in man. In order to prevent any immunogenicity in humans it is preferred that the bispecific IgG antibody according to the invention is a human IgGl. The term 'bispecific' (bs) means that one arm of the antibody binds to a first antigen whereas the second arm binds to a second antigen, wherein said first and second antigens are not identical. According to the present invention, said first and second antigens are in fact two different molecules that are located on two different cell types. The term 'one arm [of the antibody]' preferably means one Fab portion of the full length IgG antibody. Bispecific antibodies that mediate cytotoxicity by recruiting and activating endogenous immune cells are an emerging class of next-generation antibody therapeutics. This can be achieved by combining antigen binding specificities for target cells (i.e., tumor cells) and effector cells (i.e., T cells, NK cells, and macrophages) in one molecule (Cui et al. JBC 2012 (287) 28206-28214; Kontermann, MABS 2012 (4) 182-197; Chames and Baty, MABS 2009 (1) 539-547; Moore et al. Blood 2011 (117) 4542-4551 ; Loffler et al. 2000 Blood 95:2098; Zeidler et al. 1999 J. Immunol. 163: 1246). According to the invention, bispecific antibodies are provided wherein one arm binds the CLEC12A antigen on aberrant (tumor) cells whereas the second arm binds an antigen on immune effector cells.
[0032] Also contemplated are antibodies wherein a VH is capable of specifically recognizing a first antigen and the VL, paired with the VH in a immunoglobulin variable region, is capable of specifically recognizing a second antigen. The resulting VH/VL pair will bind either antigen 1 or antigen 2. Such so called "two-in-one antibodies", described in for instance WO 2008/027236, WO 2010/108127 and Schaefer et al (Cancer Cell 20, 472-486, October 2011), are also encompassed by the term "bispecific antibody" because they also have the capability of binding two different antigens. In one embodiment, a VH is used that specifically recognizes CLEC12A, or a functional equivalent thereof, and a VL is used that specifically recognizes an antigen on immune effector cells. Alternatively, an antibody according to the present invention comprises a VH that specifically recognizes an antigen on immune effector cells, and a VL that specifically recognizes CLEC12A, or a functional equivalent thereof. Either way, the resulting antibody typically contains two VH/VL pairs, wherein each VH/VL pair will bind either CLEC12A (or a functional equivalent thereof), or an antigen on immune effector cells. Two-in-one antibodies will typically either bind two similar antigens (AA or BB; monospecific bivalent) or bind two different antigens (AB; bispecific). Hence, if two-in-one antibodies are used for therapeutic applications according to the present invention, a portion of these antibodies will not exert the desired effect due to their binding to either two CLEC12A molecules (or functional equivalents thereof) or two antigens on immune effector cells, such as CD3. Since the therapeutic goal can still be achieved with a portion of the administered antibodies, two-in-one antibodies are nevertheless suitable.
[0033] The term 'CLEC12A' as used herein refers to C-type lectin domain family 12 member A, also known as C-type lectin-like molecule- 1 (CLL-1), an antigen that is expressed on leukemic blast cells and on leukemic stem cells in acute myeloid leukemia (AML), including the CD34 negative or CD34 low expressing leukemic stem cells (side population) (A.B. Bakker et al. Cancer Res 2004, 64, p8443-50; Van Rhenen et al. 2007 Blood 110:2659; Moshaver et al. 2008 Stem Cells 26:3059). Expression of CLEC12A is otherwise restricted to the hematopoietic lineage, particularly to myeloid cells in peripheral blood and bone marrow, i.e., granulocytes, monocytes and dendritic cell precursors. More importantly, CLEC12A is absent on hematopoietic stem cells. This expression profile makes CLEC12A a particularly favorable target in AML. Alternative names for CLEC12A include dendritic cell-associated C-type lectin-2 (DCAL-2), myeloid inhibitory C-type lectin-like receptor (MICL) and killer cell lectin-like receptor subfamily L, member 1 (KLRL1) (Zhang W. et al. GenBankTM access.no: AF247788; A.S. Marshall, et al. J Biol Chem 2004, 279, pl4792-802; GenBankTM access.no: AY498550; Y.Han et al. Blood 2004, 104, p2858-66; H.Floyd, et al. GenBankTM access.no: AY426759; C.H.Chen, et al. Blood 2006, 107, p 1459-67). An alignment of these sequences is represented in FIG. 1. The full length form of CLEC12A comprises 275 amino acid residues, including an additional intracellular stretch of 10 amino acids which is absent in most other isoforms, and shows the strictly myeloid expression profile (surface expression and mRNA level). The term 'CLEC12A or functional equivalent thereof means all variants that are referenced above and isoforms thereof that retain the strict myeloid expression profile (both at surface expression level and mRNA level) as described in Bakker et al. Cancer Res 2004, 64, p8443-50. Hence, the invention includes bispecific IgG antibodies wherein one arm specifically recognizes functional equivalents of CLEC12A, including those functional equivalents that lack the above mentioned additional intracellular stretch of 10 amino acids. Bispecific IgG antibodies according to the invention wherein one arm specifically recognizes the full length form of CLEC12A are, however, preferred.
[0034] The term 'aberrant cells' as used herein includes tumor cells, more specifically tumor cells of hematological origin including also pre-leukemic cells such as cells that cause myelodysplastic syndromes (MDS) and leukemic cells such as acute myeloid leukemia (AML) tumor cells or chronic myelogenous leukemia (CML) cells.
[0035] The term 'immune effector cell' or 'effector cell' as used herein refers to a cell within the natural repertoire of cells in the mammalian immune system which can be activated to affect the viability of a target cell. Immune effector cells include cells of the lymphoid lineage such as natural killer (NK) cells, T cells including cytotoxic T cells, or B cells, but also cells of the myeloid lineage can be regarded as immune effector cells, such as monocytes or macrophages, dendritic cells and neutrophilic granulocytes. Hence, said effector cell is preferably an NK cell, a T cell, a B cell, a monocyte, a macrophage, a dendritic cell or a neutrophilic granulocyte. According to the invention, recruitment of effector cells to aberrant cells means that immune effector cells are brought in close vicinity to the aberrant target cells cells such that the effector cells can directly kill, or indirectly initiate the killing of the aberrant cells that they are recruited to. In order to avoid non specific interactions it is preferred that the bispecific antibodies of the invention specifically recognize antigens on immune effector cells that are at least over-expressed by these immune effector cells compared to other cells in the body. Target antigens present on immune effector cells may include CD3, CD 16, CD25, CD28, CD64, CD89, NKG2D and NKp46, Preferably, the antigen on immune effector cells is CD3 expressed on T cells, or a functional equivalent thereof (a functional equivalent would be a CD3-like molecule with a similar distribution on T-cells and a similar function (in kind, not necessarily in amount)). As used herein, the term "CD3" also encompasses functional equivalents of CD3. The most preferred antigen on an immune effector cell is the CD3s chain. This antigen has been shown to be very effective in recruiting T cells to aberrant cells. Hence, a bispecific IgG antibody according to the present invention preferably contains one arm that specifically recognizes CD3s.
[0036] Thus, the invention provides a bispecific full length IgG antibody, wherein said bispecific antibody comprises one arm that specifically recognizes CLEC12A or a functional equivalent thereof, and a second arm that specifically recognizes an antigen on immune effector cells capable of recruiting such cells to an aberrant cell expressing CLEC12A or said functional equivalent, wherein said immune effector cells comprise T cells. In another preferred embodiment, the invention provides a bispecific IgG antibody according to the invention wherein said antigen on said immune effector cells is CD3 or functional equivalent thereof, preferably human CD3s. In another embodiment, the invention provides F(ab)'2 fragments of such bispecific IgG CLEC 12AxCD3 antibody.
[0037] It is an aspect of the invention to provide a bispecific IgG antibody according to the invention wherein both arms comprise a common light chain. The term 'common light chain' according to the invention refers to light chains which may be identical or have some amino acid sequence differences while retaining the binding specificity of the antibody. It is for instance possible within the scope of the definition of common light chains as used herein, to prepare or find light chains that are not identical but still functionally equivalent, e.g., by introducing and testing conservative amino acid changes, changes of amino acids in regions that do not or only partly contribute to binding specificity when paired with the heavy chain, and the like. The terms 'common light chain', 'common VL', 'single light chain', 'single VL', with or without the addition of the term 'rearranged' are all used herein interchangeably. It is an aspect of the present invention to use as common light chain a human light chain that can combine with different heavy chains to form antibodies with functional antigen binding domains (WO2004/009618, WO2009/ 157771, Merchant et al. 1998, Nissim et al. 1994). Preferably, the common light chain has a germline sequence. A preferred germline sequence is a light chain variable region that is frequently used in the human repertoire and has superior ability to pair with many different VH regions, and has good thermodynamic stability, yield and solubility. A most preferred germline light chain is 012, preferably the rearranged germline human kappa light chain IgVKl-39*01/IGJKl*01 (nomenclature according to the IMGT database worldwide web at imgt.org or fragment or a functional derivative thereof. The terms rearranged germline human kappa light chain IgVKl-39*01/IGJKl*01, IGKV1-39/IGKJ1, huVKl-39 light chain or in short huVKl-39 are used interchangeably throughout the application. Obviously, those of skill in the art will recognize that "common" also refers to functional equivalents of the light chain of which the amino acid sequence is not identical. Many variants of said light chain exist wherein mutations (deletions, substitutions, additions) are present that do not materially influence the formation of functional binding regions.
[0038] In a particularly preferred embodiment a bispecific IgG antibody according to the invention is provided wherein the arm that specifically recognizes CLEC12A or a functional equivalent thereof comprises a heavy chain CDR1 sequence consisting of a sequence that is at least 90% identical to SGYTFTSY and a heavy chain CDR2 sequence consisting of a sequence that is at least 90% identical to IINPSGGS and a heavy chain CDR3 sequence consisting of a sequence that is at least 90% identical to GTTGDWFD. Preferably, said heavy chain CDR 1, 2 and 3 sequences consist of a sequence that is at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99% identical to the recited CDR sequences. Typically, variations of 1, 2 or 3 amino acid residues from the recited CDR sequences are allowed while retaining the same kind of binding activity (in kind, not necessarily in amount). Hence, said heavy chain CDR 1 , 2 and 3 sequences preferably contain sequences that deviate in no more than three, preferably no more than two, more preferably no more than one amino acid from the recited CDR sequences. In a particularly preferred embodiment, said heavy chain CDR 1, 2 and 3 sequences are identical to the recited CDR sequences. The recited CDR sequences are the CDR sequences of Fab arm 4327 which, as shown in the Examples, has good CLEC12A binding properties. The heavy chain sequence of Fab arm 4327, hence the VH of CLEC12A- specific antibody 4327, is shown in Figure 20. In one preferred embodiment, a bispecific IgG antibody according to the invention comprises a variable heavy chain (VH) sequence that is at least 90% identical to this VH of antibody 4327. Further provided is therefore a bispecific IgG antibody according to the invention, wherein the arm that specifically recognizes CLEC12A or a functional equivalent thereof comprises a VH sequence consisting of a sequence that is at least 90%, preferably at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99% or even 100%, identical to the sequence QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLEWMGIINPSGGS TSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCAKGTTGDWFDYWGQGTLV TVS. As shown in the Examples, bispecific antibodies according to the invention containing the above mentioned VH sequence, together with a VH sequence of a Fab arm recognizing CD3 (and together with a common light chain), have an excellent capacity of inducing T cell mediated lysis of CLEC12A-positive AML tumor cells.
[0039] In a further preferred embodiment a bispecific IgG antibody according to the invention is provided wherein the arm that specifically recognizes CLEC12A or a functional equivalent thereof comprises a heavy chain CDR1 sequence consisting of a sequence that is at least 90% identical to SGYTFTSY and a heavy chain CDR2 sequence consisting of a sequence that is at least 90% identical to IINPSGGS and a heavy chain CDR3 sequence consisting of a sequence that is at least 90% identical to GNYGDEFDY. Preferably, said heavy chain CDR 1 , 2 and 3 sequences consist of a sequence that is at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99% identical to the recited CDR sequences. As stated above, typically variations of 1, 2 or 3 amino acid residues from the recited CDR sequences are allowed while retaining the same kind of binding activity (in kind, not necessarily in amount). Hence, said heavy chain CDR 1, 2 and 3 sequences preferably contain sequences that deviate in no more than three, preferably no more than two, more preferably no more than one amino acid from the recited CDR sequences. In a particularly preferred embodiment, said heavy chain CDR 1, 2 and 3 sequences are identical to the recited CDR sequences. The recited CDR sequences are the CDR sequences of the VH region of antibody 4331 which, as shown in the Examples, has good CLEC12A binding properties. The VH sequence of Fab arm 4331 is also shown in Figure 20. In one preferred embodiment, a bispecific IgG antibody according to the invention comprises a VH sequence that is at least 90% identical to this VH of Fab arm 4331. Further provided is therefore a bispecific IgG antibody according to the invention, wherein the arm that specifically recognizes CLEC12A or a functional equivalent thereof comprises a VH sequence consisting of a sequence that is at least 90%, preferably at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99% or even 100%, identical to the sequence EVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLEWMGIINPSGGS TSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCARGNYGDEFDYWGQGTLV TVSS. As shown in the Examples, bispecific antibodies according to the invention containing the VH sequence of Fab arm 4331, together with a VH sequence of a Fab arm recognizing CD3, (together with a common light chain) have an excellent capacity of inducing T cell mediated lysis of CLEC12A-positive AML tumor cells.
[0040] Yet another preferred embodiment provides a bispecific IgG antibody according to the invention wherein the arm that specifically recognizes CLEC12A or a functional equivalent thereof comprises a heavy chain CDR1 sequence consisting of a sequence that is at least 90% identical to SGYTFTGY and a heavy chain CDR2 sequence consisting of a sequence that is at least 90% identical to WINPNSGG and a heavy chain CDR3 sequence consisting of a sequence that is at least 90% identical to DGYFADAFDY. Preferably, said heavy chain CDR 1, 2 and 3 sequences consist of a sequence that is at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99% identical to the recited CDR sequences. Again, typically variations of 1, 2 or 3 amino acid residues from the recited CDR sequences are allowed while retaining the same kind of binding activity (in kind, not necessarily in amount). Hence, said heavy chain CDR 1 , 2 and 3 sequences preferably contain sequences that deviate in no more than three, preferably no more than two, more preferably no more than one amino acid from the recited CDR sequences. In a particularly preferred embodiment, said heavy chain CDR 1, 2 and 3 sequences are identical to the recited CDR sequences. The recited CDR sequences are the CDR sequences of the VH of antibody 3918 which, as shown in the Examples, also has good CLEC12A binding properties. The VH sequence of the antibody 3918 is also shown in Figure 20. In one preferred embodiment, a bispecific IgG antibody according to the invention comprises a VH sequence that is at least 90% identical to this VH of antibody 3918. Further provided is therefore a bispecific IgG antibody according to the invention, wherein the arm that specifically recognizes CLEC12A or a functional equivalent thereof comprises a VH sequence consisting of a sequence that is at least 90%, preferably at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99% or even 100%, identical to the sequence QVQLVQSGAEVKKPGASVKVSCKASGYTFTGYYMHWVRQAPGQGLEWMGWINPNSG GTNYAQKFQGRVTMTRDTSISTAYMELSRLRSDDTAVYYCARDGYFADAFDYWGQGT LVTVSS. As shown in the Examples, bispecific antibodies according to the invention containing the VH sequence of Fab arm 3918, together with the VH sequence of a Fab arm recognizing CD3, (and together with a common light chain) also have a good capacity of inducing T cell mediated lysis of CLEC12A-positive AML tumor cells.
[0041] In a further preferred embodiment a bispecific IgG antibody according to the invention is provided wherein the second arm specifically recognizes CD3 and comprises a heavy chain CDR1 sequence consisting of the sequence SYGMH and a heavy chain CDR2 sequence consisting of the sequence IIWYSGSKKNYADSVKG and a heavy chain CDR3 sequence consisting of the sequence GTGYNWFDP. Preferably, said CD3-specific arm comprises a VH sequence consisting of the sequence
QVQLVESGGGVVQPGRSLRLSCAASGFTFRSYGMHWVRQAPGKGLEWVAIIWYSGSK KNYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARGTGYNWFDPWGQGTLV TVSS. The recited CDR sequences and VH sequence are the sequences of antibody 3896. These sequences are also depicted in Figure 20. A heavy chain comprising these CD3-specific CDR sequences and/or the recited VH sequence of Fab arm 3896 is preferred for a bispecific IgG antibody according to the invention, because these sequences provide the bispecific antibody with an optimal affinity for CD3-expressing immune cells, while simultaneously allowing sufficient binding to CLEC12A-positive AML tumor cells. Without wishing to be bound to theory, it is thought that the overall effect of a bispecific antibody is determined by the combined effect of the affinity of one arm for antigen 1 and the affinity of the other arm for antigen 2. For an antibody of the present invention, having a specificity for CLEC12A (or a functional equivalent thereof) and an antigen on immune effector cells (preferably CD3), an optimized timing of binding to CD3-positive immune cells and CLEC12A-expressing tumor cells is preferred in order to efficiently induce T cell mediated lysis of CLEC12A-positive tumor cells. It is hypothesized that the balance between affinities of a CLEC12A/CD3 bispecific antibody is of utmost importance. It is thought that a significantly higher affinity for CD3 versus a much lower affinity for CLEC12A (i.e., a too high affinity for CD3) will result in a situation wherein the antibodies would primarily bind CD3 expressing T cells. Such 'bispecific antibody-loaded' T- cells may either internalize their CD3 or may invade the tissues thereby leaving the circulation before they have even encountered a CLEC12A-positve tumor cell. This would diminish the therapeutic effect of the bispecific antibody.
[0042] In a more favorable mode of action, CLEC12A -positive tumor cells are first bound by one or more bispecific antibodies according to the invention, where after T cells are attracted by the free CD3 arm of the bispecific antibody and subsequent T cell activation takes place. Alternatively, CD3 positive T cells and CLEC12A-positve tumor cells are bound essentially simultaneously by the bispecific antibody. Hence, the affinities for both CLEC12A (or a functional equivalent thereof) and for an antigen on immune effector cells (preferably CD3) are preferably chosen or modulated such that the right balance is achieved, i.e. that the resulting bispecific antibodies will either bind CLEC12A and CD3 essentially simultaneously or that the bispecific antibodies have a tendency to bind CLEC12A-positive tumor cells to a sufficient extent, where after T cell activation takes place and the tumor cells are lysed. According to the present invention, such excellent balance between the binding affinities for CD3 and CLEC12A is preferably achieved by combining a VH having the CDR sequences (or whole VH sequence) of Fab arm 3896 (which is specific for CD3) with a VH having the CDR sequences (or whole VH sequence) of either Fab arms 4327 or 4331 or 3918 or 3116 (which are specific for CLEC12A). Such resulting bispecific antibodies display a favorable balance between the binding affinities for CD3 and CLEC12A, so that T cells and CLEC12A-positive AML tumor cells are efficiently brought together, and T cell mediated lysis of CLEC12A-positive AML tumor cells is optimally induced.
[0043] As described herein before, a bispecific IgG antibody according to the present invention is preferably provided wherein both arms comprise a common light chain variable domain. A particularly preferred common light chain is the human rearranged kappa light chain IgVKl-39*01/IgJKl*01, also named 012. The nucleotide and amino acid sequence of the 012 VL are also depicted in Figure 20. The CDR sequences are bold and underlined. A bispecific antibody according to the invention containing a common light chain that at least comprises the CDR sequences of 012 is therefore preferred. One aspect of the invention therefore provides a bispecific IgG antibody according to the invention, wherein the first and the second arms further comprise a light chain CDR1 sequence consisting of a sequence that is at least 90% identical to RASQSISSYLN and a light chain CDR2 sequence consisting of a sequence that is at least 90% identical to AASSLQS and a light chain CDR3 sequence consisting of a sequence that is at least 90% identical to QQS YSTPPT. Preferably, said light chain CDR 1 , 2 and 3 sequences consist of a sequence that is at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99% identical to the recited CDR sequences. Again, typically variations of 1, 2 or 3 amino acid residues from the recited CDR sequences are allowed. Hence, said light chain CDR 1, 2 and 3 sequences preferably contain sequences that deviate in no more than three, preferably no more than two, more preferably no more than one amino acid from the recited CDR sequences. In a particularly preferred embodiment, said light chain CDR 1, 2 and 3 sequences are identical to the recited CDR sequences. In one preferred embodiment, a bispecific IgG antibody according to the invention comprises a VL sequence that is at least 90% identical to the 012 VL chain. Further provided is therefore a bispecific IgG antibody according to the invention, wherein first and the second arms comprise a VL sequence consisting of a sequence that is at least 90%, preferably at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99% or even 100%, identical to the sequence DIQMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQKPGKAPKLLIYAASSLQSGVPSR FSGSGSGTDFTLTISSLQPEDFATYYCQQSYSTPPTFGQGTKVEIK.
[0044] The term "% identical to" is defined herein as the percentage of residues in a candidate amino acid sequence that is identical with the residues in a reference sequence after aligning the two sequences and introducing gaps, if necessary, to achieve the maximum percent identity. Methods and computer programs for the alignment are well known in the art. One computer program which may be used or adapted for purposes of determining whether a candidate sequence falls within this definition is "Align 2", authored by Genentech, Inc., which was filed with user documentation in the United States Copyright Office, Washington, D.C. 20559, on Dec. 10, 1991.
[0045] A bispecific full length IgG antibody according to the invention by definition has two different antigen binding sites but the Fc region of the IgG also comprises a third binding site for an Fc receptor. If a cell carries both an Fc receptor and one of the targets of the bispecific antibody, cross-linking of the Fc receptor and said target on the surface of said cell may occur, which may lead to undesired effects. In a preferred embodiment the invention provides a bispecific full length IgG antibody according to the invention, wherein said bispecific IgG antibody has mutated lower hinge and/or CH2 domains such that interaction of said bispecific IgG antibody with Fc gamma (Fey) receptors is significantly reduced. As used herein, the term "such that interaction of said bispecific IgG antibody with Fc gamma receptors is significantly reduced" means that the capability of said bispecific IgG antibody of interacting with Fc gamma receptors, if such Fc gamma receptors are present in the vicinity of said antibody, is reduced. Thus, according to the invention a region of the antibody, preferably the lower hinge and/or the CH2 domain of the antibody is mutated (typically by expressing a mutated nucleic acid sequence encoding it) whereby the ability to interact with an Fc receptor is diminished. It is preferred that the interaction with the Fc receptor is essentially abolished. Amino acid residues in human IgGl that are involved in binding to Fey receptors have been mapped previously. In addition to residues which, when altered, improved binding only to specific receptors or simultaneously improved binding to one type of receptor and reduced binding to another type, several residues were found that abrogated binding to one or more of the receptors (Shields RL et al. JBC 2001 (276) 6591-6604; Armour et al. Mol. Immunol. 2003 (40) 585-593). In a further preferred embodiment, said mutated lower hinge and/or CH2 domains comprise at least one substitution at amino acids positions 235 and/or 236 (numbering according to Kabat). Preferably, both amino acids positions 235 and 236 are substituted. It is shown in the examples that substitutions at these sites are capable of essentially preventing the interaction between the bispecific antibody and the Fc receptor present on the tumor cells. In particular it is shown that substitutions L235G and/or G236R are very suitable for that purpose. A bispecific IgG antibody according to the invention, wherein said mutated CH2 and/or lower hinge domains comprise substitution L235G and/or G236R, is therefore also provided herein. Preferably, both L235G and G236R are substituted. Alternatively, a person skilled in the art may introduce lower hinge and/or the CH2 domain mutations that comprise the substitutions 234F, 235E and/or 33 IS (Oganesyan et al. Biol. Crystall. 2008(D64)700). Preferably, all three substitutions are introduced in this alternative. [0046] In our US provisional application 61/635,935, which has been followed up by
US regular application No. 13/866,747 and PCT application No. PCT/NL2013/050294, (incorporated herein by reference), we disclose methods and means for producing bispecific antibodies from a single cell, whereby means are provided that favor the formation of bispecific antibodies over the formation of monospecific antibodies. These methods can also be favorably employed in the present invention. Thus the invention provides a method for producing a bispecific full length IgG antibody according to the invention from a single cell, wherein said bispecific full length IgG antibody comprises two CH3 domains that are capable of forming an interface, said method comprising providing in said cell a) a first nucleic acid molecule encoding a 1st CH3 domain-comprising polypeptide chain, b) a second nucleic acid molecule encoding a 2nd CH3 domain-comprising polypeptide chain, wherein said nucleic acid molecules are provided with means for preferential pairing of said 1st and 2nd CH3 domain-comprising polypeptides, said method further comprising the step of culturing said host cell and allowing for expression of said two nucleic acid molecules and harvesting said bispecific full length IgG antibody from the culture. Said first and second nucleic acid molecules may be part of the same vector or gene delivery vehicle and may be integrated at the same site of the host cell's genome. Alternatively, said first and second nucleic acid molecules are separately provided to said cell.
[0047] A preferred embodiment provides a method for producing a full length bispecific IgG antibody according to the invention from a single cell, wherein said bispecific IgG antibody comprises two CH3 domains that are capable of forming an interface, said method comprising providing: - a cell having a) a first nucleic acid sequence encoding a IgG heavy chain that specifically recognizes CLEC12A and that contains a 1st CH3 domain, and b) a second nucleic acid sequence encoding a IgG heavy chain that specifically recognizes an antigen on immune effector cells, preferably CD3, and that contains a 2nd CH3 domain, wherein said nucleic acid sequences are provided with means for preferential pairing of said 1st and 2nd CH3 domains, said method further comprising the step of culturing said cell and allowing for expression of said two nucleic acid sequences and harvesting said bispecific IgG antibody from the culture. In a particularly preferred embodiment, said cell also has a third nucleic acid sequence encoding a common light chain. A preferred common light chain is 012, preferably the rearranged germline human kappa light chain IgVKl-39*01/IGJKl*01, as described above. The preferred mutations to produce essentially only bispecific full length IgG molecules are the amino acid substitutions L351K and T366K (numbering according to Kabat) in the first CH3 domain and the amino acid substitutions L351D and L368E in the second CH3 domain, or vice versa. Further provided is therefore a method according to the invention for producing a bispecific IgGl antibody, wherein said first CH3 domain comprises the amino acid substitutions L351K and T366K (numbering according to Kabat) and wherein said second CH3 domain comprises the amino acid substitutions L351D and L368E, said method further comprising the step of culturing said cell and allowing for expression of said nucleic acid sequences and harvesting said bispecific antibody from the culture. Also provided is a method according to the invention for producing a bispecific IgGl antibody, wherein said first CH3 domain comprises the amino acid substitutions L351D and L368E (numbering according to Kabat) and wherein said second CH3 domain comprises the amino acid substitutions L351K and T366K, said method further comprising the step of culturing said cell and allowing for expression of said nucleic acid sequences and harvesting said bispecific antibody from the culture. Antibodies that can be produced by these methods are also part of the present invention.
[0048] The invention further provides a pharmaceutical composition comprising a bispecific IgG antibody according to the invention and a pharmaceutically acceptable carrier. As used herein, such 'pharmaceutically acceptable carrier' includes any and all solvents, salts, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Depending on the route of administration (e.g., intravenously, subcutaneously, intra-articularly and the like) the active compound may be coated in a material to protect the compound from the action of acids and other natural conditions that may inactivate the compound.
[0049] The antibodies and pharmaceutical compositions according to the invention find their use in the treatment of various leukemias and pre-leukemic diseases of myeloid origin but also B cell lymphomas. Diseases that can be treated according to the invention include myeloid leukemias or pre-leukemic diseases such as AML, MDS and CML and Hodgkin's lymphomas and most non-Hodgkin's lymphomas. Thus the invention provides a bispecific full length IgG antibody according to the invention for use as a pharmaceutical in the treatment of myelodysplastic syndrome (MDS), chronic myelogenous leukemia (CML) or preferably acute myeloid leukemia (AML). Also provided is a use of a bispecific IgG antibody according to the invention in the preparation of a medicament for the treatment or prevention of myelodysplastic syndrome (MDS), chronic myelogenous leukemia (CML) or preferably acute myeloid leukemia (AML).
[0050] The amount of antibody according to the invention to be administered to a patient is typically in the therapeutic window, meaning that a sufficient quantity is used for obtaining a therapeutic effect, while the amount does not exceed a threshold value leading to an unacceptable extent of side-effects. The lower the amount of antibody needed for obtaining a desired therapeutic effect, the larger the therapeutic window will typically be. An antibody according to the invention exerting sufficient therapeutic effects at low dosage is, therefore, preferred.
[0051] Approximately 30.000 patients are diagnosed each year with acute myeloid leukemia (AML) in Europe and US. The majority of these patients are 60 years of age or older. Older age is a major negative determinant of outcome in AML and long-term survival (at 5 years) of intensively treated older AML patients is approximately 10%. In almost all patients that have achieved remission upon induction chemotherapy, disease progression is observed within 3 years. Current post-remission treatment has shown limited, if any, value in older patients with AML. Therefore, a significant load of residual resistant leukemia remains, and the surviving subpopulation of drug-resistant leukemic cells rapidly generates recurrence. Novel types of drugs with entirely different modes of action are needed to target these chemotherapy non-responsive AML tumour cells in efforts to induce and sustain complete remissions. Although complete remission (CR) can be achieved with a number of intensive chemotherapy combinations in more than 50% of elderly AML patients and around 80% in younger patients, advancements of response or survival have remained a major investigational challenge. In a recently published network meta-analysis of 65 randomized clinical trials (15.110 patients) in older patients with AML most of the amended investigational induction regimens have similar or even worse efficacy profiles as compared to the conventional 3+7 induction regimen with daunorubicin and cytarabine. This standard treatment of AML is associated with high morbidity and even mortality. The majority of the patients in CR relapse due to remaining leukemic stem cells after chemotherapy. Further dose intensification is limited due to unacceptable toxicity. An urgent need for new treatment modalities preferably with less toxicity is thus emerging especially in elderly patients with AML. [0052] Treatment of chemotherapy unresponsive AML could be achieved by engaging
T cells from the patient's own immune system and AML tumour cells using a bispecific antibody. In this manner, the patients' immune system is strengthened and retargeted to attack and eradicate the AML tumour cells. The present invention provides CD3xCLEC12A bispecific IgG antibodies that efficiently induce AML tumour cell lysis. CD3xCLEC12A bispecific antibodies thus are a targeted therapy with fewer side effects that specifically eradicates leukemic stem cells in order to improve the prognosis of AML patients. Because CLEC12A is expressed on leukemic stem cells (LSC) and not on normal haematopoietic stem cells, therapy directed against this antigen (as has been shown in vitro) will eradicate the LSC while sparing the normal stem cell. It most probably will have the greatest impact in situations of Minimal Residual Disease (MRD). The expectancy is that relapse percentage will drop due to the eradication of the MRD. So the impact for the AML patient of this new treatment modality would be a less toxic treatment with a lesser percentage of relapse resulting in an improvement of outcome associated with a better quality of life. These full length IgG bispecific antibodies are clinically evaluated in relapsed AML patients. The clinical efficacy is analyzed using AML blast reduction in the bone marrow as an objective response criterion. An efficacious bispecific IgG for AML provides a novel therapeutic option for a large patient segment for which there is currently no treatment available. In addition to providing a means to achieve durable remissions, this treatment option also has a curative potential for AML when applied during remission.
EXAMPLES
[0053] Example 1 : Generation and functional characterization of a candidate
CD3xCLEC12 bispecific IgGl
To validate the concept of targeting an immune effector cell to an aberrant cell with a bispecific full length IgG, a candidate CD3XCLEC12A bispecific IgGl was generated for which the CD3 and CLEC12A Fab arms were derived from antibodies previously described. In the CD3 Fab arm, the VH region from anti-CD3 antibody 15C3, one of the CD3-specific antibodies as disclosed in WO2005/118635, was used and this VH is referred to as '3056'. In the CLEC12A Fab arm, the VH region from scFv SC02-357, one of the CLEC12A-specific antibodies as disclosed in WO2005/000894, was used (hereafter named 'CLEC12A benchmark [Fab arm or antibody]'; alternatively this VH is referred to as '3116'). The nucleotide and amino acid sequences of the VH of the CD3 arm (3056), as well as the nucleotide and amino acid sequences of the VH of the CLEC12A arm (3116) of this candidate molecule, which is referred to as candidate 3056x31 16, are provided in Figure 20. The nucleotide and amino acid sequences of the common VL (huVKl-39; 012) are also provided in Figure 20.
[0054] The respective VH regions were cloned into expression vectors using methods known in the art for production of bispecific IgGl (Gunasekaran et al. JBC 2010 (285) 19637-19646; WO2009/089004), in conjunction with the rearranged human IGKV1-39/IGKJ1 (huVKl-39) light chain. The huVKl-39 was previously shown to be able to pair with more than one heavy chain thereby giving rise to antibodies with diverse specificities, which facilitates the generation of bispecific molecules (De Wildt RM et al. J. Mol. Biol. 1999 (285) 895-901 ; De Kruif et al. J. Mol. Biol. 2009 (387) 548-58; WO2009/157771).
[0055] First, the binding of the candidate 3056x3116 CD3xCLEC12A bispecific IgGl to CD3s on HPB-ALL cells was demonstrated by flow cytometry, which was performed according to standard procedures known in the art (Table 1). Binding to cell-expressed CD3s is confirmed using CHO cell transfected with CD35/8 or CD3y/8. The binding of the candidate 3056x31 16 bispecific IgGl to CLEC12A was determined using CHO cells transfected with a CLEC12A expression construct; CD3 monospecific antibody (3056x3056) and CLEC12A monospecific antibody (3116x3116), as well as an irrelevant IgGl isotype control mAb were taken as control.
Table 1 : Binding to cell-expressed CD3 and CLEC12A by flow cytometry.
Figure imgf000023_0001
Results are given as the mean fluorescent intensity. [0056] Affinity measurements of the candidate 3056x3116 bispecific IgGl for CD35/8 and the extracellular domain of CLEC12A are determined by surface plasmon resonance (BIAcore). Briefly, purified recombinant antigens are covalently coupled to the surface of a CM5 sensor chip using free amine chemistry: antigens are diluted in a kAc buffer to 10μg/ml and coupled to a surface that is activated with NHS/EDC (according to the manufacturer's recommendations). To determine the affinities of the Fab arms present in bispecific antibodies, these are serially diluted to 100, 50, 20, 10, 1 and 0.1 nM in Hepes buffered saline (HBS) and flowed over the antigen-coupled surface of the CM5 sensor chip at a high (30μ1/ηΉη) flow rate (to prevent re-binding). Flow cell 1 (FC1) is used as a control (blanc) surface and the responses (sensor grams) resulting from this surface are subtracted from the responses measured on other flow cells (FC). FC2 and FC3 are used for the two different antigens recognized by the bispecific antibody, to be able to measure the affinities of both Fab arms in a single kinetic run over all three surfaces. As the concentration of antibody does not significantly change when it is flowed over an antigen-coupled surface, the on-rates (that are concentration-dependent) of bispecific antibodies are simultaneously measured on the two different antigens they recognize. Sensorgrams of the association and dissociation phases of the different bispecific proteins are thus obtained. Using the BIA evaluation software and curve-fitting employing a 1: 1 interaction model (for monovalent interaction), the affinities of the Fab arms are determined. In case the binding of the bispecific protein to the antigen-coated surface of the sensor chip is compromised (i.e., when very little protein binds, resulting in low responses and/or very fast off-rates), the setup of the experiment is reversed: the bispecific antibody is covalently coupled to the surface of the sensor chip using free amine chemistry and recombinant purified antigen is flowed over the surface at a high (30μ1/ηΉη) flow rate to measure the affinity of the Fab arm directed to that antigen.
[0057] Next, the functionality of the candidate 3056x3116 CD3xCLEC12A bispecific
Ig was tested. First, the T-cell stimulatory capacity was investigated with healthy donor resting T-cells. Briefly, peripheral blood was obtained from healthy donors after informed consent. T-cells were isolated by standard density gradient isolation to enrich for peripheral blood mononuclear cells (PBMC), followed by negative selection using magnetic beads (pan T-cell kit, Miltenyi Biotec, cat.no.130-091-155). Using this purification strategy, T-cells were so-called 'untouched' (i.e., not stained by antibodies, so-called 'resting T cells') to limit the possibility of pre-activation. Purified resting T-cells were subsequently incubated with cells from the leukemia-derived HL60 cell line in 10% fetal bovine serum (FBS) or 10% normal human serum (HS) at an effector: target cell ratio of 10: 1 for two days. Results were expressed as the percentage of CD69-positive or CD25-positive cells within the CD4-positive or CD8-positive T-cell population.
[0058] Both the bivalent CD3 IgG and the CD3XCLEC12A bispecific IgG efficiently induced upregulation of the T-cell activation markers CD69 and CD25 on CD4-positive and CD8-positive T-cells (FIG. 2). In the presence of FBS which did not block Fc receptors present on HL60 cells (Liesveld et al. 1988, J. Immunol. 140(5), pages 1527-1533), also the control bispecific molecule CD3Xisotype control IgG was shown to induce T-cell activation. This effect was diminished in the presence of HS, suggesting that the observed T-cell activation by monovalent CD3 binding of the CD3Xisotype control IgG was dependent on Fc cross-linking. However, T-cell activation induced by the candidate 3056x3116 CD3xCLEC12A bispecific IgG was only partially dependent on Fc-inter actions, as the potency to upregulate CD69 and CD25 was largely maintained in the presence of HS (FIG. 2). This indicated that the intrinsic potency of monovalent CD3 binding was sufficient to activate T-cells when the binding molecule bridged to the CLEC12A antigen on the HL60 target cells following binding with the other Fab arm.
[0059] To investigate whether the extent of T-cell activation by the candidate
3056x3116 CD3XCLEC12A bispecific IgG is sufficient to induce target cell lysis, the HL60 cells in this assay were labeled with carboxyfluorescein diacetate succimidyl ester (CFSE) and cocultured with T-cells at various effector: target cell ratios. After one, two or three days, surviving CFSE-positive HL60 cells were quantified by flow cytometry. Results were expressed as the percentage of specific lysis related to PBS.
[0060] As expected, CD3 monospecific bivalent IgG induced resting T-cell mediated killing of HL60 cells (FIG. 3). Surprisingly, CD3XCLEC12A bispecific monovalent IgG and the control CD3Xisotype control also induced resting T-cell mediated killing of HL60 cells. These effects were most prominent when the assay was performed in the absence of excess of human IgG, i.e., when the Fc receptors on the HL60 target cells were not blocked (FBS condition; FIG. 3). Surprisingly, even in the presence of excess human IgG (10% HS condition) the CD3XCLEC12A bispecific IgG was very efficient in killing HL60 cells indicating that the induction of HL60 lysis is not dependent on Fey receptor interactions. On day 3 also HL60 lysis induced by the CD3Xisotype control was observed, probably due to incomplete Fc-gamma receptor blockade upon extended incubation periods. HL60 target cell killing varied with different effector: target cell ratios (FIG 4).
[0061] In conclusion, this example demonstrates that a CD3xCLEC12A bispecific molecule is a potent inducer of T-cell mediated tumor cell lysis and confirms our hypothesis that T cell engagement for effective killing of aberrant cells can be mediated by a CD3xCLEC12A full length IgGl bispecific antibody. Surprisingly, the activity induced by the CD3XCLEC12A bispecific IgG is not dependent on Fey receptor interactions. To extend the panel of CD3XCLEC12A bispecific full length IgG in order to arrive at a final clinical candidate, panels of CD3 Fab arms and CLEC12A Fab arms are generated. Validation of specificity and functionality of CD3 and CLEC12A Fab arms is done by fixing the other arm using the respective Fab from the candidate 3056x3116 CD3XCLEC12A bispecific IgG shown in the current example. [0062] Example 2: Generation and characterization of CD3 Fab arms for
CD3xCLEC12 bsAb
[0063] Example 1 showed that CD3xCLEC12A bispecific molecules can be potent inducers of T-cell mediated tumor cell lysis. Therefore, to generate more extensive panels of such bispecific molecules separate panels of CD3 binders as well as CLEC12A binders were generated.
[0064] For generation of a panel of CD3 binders, CD3s-specific VH regions are generated by immunization of mice transgenic for the huVKl-39 light chain (WO2009/ 157771) and for a human heavy chain (HC) minilocus with CD3s in various formats: (1) isolated CD35/8 or CD3y/s that may be fused of coupled to a carrier molecule (such as human IgG-Fc or a His-tag) as known in the art with or without adjuvant, (2) cells expressing CD35/8 or CD3y/s, or (3) DNA construct encoding CD35/8 or CD3y/8, or a combination of these strategies. From immunized mice displaying a sufficient antigen-specific titer as determined by ELISA and/or flow cytometry, spleens and/or lymph nodes are harvested from which Fab phage libraries are generated. Alternatively, VH region sequences are derived directly from spleen and lymph node material by deep sequencing (co-pending US provisional application 61/539,116). [0065] Antigen- specific Fab arms are selected from phage libraries from immunized mice or from synthetic phage display libraries which contain the VL region of the huVKl-39 light chain and a collection of human VH regions. For generation of synthetic libraries, randomized CDR3 primers were used as described in De Kruif et al. 1995, J Mol Biol 248(1), pages 97-105. Bacteriophages from these libraries are selected in multiple rounds for binding to isolated CD35/8 protein that may be coupled to a carrier molecule (see above) or to cells expressing CD3s such as HPB-ALL or cells transfected to express CD35/8 or CD3y/8, or a combination of these strategies. Non-binding phages are removed and binding phages are eluted with an acidic buffer or, to direct the selected Fab repertoire to a desired specificity, with antibodies against a specific epitope, for example with antibodies that are cross-reactive to cynomolgous CD3s. These phages are then transfected into competent bacteria which were grown under selection pressure for phage-containing bacteria. After picking a number of surviving bacterial colonies, phages are rescued and submitted to the next selection round.
[0066] After completing selection, the remaining phages are screened for binding to cell-expressed antigen by flow cytometry and to isolated antigen by ELISA. As a positive control for binding, benchmark CD3 antibodies are used such are known in the art, e.g., OKT-3. Nucleotide material from essentially all phages that showed specific binding to antigen-expressing cells is submitted to colony PCR to amplify the VH regions and sequence PCR to determine the VH region sequence. The resulting sequences are clustered based on uniqueness of their HCDR3. For sequences derived from immunized mice, in which (limited) somatic hypermutation can occur, VH sequences are further grouped based on the likelihood of a unique VDJ (i.e., if HCDR3 in different clusters contain <2 amino acids difference, they are considered part of the same cluster and are grouped together). From each cluster, one or a few VH regions per cluster are selected for cloning into vectors for expression in a IgG monospecific bivalent format in conjunction with the huVKl-39 light chain. VH regions for which specific binding to isolated antigen and cell-expressed antigen is confirmed are subsequently cloned in vectors for expression in a CD3XCLEC12A bispecific format. These are then characterized to select a candidate with therapeutic potential (see following examples). [0067] Example 3: Generation and characterization of CLEC12 Fab arms for
CD3xCLEC12 bsAb [0068] As it was demonstrated in Example 1 that CD3xCLEC12A bispecific molecules have the potency to induce T-cell mediated tumor cell lysis, we next wished to establish more extensive panels of such bispecific molecules. In addition to the panel of CD3 binders as described in Example 2 we also generated a panel of CLEC12A binders.
[0069] Briefly, CLEC 12 A- specific Fab arms were selected from Fab synthetic phage display libraries which contained the rearranged human IGKV1-39/IGKJ1 VL region and a collection of human VH regions (De Kruif et al. Biotechnol Bioeng. 2010 (106)741-50). Bacteriophages from these banks were selected in two rounds for binding to CLEC12A. This was done by incubation with the extracellular domain of CLEC12A (amino acids 75 to 275) coupled to a His-tag (Sino Biological, cat.no. 11896-H07H) which was coated to a surface. Non-binding phages were removed, binding phages were chemically eluted, and used to infect bacteria which were grown under selection pressure for phage-containing bacteria. After picking a number of surviving bacterial colonies, phages were rescued and submitted to the next round of selection and propagation.
[0070] After completing selection, the remaining phages were screened for binding to
CLEC12A expressed on the tumor cell line HL60 by flow cytometry. As a positive control for binding, the CLEC12A benchmark antibody was used. Nucleotide material from essentially all phages that showed specific binding to CLEC12A-expressing cells was submitted to colony PCR to amplify the VH regions and sequence PCR to determine the VH region sequence. The resulting sequences were clustered based on uniqueness of their HCDR3. The VH regions from each unique HCDR3 cluster were cloned into vectors for expression in IgG monospecific or bispecific formats in conjunction with the rearranged human IGKV1-39/IGKJ1 LC.
[0071] Three selected CLEC12A binding molecules with a unique HCDR3 sequence showed the desired profile in IgG format, which comprised the following characteristics (Table 2 and data not shown):
[0072] Specific binding to isolated extracellular domain of CLEC12A.
[0073] Specific binding to CLEC12A expressed on a tumor cell line.
[0074] Confirmation of myeloid lineage-specific expression pattern on human PBMC.
[0075] Table 2: Characterization of CLEC 12A Fab arms.
Figure imgf000029_0001
* Tested in ELISA, extracellular domain of CLEC12A (Sino Biological) coated at 2 μg/ml, results given as optical density (background signal isotype control: 0.127).
** Tested by flow cytometry on HL60 cells with optimized IgG concentration, results given as mean fluorescent intensity (background signal isotype control: 116).
*** Tested in ELISA with Fab format, against bench mark IgG at 20 μg/ml.
[0076] Example 4: Selection of functional CLEC 12 Fab arm for CD3xCLEC 12 bsAb
[0077] The selected CLEC12A Fab arms as described in Example 3 were subsequently expressed in bispecific IgG format with a new CD3 Fab arm as a fixed arm. This new CD3 Fab arm, referred to as '3896 CD3 IgG' or '3896' in short, also uses the huVKl-39 light chain. The nucleotide and amino acid sequences of this CD3-specific VH candidate 3896 are depicted in Figure 20. Hence, various bispecific CD3XCLEC12A molecules were expressed that all had the same 3896 anti-CD3 arm but that differed in the CLEC12A arm (either the CLEC12A benchmark arm, or any one of the candidate CLEC12A Fab arms 4327, 4331 or 3918). These CD3XCLEC12A bispecific molecules were then functionally tested in a target cell lysis assay as described in Example 1. Results were expressed as the percentage of specific lysis related to the isotype control. All candidate CLEC12A Fab arms showed a dose-dependent specific lysis of HL60 target cells in the bispecific format, with kinetics that were similar to or better than when the CLEC12A benchmark Fab arm used (FIG. 5). [0078] Also, the CD3xisotype control bsAb showed a dose-dependent target cell lysis, although 1 log higher concentrations were required for the same extent of specific lysis. Despite the presence of excess human IgG via addition of HS, killing activity of this monovalent CD3 IgG was still apparent, probably by Fc-mediated cross-linking. As will be clear from Example 7, this target non-specific lysis can indeed be fully abrogated via silencing Fc receptor interaction by CH2 engineering.
[0079] Example 5: efficacy of CD3xCLEC12 product candidates using AML T cells and/or AML tumor cells
[0080] Examples 1 and 4 demonstrated the potency of CD3XCLEC12A bispecific IgG using either CD3 Fab arm 3056 or 3896 and using the CLEC12A Fab arm candidates 4327, 4331 or 3918 or the CLEC12A benchmark Fab arm 3116 in inducing HL60 target cell lysis mediated by healthy donor resting T-cells. In the current example, it is investigated whether T-cells derived from patients with AML, one of the primary indications for therapeutic application of a CD3XCLEC12A bispecific drug, can be stimulated to kill tumor targets upon stimulation with a CD3XCLEC12A bispecific full length IgG. Next, it is determined whether patient-derived T-cells can kill autologous AML tumor cell blasts upon stimulation with a CD3XCLEC12A bispecific full length IgG.
[0081] T-cells are isolated from peripheral blood of AML patients according to procedures as described in Example 1. Purified patient-derived T-cells are then incubated with
CFSE-labeled HL60 cells and monitored for cell lysis as described in Example 1.
[0082] In addition, the T-cell mediated target cell lysis assay is performed with AML tumor blasts isolated from the same patient (Norde et al. Blood 2009 (113)2312). Isolated blasts are then labeled with CFSE and cocultured with autologous patient-derived T-cells in the presence of the cytokine mixture as described in Norde et al. and in the presence of the
CD3XCLEC12A bispecific IgG or controls. Target cell lysis is monitored as described in
Example 1.
[0083] Example 6: Cytokine release by T cells after contact with CD3XCLEC12A bispecific IgG [0084] Using T-cell stimulatory biologicals, overstimulation of T-cells is a serious risk as this may lead to cytokine release syndrome (Suntharalingam et al. 2006, New England J Med 355(10), pages 1018-1028; Chatenoud et al. 1990, Transplantation 49(4), pages 697-702). To investigate the extent of T-cell stimulation induced by CD3XCLEC12A bispecific IgG, the induction of T-cell cytokines was studied in a coculture of T-cells and Fc receptor-expressing target cells.
[0085] Briefly, healthy donor resting T-cells were cocultured with HL60 target cells in the presence of the candidate 3056x3116 CD3XCLEC12A bispecific IgG (1 μg/ml) or control IgG as described in Example 1. After two days, the supernatant was sampled and cytokine production levels were determined in a Luminex assay as known in the art using the human Cytokine Human 10-Plex Panel (lnvitrogen, cat.no.LHCOOOl). This panel covers the ten major Thl and Th2 cytokines.
[0086] As expected, the CD3 monospecific bivalent IgG induced strong production of
IFNy, TNFa and IL-2 (Table 3), which are considered to mainly drive cytokine release syndrome. In addition, production of IL-4, IL-6, IL-8 and IL-10 was increased by incubation with CD3 IgG. In contrast, the CD3XCLEC12A bispecific IgG only induced IL-8 production to a similar level as CD3 IgG; the other cytokines were not significantly induced by the bispecific IgG. GM-CSF was below the detection limit in any condition.
[0087] Table 3: antibody induced cytokine release by T cells.
Figure imgf000031_0001
Results are given as the average concentration of cytokine in pg/ml of two donor ± standard deviation. [0088] The data shown here suggest a favorable therapeutic profile for the different
CD3XCLEC12A bispecific IgG molecules, as they potently induce target cell lysis (Examples 1 and 4) without triggering T-cells to secrete potentially harmful amounts of pro-inflammatory cytokines as observed with CD3 IgG.
[0089] Example 7: Effect of Fc silencing on in vitro efficacy of CD3XCLEC12A bsAb
[0090] The dose-dependent target cell lysis by the CD3Xisotype control bsAb shown in
Example 4 was suggested to be due to interaction of the bsAb Fc part with Fc receptors on HL60 target cells. As such target non-specific cell lysis may als occur in vivo, either by interaction with Fc receptors on target cells or on bystander cells such as NK cells, engineering of the CH2/lower hinge region was employed to induce silencing of Fc-mediated activity of the bsAb.
[0091] For this, two Fc mutation strategies were examined, using either a 235G 236R double mutation (DM; DM-Fc) or a 234F 235E 33 IS triple mutation (TM; TM-Fc). CD3XCLEC12A bsAbs (3056x3116) with either a DM-Fc or a TM-Fc were generated and confirmed to bind CLEC12A-expressing cells by flow cytometry with the same intensity as the bsAb with wild type Fc (data not shown). Next, these bsAbs and the wild type, DM-Fc and TM-Fc versions of the CD3Xisotype control bsAb were tested in the HL60 target cell lysis assay (see Examples 1 and 4). Results were expressed as the percentage of specific lysis related to the isotype control.
[0092] Fc silencing either by the DM or by the TM had no or only a minor influence on the extent of HL60 cell specific lysis induced by CD3XCLEC12A bsAb (FIG. 6). For the CD3Xisotype control bsAb, however, the potency to induce lysis of HL60 cells was significantly reduced with the TM and even further with the DM.
[0093] This demonstrates that Fc silencing by CH2/lower hinge engineering further contributes to target- specific killing of aberrant cells by creating a bispecific CD3xCLEC12A IgGl format that efficiently and specifically recruits effector cells, and diminishes the potential non-specific immune activation mediated by normal Fey receptor expressing accessory cells. [0094] Example 8: Effect of Fc-silencing on binding to FcRn, CD 16, CD32, CD64 and
Clq [0095] Binding of the candidate 3056x3116 CD3XCLEC12A bsAb with WT Fc or with silenced DM-Fc or a TM-Fc to human FcRn was determined by Bio-Layer Interferometry (BLI, Octet QK, ForteBio). Briefly, purified CD3XCLEC12A WT Fc IgGl, DM-Fc IgGl or TM-Fc IgGl was captured to Protein L biosensors (ForteBio, Cat no 18-5085) at a concentration of 50 μ^ιηΐ in 0.1 M phosphate buffer/0.002%Tween20 containing l.Omg/ml BSA pH6.0 (FcRn-Binding buffer) at RT. Subsequently soluble human FcRn (Sino Biological Inc, CT009-H08H) was added at concentration of 1 μg/ml in FcRn-Binding buffer) at RT. Data analysis using the Octet QK analysis software showed upon normalization for IgG binding to the ProtL sensor that the subsequent binding of CD3XCLEC12A bsAb with DM or TM silenced Fc to human FcRn was comparable to CD3XCLEC12A bsAb with wild-type Fc-tail (FIG. 7) and Fc silencing did thus not affect FcRn binding.
[0096] Binding of CD3XCLEC12A bsAb with silenced Fc to CD 16, CD32 and CD64 is determined by Bio-Layer Interferometry (BLI, Octet QK, ForteBio). Protocol in short: purified CD3XCLEC12A WT Fc IgGl, DM-Fc IgGl or TM-Fc IgGl is captured to Protein L biosensors (ForteBio, Cat no 18-5085) at a concentration of 50 μg/ml in lx Kinetics Buffer (ForteBio 18-5032) at RT. Subsequently recombinant CD 16 (Sino Biological Inc, 10389-H08H1), CD32 (Sino Biological Inc, 10374-H08H) and CD64 (Sino Biological Inc, 10256-H08H) protein is added at concentration of 1.0 μg ml in Kinetics Buffer (ForteBio 18-5032) at RT. Binding of FcR receptors to bsAb is analyzed using Octet QK analysis software.
[0097] Binding of CD3XCLEC 12A bs Ab with silenced Fc to human C 1 q is determined by capture ELISA. To this end purified CD3XCLEC12A WT Fc IgGl, DM-Fc IgGl or TM-Fc IgGl is coated in a concentration range of 25-0.012 μg/ml in PBS on Nunc-Immuno maxisorp F96 plate (Nunc, 439454) O/N at 4C. Subsequently human Clq (Quidel, A400) is added at 2.0 μg/ml in ELISA buffer (2%MILK/PBST). The complex is then visualized using sheep-anti-human Clq polyclonal IgG (Meridian, K90020C) and rabbit-anti-sheep HRP conjugated polyclonal IgG (Southern Biotech, 6150-05). Finally, using TMB substrate (BD 51-2606KC/51-2607KC) binding is developed and OD450 is quantified using a Micro plate reader (Multiskan EX, Thermo Electron Corporation).
[0098] Example 9: Evaluation of in vivo efficacy of CD3xCLEC12A bispecific IgG. [0099] Animal xenograft studies using luciferase expressing HL60 cells (HL60(-Luc) cells) are performed to confirm and extend the in vitro findings using the CD3xCLEC12A bispecific IgGl. More specifically these studies are performed to determine the steady state plasma concentrations at effective doses, which will be taken into account in setting the starting dose for the Phase 1 clinical evaluation. To this purpose NOD/SCID mice (or comparable immune-compromised mice) are injected subcutaneously with an amount of viable HL60(-Luc) cells that results in the establishment of subcutaneous HL60 tumors in the majority of the animals within two weeks upon injection. In parallel with the HL60(Luc) inoculation, or upon initial tumor take, 5xl0E6 or lxlOE7 human PBMC are administered. CD3xCLEC12A bispecific IgG or control monospecific or control bispecific IgG are administered intravenously at several dose levels at the first day of PBMC administration, and 3, 6, and 9 days later. Tumor dimensions are scored 1 week after the initial HL60(Luc) inoculation. The arithmetic average of tumor dimensions (either denoted as tumor volumes or as total bioluminescence) from each group is plotted against time.
[00100] Example 10: Use of a bispecific full length IgGl antibody CD3xCLEC12A in a phase Ia/Ib study.
[00101] The final lead CD3xCLEC12A bispecific full length IgGl candidate is used to manufacture GMP grade material and is clinically evaluated in AML patients. First, a formal non-clinical safety analysis of the product candidate is performed to establish a safe starting dose for first in man studies. Hereafter, an open-label, multi-centre dose escalation Phase Ia/b trial is performed in relapsed and/or refractory AML and in patients unfit for intensive treatment, to explore the safety and tolerability of the CD3xCLEC12A bispecific IgG upon i.v. administration. Secondary endpoints include pharmacokinetic and pharmacodynamic characterization and preliminary efficacy analysis. Overall response rates are assessed by evaluation of the AML blast reduction in the bone marrow. In Phase la the maximum tolerated dose (MTD) is assessed upon single/multiple dose escalation. After interim PK analysis, the Phase lb part of the study entails a dose extension cohort at the MTD or entails further exploration of the dosing frequency.
[00102] Example 11 : Capacity of CD3xCLEC 12A bsAb to induce T cell proliferation. [00103] In patients with AML T cell numbers are low compared to the amount of AML blasts at diagnosis. It is well known that T cells undergo proliferation upon activation resulting in an increased number of T cells. Moreover, in example 1 we have demonstrated that a CD3xCLEC12A bsAb can activate T cells and has the potency to induce T-cell mediated tumor cell lysis. We hypothesized that AML patients treated with CD3xCLEC12A bsAb benefit from expansion of T cell subsets upon CD3xCLEC12A bispecific molecule mediated T cell activation as T cell proliferation will result in an increased number of effector T cells. To demonstrate that CD3xCLEC12A bsAb induces in vitro T cell proliferation, resting T cells were purified, labeled with carboxyfluorescein diacetate succimidyl ester (CFSE) and co-cultured with autologous CLEC12A+ monocytes in the presence of CD3xCLEC12A bsAb or control Abs. To specifically investigate the CD3xCLEC12A induced T cell proliferation without non-specific Fcgamma activation CD3xCLEC12A bsAbs with the DM-Fc tail, as described in Examples 7 and 8, was used. As controls, a CD3xisotype control WT-Fc bsAb, a CD3xisotype control DM-Fc bsAb, a monoclonal CD3 with WT-Fc and an irrelevant isotype control (IgG with WT-FC) were included. Monocytes and T cells from healthy donor peripheral blood were isolated by standard density gradient isolation to enrich for peripheral blood mononuclear cells (PBMC), followed by a CD 14 positive selection for monocytes using CD 14 microbeads (human CD 14 microbeads, Miltenyi Biotec, cat.no. 130-050-201) and a negative selection of untouched T cells using magnetic beads against other leukocytes (pan T-cell isolation kit, Miltenyi Biotec, cat.no. 130- 096-535). The pan T-cell isolation kit allows isolation of resting (untouched) T cells (i.e. not stained with antibodies) avoiding the possibility of pre- activation of T cells.
[00104] CFSE-labeled purified resting T cells were subsequently incubated with purified monocytes and bsAbs in medium with 10% normal human serum (HS) at an effector: target cell ratio of 5: 1 for seven days. At day 7 decrease of CFSE signal as read-out for T cell proliferation was measured by flow cytometry. Results were expressed CFSE signal per CD3+, CD3+CD4+ or CD3+CD8+ T cells in histograms.
[00105] Positive control CD3 WT-Fc Ab induced T cell proliferation whereas isotype control IgG with WT-Fc did not induce T cell proliferation (Figure 8). As expected the CD3xisotype control WT-Fc bsAb did induce T cell proliferation, but to a lower extend when compared to the bivalent monospecific anti-CD3 IgG control. In contrast, the CD3xisotype control DM-Fc bsAb did not induce T cell proliferation due to its silenced Fc-tail. The CD3xCLEC12A DM-Fc bsAb also induced the desired T cell proliferation mediated by specifically bridging CD3 with the CLEC12A antigen
[00106] This shows that a CD3xCLEC12A bsAb is not only capable of target specific induction of T cell mediated tumor lysis as demonstrated previously, but can also potently induce target specific T cell proliferation resulting in an increased number of T cells. Moreover this further demonstrates that Fc silencing by CH2/lower hinge engineering not only contributes to target-specific killing of aberrant cells but also to target-specific induction of T cell proliferation by the CD3xCLEC12A DM-Fc bsAb IgG. [00107] Example 12: Evaluation of CD3xCLEC12A induced expansion of TEMRA subset from AML patients.
[00108] As activation of T cell proliferation was demonstrated for CD3xCLEC12 DM-Fc bsAb, we next wished to investigate whether CD3xCLEC12A DM-Fc bsAb is capable of inducing proliferation of the CD8+ cytotoxic T cell compartment in AML patients. CD8+ cytotoxic T cells have been recognized as the main effectors mediating tumor regression (Sluijter et al., 2010). CD8+ T cells can be divided into four subsets: naive (CCR7+CD45RA+), central memory (TCM, CCR7+CD45RA-), effector memory (TEM, CCR7-CD45RA-), and CD45RA+ effector memory (TEMRA, CCR7-CD45RA+) cells. Studies have shown that naive and memory CD8+ T-cell subsets have different capacities to proliferate and differentiate in response to TCR stimulation (Geginat et al., 2003).
[00109] First the CD8+ compartment in peripheral blood of AML patients in clinical remission was analyzed in comparison to healthy donors. To this end PBMC were isolated from frozen peripheral blood samples from AML patients and healthy donors by standard density gradient isolation. Next, PBMCs were stained with CCR7, CD3, CD4, CD8, CD45RA and CD45RO antibodies to analyze for the CD8+ T cell subsets by flow cytometry. Results were expressed as percentage of a subset in the total CD8+ T cell compartment.
[00110] Analogous to what was previously described, it was observed that the naive CD8+ T cell subset was reduced in blood from AML patients compared to the naive CD8+ T cell subset from healthy individuals, whereas the TEMRA compartment (CCR7-CD45RA+) was increased in AML patients compared to healthy donors (Figure 9). [00111] Next, experiments are performed to study tumor target specific T cell proliferation of the AML patient T cell compartment. More specifically, these experiments are performed to determine if the CD3xCLEC12A DM-Fc bsAb can enhance T cell proliferation and outgrowth of the effector T cell subsets (TEM and TEMRA) of AML patients relative to the naive CD8+ T cells of AML patients.
[00112] To this end resting T cells from AML patients in clinical remission are purified according to example 1 1. Composition of the CD8+ T cell subsets at day = 0 is analyzed by staining of the PBMC with CCR7, CD3, CD4, CD8, CD45RA and CD45RO antibodies, followed by flow cytometric analysis. In addition, resting T cells are either labeled with CFSE or not labeled (CFSE labeling as described in example 11) and co-cultured with HL60 leukemia cells at an E:T ratio 5: 1 with control or test antibodies for 7 days. CFSE labeled T cells are used for quantification of T cell proliferation, whereas unlabeled T cells are used to determine the percentage of proliferated T cell subsets. CFSE-labeled and unlabeled T cells are incubated with PBS, isotype control WT-Fc Ab, CD3xCLEC12A DM-Fc bsAb, CD3xisotype control DM-Fc bsAb and CD3 monoclonal Ab with WT-Fc at 1 μg/ml. After 7 days, CFSE labeled T cell are stained with CD3, CD4 and CD8 antibodies and subjected to FACS analysis to determine the absolute T cells numbers and number of cell divisions, whereas unlabelled CFSE T cells are stained with CCR7, CD3, CD4, CD8, CD45RA and CD45RO antibodies to determine composition of the proliferated CD8+ T cell subsets by flow cytometry. T cell proliferation is expressed as CFSE signal per T cell subset in histograms and the size of the four CD8+ T cells subsets is expressed as percentage within the total CD8+ T cell compartment.
[00113] Example 13: Efficacy of CD3xCLEC12A bsAb to induce AML patient T cell mediated tumour cell lysis.
[00114] In example 1 it was demonstrated that a CD3xCLEC12A bsAb can induce killing of CLEC12A-positive HL60 cells by resting T cells from healthy donors. Next we investigated the capacity of the CD3xCLEC12A bsAb to induce target-specific activation of AML patient T cells and its capacity to induce AML patient T cell mediated killing of HL60 cells.
[00115] T cells were isolated from frozen peripheral blood of AML patients (AML FAB classification AML-M1/M2, M4 or M5) in clinical remission using pan T-cell isolation kit as described in example 1 1. Purified AML patient derived resting T-cells were subsequently incubated with CSFE-labeled HL60 cells in medium supplemented with 10% normal HS at an effector: target cell ratio of 5: 1 for two days, in the presence of PBS, isotype control WT-Fc Ab, CD3xCLEC12A DM-Fc, CD3xisotype DM-Fc, and positive control CD 3 WT-Fc Ab (all antibodies at concentration of 1 μg/ml). After two days of co-culture, T cell activation was determined by flow cytometric analysis for CD3, CD4, and CD25. These results were expressed as percentage CD25+ cells per CD4+ T cells. Moreover, surviving CFSE-positive HL60 cells were quantified by flow cytometry. Results were expressed as the percentage of specific lysis relative to IgG.
[00116] These data show that the antigen-specific activation of healthy donor and AML patient T cells mediated by CD3xCLEC12A DM-Fc bsAb was comparable (Figure 10A). As expected the CD3xisotype control DM-Fc bsAb did not induce T cell activation of health donor nor AML patient derived T cells. It was demonstrated that the CD3xCLEC12A DM-Fc bsAb mediated lysis of HL60 cells by AML patient derived T-cells (68% HL60 cell lysis) was comparable to that by healthy donor T cells (69% HL60 cell lysis, Figure 10B). As expected, the CD3xisotype control DM-Fc bsAb did not induce killing of HL60 cells, neither by AML patient T cells nor by healthy donor T cells. Thus, the CD3xCLEC12A bispecific molecule is a potent inducer of T cell mediated tumor cell lysis, regardless of whether these T cells are AML patient derived or from healthy donors.
[00117] As it was shown that the CD3xCLEC12A bsAb has the capacity to induce potent lysis of HL60 tumor cells by AML patient T cells, subsequently the capacity of the CD3xCLEC12A bsAb to target specific activation of AML T cells was evaluated. In addition, the capacity of the CD3xCLEC12A bsAb to induce lysis of primary CLEC12A-positive AML blasts by AML patient derived autologous T cells was determined. First, frozen stored bone marrow samples from AML patients at diagnosis samples containing >70% of primary AML blasts as determined by flow cytometric analysis were thawed, cultured overnight (O/N) in IMDM medium supplemented with 10% FCS, lOOng/ml GM-CSF, lOOng/ml G-CSF, 50ng/ml IL-3, 25ng/ml SCF and 20ng/ml Flt3L as previously described (Norde et al., 2009). After O/N culture, primary AML blasts were phenotyped for surface expression of CLEC12A, CD3, CD4, CD8, CD14, CD19, CD33, CD34, CD38, CD45 and CD117 by flow cytometry and labelled with CFSE. Resting autologous patient derived T cells, collected when the patient had achieved clinical remission, were isolated from the peripheral blood using the pan T-cell isolation kit as described in example 11. Subsequently, AML blasts were co-cultured with resting autologous T cells at an E:T ratio of 5: 1 in medium with 10% HS for two days. The conditions tested included PBS, isotype control Ab WT-Fc, CD3xCLEC12A DM-Fc, CD3xisotype control DM-Fc and positive control CD3 WT-Fc Ab (all antibodies at 1 μg/ml). After two days of co-culture, T cell activation was determined by flow cytometric analysis for CD3, CD4, CD8, and CD25. These results were expressed as percentage CD25+ cells per CD4+ or CD8+ AML T cells. AML blast lysis was determined by quantification of the surviving CFSE7CD4510W double positive AML blasts by flow cytometry. Results were expressed as the percentage of specific blast lysis relative to IgG.
[00118] These data demonstrate that the CD3xCLEC12A DM-Fc bsAb has the capacity to induce AML blast target specific activation of AML T cells comparable to the monoclonal CD3 WT-Fc positive control Ab (Figure 11A/B). Moreover these data demonstrate that the CD3xCLEC12A bsAb induced potent killing of autologous AML blasts by AML patient-derived T cells is as potent as the killing induced by the monoclonal CD3 WT-Fc positive control Ab (Figure 11C). As expected, no or minor AML blast killing was induced by the CD3xisotype control DM-Fc Ab, which indicates that the observed AML blast killing mediated by the CD3xCLEC12A bsAb is primarily the result of antigen-specific activation of T cells and specific lysis of CLEC12A+ AML tumor cells. Overall, this study demonstrates that CD3xCLEC12A bsAb can efficiently induce killing of CLEC12A positive tumor cells by AML patient T cells.
[00119] Example 14: Effect of Fc-silencing on non-specific cytokine release
[00120] In examples 7 and 8 it was demonstrated that CD3xCLEC12A bsAb IgGl format with Fc silencing by CH2/lower hinge engineering (DM-Fc) resulted in reduced affinity for Fcgamma receptors and abrogated non-specific Fc receptor mediated cytotoxicity of the leukemia-derived HL60 cell line. Next, it was investigated whether the bsAb IgGl format with DM-Fc silencing abrogated non-specific Fc receptor mediated cytotoxicity in the presence of Fc receptor-positive bystander cells such as NK cells. In this study, autologous healthy donor derived resting T cells were redirected against CLEC12A-positive monocytes in the presence of other Fc receptor positive bystander innate effector cells such as NK cells. To this end PBMC were isolated from heparinized peripheral blood from healthy donors by density gradient centrifugation and were plated at a density of 1*10A6 cells/ml. PBMC were cultured for two days in medium with 10% FBS in the presence of either PBS, isotype control Ab, CD3xCLEC12A WT-Fc bsAb, CD3xCLEC12A DM-Fc bsAb, CD3xisotype control WT-Fc bsAb, CD3xisotype control DM-Fc bsAb or CD3 monoclonal Ab with WT-Fc. After two days culture, surviving monocytes were quantified by flow cytometry based on CD14-expression. Results were expressed as the percentage of specific lysis related to IgG.
[00121] It was demonstrated that, for the CD3xCLEC12A bispecific antibody, Fc silencing through the presence of the DM-Fc region only had a minor effect on monocytes lysis (Figure 12). In contrast, for the CD3xisotype control bsAb, Fc silencing through the presence of the DM-Fc region significantly reduced the non-specific lysis of monocytes. It is thus concluded that Fc silencing in the CD3xCLEC12A bsAb further contributes to target-specific killing: the CD3xCLEC12A DM-Fc bsAb specifically recruits T cells and diminishes non-specific immune activation mediated by normal Fey receptor expressing accessory cells.
[00122] Next it was questioned whether the Fc silencing by the DM mutation in the CD3xCLEC12A bsAb abrogates the Fc receptor-mediated release of cytokines, known to be associated with cytokine release syndrome (CRS), a common clinical event with antibody therapies brought about by accessory cells. To this end the cytokine profile in the supernatants of the monocyte killing assay described in Figure 13 was analyzed using the cytokine human 10- plex panel for the Luminex platform (lnvitrogen, LHCOOOl) according to manufacturer instructions. The profile of the following human cytokines was measured in day 2 supernatant: GM-CSF, IFN-γ, IL-Ιβ, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10 and TNF-a. Results shown are of cytokine concentration measured in pg/ml. The levels of GM-CSF, IL-4 and IL-5 cytokines were below detection limit of this assay (data not shown).
[00123] The data show that CD3xCLEC12A and CD3xisotype control bsAb, both with WT-Fc tail induced release of IL-Ιβ, IL-6, TNF-a, IL-10, IL-2 and IFN-γ (Figue 13). However, no or only very low levels of those cytokines were found in CD3xCLEC12A and CD3xisotype control bsAb when carrying the DM-Fc tail, with an exception for IL-8. As monocytes are the main source of IL-8, the high IL-8 levels are assumed to be released from the lysed monocytes and are not a result from a-specific FcR mediated release. It is concluded that Fc silencing through the DM mutations in the bsAb IgG format significantly eliminates the Fc receptor mediated release of IL-lb, IL-6, TNF-a, IL-2 and IFN-γ cytokines associated with CRS. Overall, these data demonstrate that the Fc silencing by the DM mutation in the CH2/lower hinge region contributes to the enhancement of the efficiency and specific recruitment of effector cells by CD3xCLEC12A DM-bsAb by diminishing the potential non-specific immune activation mediated by normal Fey receptor expressing accessory cells and associated release of proinflammatory cytokines.
[00124] Example 15
[00125] The binding of candidate 3896 as full length bivalent monoclonal anti-CD3 IgG to membrane bound CD3 was compared with candidate 3056 as full length bivalent monoclonal anti-CD3 IgG by FACS analysis using CD3 expressing HPB-ALL cells. An irrelevant human IgGl served as an isotype control IgG. Flow cytometry was performed according to standard procedures known in the art. As shown in Figure 14A, the 3896 CD3 IgG dose-dependently bound to CD3 on HPB-ALL cells, as did the 3056 CD3 IgG.
[00126] Next, the ability of 3896 CD3 IgG to induce T-cell proliferation was tested in direct comparison to murine OKT3 CD3 antibody, 3056 CD3 IgG, and isotype control IgG. Briefly, the antibodies were serially diluted and immobilized onto 96-well plates. Upon removal of unbound IgG, CFSE-labeled T cells were added and incubated at 37°C. At day 5, the level of induced T cell proliferation was analyzed by flow cytometry. Results are expressed as the percentage of viable T cells displaying at least a twofold reduction in CFSE expression level and are shown in figure 14B. It was demonstrated that the 3896 CD3 IgG as a bivalent monospecific antibody was less potent in inducing T cell proliferation as compared to the candidate 3056 CD3 IgG and murine OKT3. These data suggest that the reduced level of T cell proliferation as induced by 3896 when compared to the 3056 CD3 IgG reflect the reduced CD3 binding capacity as analyzed by flow cytometry. This difference in binding allows for choosing an arm with a desired affinity, resulting in a bispecific antibody that displays a favorable balance between the binding affinities for CD3 and CLEC12A, so that T cells and CLEC12A-positive AML tumor cells are efficiently brought together, and T cell mediated lysis of CLEC12A-positive AML tumor cells is optimally induced.
[00127] To test the potency of the new 3896 anti-CD3 arm versus the 3056 anti-CD3 arm in a CD3xCLEC12A bispecific antibody format, the 3896xCLEC12A benchmark bispecific antibody of example 4 (candidate 3896x3116) and the 3056xCLEC12A benchmark bs antibody of example 1 (candidate 3056x3116) were directly compared in the HL60 cytotoxicity assay as previously described. The results are shown in figure 15. It was observed that the 3896xCLEC12A benchmark bsAb has similar potency as the 3056xCLEC12A benchmark bsAb. Hence, as both bispecific antibodies differ only in their CD3 Fab arm whilst the CLEC12A Fab arm is the same, it is concluded that the functionality of the 3896 CD3 Fab arm is similar to that of the 3056 CD3 Fab arm in a CD3xCLEC 12A bispecific Ab. It is noted that at lower concentrations the candidate 3896x3116 is even better than the candidate 3056x3116. This is favourable because it provides a larger therapeutic window, as explained herein before.
[00128] Example 16
In example 3, a panel of CLEC12A-specific Fab arms was selected from phage display libraries. All CLEC12A binding molecules contained the huVkl-39 light chain. Three CLEC12A binding molecules were selected: Fabs 3918, 4327 and 4331. These Fabs were expressed as full length human IgGl: 3918 CLEC12A IgG, 4327 CLEC12A IgG and 4331 CLEC12A IgG.
The nucleotide and amino acid sequences of the VH of 3918 CLEC12A IgG, the VH of 4327 CLEC12A IgG, the VH of 4331 CLEC12A IgG and the common VL (IGKV1-39; 012) are provided in Figure 20.
The full length CLEC12A antibodies were tested for binding to CLEC12A expressed by HL60 cells.
[00129] The binding of 3918 CLEC12A IgG, 4327 CLEC12A IgG and 4331 CLEC12A IgG to membrane bound CLEC12A was compared with the CLEC12A benchmark antibody (3116) by FACS analysis using CLEC12A expressing HL60 cells. An irrelevant human IgGl served as an isotype control IgG. Flow cytometry was performed according to standard procedures known in the art. As shown in Figure 16, the 4327 CLEC12A IgG bound to CLEC12A in a similar fashion as the CLEC12A benchmark antibody. The other two antibodies, 3918 CLEC 12A IgG and 4331 CLEC 12A IgG also demonstrated a good dose-dependent binding to CLEC12A on HL60 cells. Their binding to CLEC12A seemed somewhat lower as compared to the CLEC12A benchmark antibody.
In conclusion, Fabs 3918, 4327 and 4331 are good CLEC12A binding arms.
[00130]
[00131] Example 17 It was tested whether bispecific molecules containing the 3896 CD3 Fab arm and the CLEC12A Fab arm 3981, 4327 or 4331 were functional.
For this, the VH sequence of the 3896 CD3 Fab arm and the VH region of either the CLEC12A benchmark antibody, the 3918 CLEC12A Fab, the 4327 CLEC12A Fab or the 4331 CLEC12A Fab were cloned into expression vectors using methods known in the art for production of bispecific IgGl (Gunasekaran et al., WO2009/089004) in conjunction with the rearranged huVKl-39 light chain to result in bispecific antibodies; 3896xCLEC12A benchmark, 3896x3918, 3896x4327 and 3896x4331.
These bispecifics were tested for functionality in the previously described HL60 cytotoxicity assay. Resting T cells from two healthy donors (HD 1 and HD2) were co-cultured with CFSE- labeled HL60 cells in the presence of various concentrations of bispecific antibody at an E:T ratio 5: 1 or 48 hours in the presence of 10% HS. Surviving CFSE-positive HL60 cells were quantified by flow cytometry at day 2. Results in Figure 17 are expressed as the percentage specific lysis. For the two individual experiments with T cells from donor 1 (HD1; Figure 17 upper panel) and T cells for donor 2 (HD2; figure 17 lower panel), it was demonstrated that all bispecifics were as potent as the 3896xCLEC12A benchmark bispecific when incubated at high concentration.
[00132] Of note, especially at lower concentrations of bispecific antibodies, it was observed that the 3896x4327 and 3896x4331 bispecific antibodies were more potent than the 3896xCLEC12A benchmark bispecific. Hence, as these bispecific antibodies differ only in their CLEC12A Fab arm whilst the CD3 Fab arm is the same, it can be concluded that the functionality of the 4327 and 4331 CLEC12A Fab arms is more potent as compared to the CLEC12A benchmark Fab arm. Without wishing to be bound to theory, the observed differences between the 3896x4327 and 3896x4331 versus the 3896xCLEC12A benchmark bispecific IgG may reflect a difference in binding affinity of these novel anti-CLEC12A Fab arms or they might be targeting a different CLEC12A epitope that allows a more efficient crosslinking of the tumor cells with CD3 expressing T cells.
[00133] Example 18
In example 2 it was demonstrated that the CLEC12A Fabs 3918 and 4331 competed for binding to an epitope on CLEC12A when tested in ELISA as Fab format against Fab fragments of the CLEC12A benchmark antibody. The 4327 CLEC12A Fab, however, did not compete with CLEC12A benchmark IgG for binding in this assay (Table 2).
In this experiment, it was tested whether the full length IgG of the 4327 CLEC12A IgG competed for binding to CLEC12A with the CLEC12A benchmark antibody. Briefly, HL60 cells were pre-incubated with primary antibody at 50 μg/ml on ice for 20 minutes. Subsequently,
Oregon Green (OG)-labeled (Invitrogen, cat.no. A 10476) second antibody was added at 1 μg/ml to the cells plus first antibody (concentration of first antibody after addition of OG-labeled IgG -45 μg/ml). After 20 minutes cells were washed and analyzed by FACS.
[00134] The results are shown in Figure 18: it was concluded that 4327 CLEC12A IgG and CLEC12A benchmark IgG compete for binding to CLEC12A. This suggests that both IgGs bind either a closely related epitope on the CLEC12A antigen or that they bind to different epitopes which do not allow simultaneous binding of both IgGs due to steric hindrance.
[00135] Example 19
[00136] In previous examples it was demonstrated that the CLEC12A Fab arms 4327, 4331, 3918 as well as 3116 are good binders to CLEC12A and potent inducers of T cell mediated killing in a CD3xCLEC12A bispecific format. So-far, bispecific antibodies were obtained using known methods for driving immunoglobulin heavy chain heterodimerization (Gunasekaran et al.).
In our co-pending US and PCT applications (US regular application NO: 13/866,747 and
PCT/NL2013/050294; incorporated herein by reference) we have disclosed methods and means for producing bispecific antibodies from a single cell, whereby means are provided that favor the formation of bispecific antibodies over the formation of monospecific antibodies. These methods can also be favorably employed in the present invention. Specifically, preferred mutations to produce essentially only bispecific full length IgG molecules are the amino acid substitutions L351K and T366K (numbering according to Kabat) in the first CH3 domain (the 'KK-variant' heavy chain) and the amino acid substitutions L351D and L368E in the second CH3 domain (the 'DE- variant' heavy chain), or vice versa. It was previously demonstrated in our co-pending US 13/866,747 and PCT/NL2013/050294 applications that the DE-variant and KK- variant preferentially pair to form heterodimers (so-called 'DEKK' bispecific molecules).
Homodimerization of DE-variant heavy chains (DEDE homodimers) or KK-variant heavy chains (KKKK homodimers) hardly occurs due to strong repulsion between the charged residues in the CH3-CH3 interface between identical heavy chains.
To demonstrate that the effect of CD3xCLEC12A bispecific molecules is not influenced by either the known mutations for heterodimerization (Gunasekaran) or the DEKK mutations, the DE- variant and KK-variant heavy chains were used to drive heterodimerization of the different heavy chains for making CD3xCLEC12A bispecifics. In addition the CH2 / lower hinge double mutations (L235G and G236R; DM) were introduced in these DE- and KK-variant heavy chains. The Fc tail of these resulting bispecific molecules is referred to as 'DM DEKK'.
[00137] Briefly, the VH regions of either the 3116, 4327 or 4331 CLEC12A Fab arms were cloned into expression vectors containing the DE-variant + DM heavy chain whereas the VH region of the 3056 CD3 antibody was cloned into an expression vector containing the KK- variant + DM heavy chain (US regular application NO: 13/866,747 and PCT/NL2013/050294) and these expression vectors, together with a nucleic acid molecule encoding the rearranged human IGKV1-39/IGKJ1 (huVKl-39) light chain, were provided to a host cell such that the host cell expressed and produced bispecific antibodies. The resulting 3056x3116 DM DEKK, 3056x4327 DM DEKK and 3056x4331 DM DEKK bispecific antibodies were subsequently tested for potency in the HL60 cytotoxicity assay as previously described. The results are shown in figure 19: it was demonstrated that all variants are still capable of efficient tumor cell lysis and it was thus concluded that the DM and DEKK mutations can be introduced into the Fc region of the CD3xCLEC12A bispecific antibody, while maintaining the capacity of inducing tumor cell lysis.
REFERENCES Armour et al. Mol. Immunol. 2003 (40) 585-593
Bakker A.B. et al. Cancer Res 2004, 64, p8443-50
Bargou et al. 2008 Science 321 :974
Bluemel et al. 2010 Cancer Immunol. Immunother. 59: 1197 Chames and Baty, MABS 2009 (1) 539-547
Chatenoud et al. 1990, Transplantation 49(4), pages 697-702 Chen C.H. et al. Blood 2006, 107, pl459-67
Cui et al. JBC 2012 (287) 28206-28214
De Kruif et al. 1995, J Mol Biol 248(1), pages 97-105 De Kruif et al. J. Mol. Biol. 2009 (387) 548-58
De Kruif et al. Biotechnol Bioeng. 2010 (106)741-50 De Wildt RM et al. J. Mol. Biol. 1999 (285) 895-901 ;
Dreier et al. 2002 IntJ.Canc. 100:690
Geginat, J. et al. Blood, 2003. 101(11), p. 4260-6 Gunasekaran et al. JBC 2010 (285) 19637-19646
Haagen et al. 1995 Blood 85:3208
Han Y. et al. Blood 2004, 104, p2858-66
Kipriyanov et al. 1998 Int.J.Can. 77:763
Kontermann, MABS 2012 (4) 182-197
Lanzavecchia et al. 1987, Eur.J.Imm. 17: 105
Liu et al. 1985 PNAS 82: 8648
Liesveld et al. 1988, J. Immunol. 140(5), pages 1527-1533
Loffler et al. 2000 Blood 95:2098
Marshall A.S. et al. J Biol Chem 2004, 279, p 14792-802
Merchant et al. Nature Biotechnology 1998 Volume 16, pp 677-681 Moore et al. Blood 2011 (117) 4542-4551
Moshaver et al. 2008 Stem Cells 26:3059
Nissim et al. The EMBO Journal vol.13 no.3 pp.692 - 698. 1994 Norde WJ. et al. Blood 2009 (1 13) (10): p. 2312-23 Offner et al. Molecular Immunology 2006 (43) 763-771 Oganesyan et al. Biol. Crystall. 2008(D64)700 Schaefer et al (Cancer Cell 20, 472-486, October 2011 Sheridan C, Nat Biotechnol. 2012 (30):300-l Staerz et al. 1986 PNAS 83: 1453
Shields RL et al. JBC 2001 (276) 6591-6604 Sluijter, B.J., et al. Clin Immunol, 2010. 137(2), p. 221-33 Suntharalingam et al. 2006, New England J Med 355(10), pages 1018-1028 Van Rhenen et al. 2007 Blood 110:2659 Zeidler et al. 1999 J. Immunol. 163: 1246
WO2004/009618
WO2005/118635
WO2005/000894
WO2005/000894
WO 2008/027236
WO2009/089004
WO2009/ 157771
WO 2010/108127

Claims

1. A bispecific IgG antibody, wherein said bispecific IgG antibody comprises one arm that specifically recognizes CLEC12A or a functional equivalent thereof, and a second arm that specifically recognizes an antigen on immune effector cells capable of recruiting such cells to an aberrant cell expressing CLEC12A or said functional equivalent.
2. The bispecific IgG antibody according to claim 1 , wherein said immune effector cells comprise T cells.
3. The bispecific IgG antibody according to claim 1 or 2, wherein said antigen on said immune effector cells is CD3.
4. The bispecific IgG antibody according to claim 3, wherein said antibody specifically recognizes CD3s.
5. The bispecific IgG antibody according to any one of claims 1-4, wherein both arms comprise a common light chain.
6. The bispecific IgG antibody according to any one of claims 1-5, wherein said common light chain is a germline light chain, preferably the rearranged germline human kappa light chain IgVKl-39*01/IGJKl*01.
7. The bispecific IgG antibody according to any one of claims 1-6, wherein said bispecific antibody is a human IgGl.
8. The bispecific IgG antibody according to any one of claims 1-7, wherein the arm that specifically recognizes CLEC12A or a functional equivalent thereof comprises a heavy chain CDR1 sequence consisting of a sequence that is at least 90% identical to SGYTFTSY and a heavy chain CDR2 sequence consisting of a sequence that is at least 90% identical to IINPSGGS and a heavy chain CDR3 sequence consisting of a sequence that is at least 90% identical to GTTGDWFDY.
9. The bispecific IgG antibody according to claim 8, wherein the arm that specifically recognizes CLEC12A or a functional equivalent thereof comprises a variable heavy chain sequence consisting of a sequence that is at least 90% identical to
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLEWMGIINPSGGS TSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCAKGTTGDWFDYWGQGTLV TVSS.
10. The bispecific IgG antibody according to any one of claims 1-7, wherein the arm that specifically recognizes CLEC12A or a functional equivalent thereof comprises a heavy chain CDR1 sequence consisting of a sequence that is at least 90% identical to SGYTFTSY and a heavy chain CDR2 sequence consisting of a sequence that is at least 90% identical to IINPSGGS and a heavy chain CDR3 sequence consisting of a sequence that is at least 90% identical to GNYGDEFDY.
11. The bispecific IgG antibody according to claim 10, wherein the arm that specifically recognizes CLEC12A or a functional equivalent thereof comprises a variable heavy chain sequence consisting of a sequence that is at least 90% identical to
EVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLEWMGIINPSGGS TSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCARGNYGDEFDYWGQGTLV TVSS.
12. The bispecific IgG antibody according to any one of claims 1-7, wherein the arm that specifically recognizes CLEC12A or a functional equivalent thereof comprises a heavy chain CDR1 sequence consisting of a sequence that is at least 90% identical to SGYTFTGY and a heavy chain CDR2 sequence consisting of a sequence that is at least 90% identical to
WINPNSGG and a heavy chain CDR3 sequence consisting of a sequence that is at least 90% identical to DGYFADAFDY.
13. The bispecific IgG antibody according to claim 12, wherein the arm that specifically recognizes CLEC12A or a functional equivalent thereof comprises a variable heavy chain sequence consisting of a sequence that is at least 90% identical to
QVQLVQSGAEVKKPGASVKVSCKASGYTFTGYYMHWVRQAPGQGLEWMGWINPNSG GTNYAQKFQGRVTMTRDTSISTAYMELSRLRSDDTAVYYCARDGYFADAFDYWGQGT LVTVSS.
14. The bispecific IgG antibody according to any one of claims 3-13, wherein the second arm that specifically recognizes CD3 comprises a heavy chain CDRl sequence consisting of the sequence SYGMH and a heavy chain CDR2 sequence consisting of the sequence
IIWYSGSKKNYADSVKG and a heavy chain CDR3 sequence consisting of the sequence GTGYNWFDP.
15. The bispecific IgG antibody according to claim 14, wherein the second arm that specifically recognizes CD3 comprises a variable heavy chain sequence consisting of the sequence
QVQLVESGGGVVQPGRSLRLSCAASGFTFRSYGMHWVRQAPGKGLEWVAIIWYSGSK KNYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARGTGYNWFDPWGQGTLV TVSS.
16. The bispecific IgG antibody according to any one of claims 1-15, wherein the first and the second arms further comprise a light chain CDRl sequence consisting of a sequence that is at least 90% identical to RASQSISSYLN and a light chain CDR2 sequence consisting of a sequence that is at least 90% identical to AASSLQS and a light chain CDR3 sequence consisting of a sequence that is at least 90% identical to QQSYSTPPT.
17. The bispecific IgG antibody according to claim 16, wherein the first and the second arms comprise a variable light chain sequence consisting of a sequence that is at least 90% identical to
DIQMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQKPGKAPKLLIYAASSLQSGVPSR FSGSGSGTDFTLTISSLQPEDFATYYCQQSYSTPPTFGQGTKVEIK.
18. The bispecific IgG antibody according to any one of claims 1-17, wherein said bispecific IgG antibody has mutated CH2 and/or lower hinge domains such that interaction of said bispecific IgG antibody with Fey receptors is significantly reduced.
19. The bispecific IgG antibody according to claim 18, wherein said mutated CH2 and/or lower hinge domains comprise at least one substitution at amino acids position 235 and/or 236 (numbering according to Kabat).
20. The bispecific IgG antibody according to claim 18 or 19, wherein said mutated CH2 and/or lower hinge domains comprise substitution L235G and/or G236R, preferably L235G and G236R.
21. A method for producing a bispecific IgG antibody according to any one of claims 1-20 from a single cell, wherein said bispecific IgG antibody comprises two CH3 domains that are capable of forming an interface, said method comprising providing:
- a cell having a) a first nucleic acid sequence encoding a IgG heavy chain that specifically recognizes CLEC12A and that contains a 1st CH3 domain, and b) a second nucleic acid sequence encoding a IgG heavy chain that specifically recognizes an antigen on immune effector cells, preferably CD3, and that contains a 2nd CH3 domain, wherein said nucleic acid sequences are provided with means for preferential pairing of said 1st and 2nd CH3 domains, said method further comprising the step of culturing said cell and allowing for expression of said two nucleic acid sequences and harvesting said bispecific IgG antibody from the culture.
22. A method according to claim 21 , wherein said cell has a third nucleic acid sequence encoding a common light chain, preferably the rearranged germline human kappa light chain IgVKl-39*01/IGJKl*01.
23. A method according to claim 21 or 22 for producing a bispecific IgGl antibody, wherein said first CH3 domain comprises the amino acid substitutions L351K and T366K (numbering according to Kabat) and wherein said second CH3 domain comprises the amino acid substitutions L351D and L368E, said method further comprising the step of culturing said cell and allowing for expression of said nucleic acid sequences and harvesting said bispecific antibody from the culture.
24. An antibody obtainable by a method according to any one of claims 21-23.
25. A pharmaceutical composition comprising a bispecific IgG antibody of any one of claims 1-20 or 24 and a pharmaceutically acceptable carrier.
26. A bispecific IgG antibody of any one of claims 1 -20 or 24 for use as a pharmaceutical in the treatment of myelodysplastic syndrome (MDS), chronic myelogenous leukemia (CML) or preferably acute myeloid leukemia (AML).
27. Use of a bispecific IgG antibody according to any one of claims 1-20 or 24 in the preparation of a medicament for the treatment or prevention of myelodysplastic syndrome
(MDS), chronic myelogenous leukemia (CML) or preferably acute myeloid leukemia (AML).
PCT/NL2013/050693 2012-09-27 2013-09-27 BISPECIFIC IgG ANTIBODIES AS T CELL ENGAGERS WO2014051433A1 (en)

Priority Applications (25)

Application Number Priority Date Filing Date Title
RS20181217A RS57910B1 (en) 2012-09-27 2013-09-27 Bispecific igg antibodies as t cell engagers
NZ630563A NZ630563A (en) 2012-09-27 2013-09-27 Bispecific igg antibodies as t cell engagers
EP13777349.5A EP2900694B1 (en) 2012-09-27 2013-09-27 Bispecific igg antibodies as t cell engagers
MX2015003885A MX2015003885A (en) 2012-09-27 2013-09-27 BISPECIFIC IgG ANTIBODIES AS T CELL ENGAGERS.
PL13777349T PL2900694T3 (en) 2012-09-27 2013-09-27 Bispecific igg antibodies as t cell engagers
KR1020157010993A KR102390177B1 (en) 2012-09-27 2013-09-27 Bispecific igg antibodies as t cell engagers
BR112015006824A BR112015006824A2 (en) 2012-09-27 2013-09-27 bispecific igg antibody, method for producing a bispecific igg antibody, antibody, pharmaceutical composition and use of a bispecific igg antibody
SG11201502451QA SG11201502451QA (en) 2012-09-27 2013-09-27 BISPECIFIC IgG ANTIBODIES AS T CELL ENGAGERS
ES13777349.5T ES2692951T3 (en) 2012-09-27 2013-09-27 Bispecific IgG antibodies as T cell couplers
LTEP13777349.5T LT2900694T (en) 2012-09-27 2013-09-27 Bispecific igg antibodies as t cell engagers
CA2889681A CA2889681C (en) 2012-09-27 2013-09-27 Bispecific igg antibodies as t cell engagers
EA201590640A EA201590640A1 (en) 2012-09-27 2013-09-27 IgG CLASS BISPECIFIC ANTIBODIES AS T-CELL RECRUITERS
AU2013324527A AU2013324527B9 (en) 2012-09-27 2013-09-27 Bispecific IgG antibodies as T cell engagers
EP18192737.7A EP3470431A1 (en) 2012-09-27 2013-09-27 Bispecific igg antibodies as t cell engagers
CN201811462337.9A CN110066338B (en) 2012-09-27 2013-09-27 Bispecific IgG antibodies as T cell adaptors
CN201380061615.9A CN105051066B (en) 2012-09-27 2013-09-27 Bispecific IgG antibody as T cell adapter
SI201331229T SI2900694T1 (en) 2012-09-27 2013-09-27 Bispecific igg antibodies as t cell engagers
JP2015534421A JP6471095B2 (en) 2012-09-27 2013-09-27 Bispecific IgG antibody as a T cell engager
DK13777349.5T DK2900694T3 (en) 2012-09-27 2013-09-27 BISPECIFIC IGG ANTIBODIES AS T-CELL ACTIVATORS
IL237945A IL237945B (en) 2012-09-27 2015-03-25 Bispecific igg antibodies as t cell engagers
ZA2015/02835A ZA201502835B (en) 2012-09-27 2015-04-24 Bispecific igg antibodies as t cell engagers
HK16100035.9A HK1211966A1 (en) 2012-09-27 2016-01-05 Bispecific igg antibodies as t cell engagers t igg
AU2018204173A AU2018204173B2 (en) 2012-09-27 2018-06-12 Bispecific IgG antibodies as T cell engagers
HRP20181717TT HRP20181717T1 (en) 2012-09-27 2018-10-19 Bispecific igg antibodies as t cell engagers
CY20181101099T CY1120976T1 (en) 2012-09-27 2018-10-25 TWO SPECIFIC IGG ANTIBODIES AS TYP CELL ACTIVATING FACTORS

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201261706543P 2012-09-27 2012-09-27
US61/706,543 2012-09-27
US201361834915P 2013-06-14 2013-06-14
US61/834,915 2013-06-14

Publications (1)

Publication Number Publication Date
WO2014051433A1 true WO2014051433A1 (en) 2014-04-03

Family

ID=49382559

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/NL2013/050693 WO2014051433A1 (en) 2012-09-27 2013-09-27 BISPECIFIC IgG ANTIBODIES AS T CELL ENGAGERS

Country Status (27)

Country Link
US (2) US10358492B2 (en)
EP (2) EP3470431A1 (en)
JP (2) JP6471095B2 (en)
KR (1) KR102390177B1 (en)
CN (2) CN110066338B (en)
AU (2) AU2013324527B9 (en)
BR (1) BR112015006824A2 (en)
CA (1) CA2889681C (en)
CY (1) CY1120976T1 (en)
DK (1) DK2900694T3 (en)
EA (1) EA201590640A1 (en)
ES (1) ES2692951T3 (en)
HK (1) HK1211966A1 (en)
HR (1) HRP20181717T1 (en)
HU (1) HUE042040T2 (en)
IL (1) IL237945B (en)
LT (1) LT2900694T (en)
MX (1) MX2015003885A (en)
NZ (1) NZ630563A (en)
PL (1) PL2900694T3 (en)
PT (1) PT2900694T (en)
RS (1) RS57910B1 (en)
SG (2) SG10201705787VA (en)
SI (1) SI2900694T1 (en)
TR (1) TR201815768T4 (en)
WO (1) WO2014051433A1 (en)
ZA (1) ZA201502835B (en)

Cited By (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016047722A1 (en) * 2014-09-26 2016-03-31 中外製薬株式会社 Cytotoxicity-inducing therapeutic agent
WO2016079081A1 (en) * 2014-11-20 2016-05-26 F. Hoffmann-La Roche Ag Common light chains and methods of use
WO2016205200A1 (en) * 2015-06-16 2016-12-22 Genentech, Inc. Anti-cll-1 antibodies and methods of use
WO2017010874A1 (en) 2015-07-10 2017-01-19 Merus N.V. Human cd3 binding antibody
WO2017053856A1 (en) * 2015-09-23 2017-03-30 Regeneron Pharmaceuticals, Inc. Optimized anti-cd3 bispecific antibodies and uses thereof
WO2017072208A1 (en) * 2015-10-29 2017-05-04 F. Hoffmann-La Roche Ag Transgenic rabbit with common light chain
WO2017125897A1 (en) 2016-01-21 2017-07-27 Novartis Ag Multispecific molecules targeting cll-1
US9758805B2 (en) 2012-04-20 2017-09-12 Merus N.V. Methods and means for the production of Ig-like molecules
US9828429B2 (en) 2007-09-26 2017-11-28 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
WO2018057915A1 (en) 2016-09-23 2018-03-29 The Regents Of The University Of Michigan Engineered lymphocytes
US10011858B2 (en) 2005-03-31 2018-07-03 Chugai Seiyaku Kabushiki Kaisha Methods for producing polypeptides by regulating polypeptide association
US10106610B2 (en) 2013-02-01 2018-10-23 Regeneron Pharmaceuticals, Inc. Antibodies comprising chimeric constant domains
US10227412B2 (en) 2014-09-12 2019-03-12 Genentech, Inc. Anti-CLL-1 antibodies and immunoconjugates
WO2019070047A1 (en) 2017-10-06 2019-04-11 小野薬品工業株式会社 Bispecific antibody
WO2019108065A1 (en) 2017-12-01 2019-06-06 Merus N.V. Use of bispecific antibody and il-15 for combination therapy
US10358492B2 (en) 2012-09-27 2019-07-23 Merus N.V. Bispecific IgG antibodies as T cell engagers
WO2019156199A1 (en) 2018-02-09 2019-08-15 小野薬品工業株式会社 Bispecific antibody
WO2019190327A2 (en) 2018-03-30 2019-10-03 Merus N.V. Multivalent antibody
US10550193B2 (en) 2014-03-19 2020-02-04 Regeneron Pharmaceuticals, Inc. Methods and antibody compositions for tumor treatment
US10556952B2 (en) 2015-03-30 2020-02-11 Regeneron Pharmaceuticals, Inc. Heavy chain constant regions with reduced binding to Fc gamma receptors
US10568947B2 (en) 2014-07-21 2020-02-25 Novartis Ag Treatment of cancer using a CLL-1 chimeric antigen receptor
WO2020086328A1 (en) * 2018-10-25 2020-04-30 The Medical College Of Wisconsin, Inc. Targeting clptm1l for treatment and prevention of cancer
US10662244B2 (en) 2014-11-17 2020-05-26 Regeneron Pharmaceuticals, Inc. Methods for tumor treatment using CD3XCD20 bispecific antibody
WO2020130829A1 (en) 2018-12-20 2020-06-25 Merus N.V. Clec12axcd3 bispecific antibodies and methods for the treatment of disease
WO2020141974A1 (en) 2018-12-31 2020-07-09 Merus N.V. Truncated multivalent multimers
US10781262B2 (en) 2014-11-20 2020-09-22 Hoffmann-La Roche Inc. Combination therapy of T cell activating bispecific antigen binding molecules and PD-1 axis binding antagonists
WO2020204152A1 (en) 2019-04-04 2020-10-08 小野薬品工業株式会社 Bispecific antibody
WO2020204708A1 (en) 2019-03-29 2020-10-08 Merus N.V. Cd3 binding molecules
WO2020227073A1 (en) * 2019-05-04 2020-11-12 Inhibrx, Inc. Clec12a-binding polypeptides and uses thereof
US10844127B2 (en) 2014-02-28 2020-11-24 Merus N.V. Antibodies that bind EGFR and ErbB3
WO2021006199A1 (en) 2019-07-05 2021-01-14 小野薬品工業株式会社 Treatment of hematologic cancer with pd-1/cd3 dual specificity protein
WO2021020416A1 (en) 2019-07-30 2021-02-04 小野薬品工業株式会社 Bispecific antibody
WO2021025140A1 (en) 2019-08-08 2021-02-11 小野薬品工業株式会社 Dual-specific protein
US11046784B2 (en) 2006-03-31 2021-06-29 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
WO2021133167A1 (en) 2019-12-24 2021-07-01 Merus N.V. Tgf-beta-rii binding proteins
US11066483B2 (en) 2010-11-30 2021-07-20 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
US11072666B2 (en) 2016-03-14 2021-07-27 Chugai Seiyaku Kabushiki Kaisha Cell injury inducing therapeutic drug for use in cancer therapy
US11124576B2 (en) 2013-09-27 2021-09-21 Chungai Seiyaku Kabushiki Kaisha Method for producing polypeptide heteromultimer
US11142587B2 (en) 2015-04-01 2021-10-12 Chugai Seiyaku Kabushiki Kaisha Method for producing polypeptide hetero-oligomer
US11161911B2 (en) 2017-10-23 2021-11-02 Go Therapeutics, Inc. Anti-glyco-MUC1 antibodies and their uses
US11230697B2 (en) 2006-09-01 2022-01-25 Therapeutic Human Polyclonals Inc. Enhanced expression of human or humanized immunoglobulin in non-human transgenic animals
WO2022031935A1 (en) 2020-08-05 2022-02-10 Dragonfly Therapeutics, Inc. Antibodies targeting egfr and use thereof
US11279770B2 (en) 2014-02-28 2022-03-22 Merus N.V. Antibody that binds ErbB-2 and ErbB-3
US11590223B2 (en) 2018-08-31 2023-02-28 Regeneron Pharmaceuticals, Inc. Dosing strategy that mitigates cytokine release syndrome for therapeutic antibodies
WO2023107954A1 (en) 2021-12-08 2023-06-15 Dragonfly Therapeutics, Inc. Antibodies targeting 5t4 and uses thereof
US11732043B2 (en) 2017-07-06 2023-08-22 Merus N.V. Antibodies that modulate a biological activity expressed by a cell
US11753470B2 (en) 2017-07-06 2023-09-12 Merus N.V. Bispecific anti PD1-anti TIM3 antibodies
US11773170B2 (en) 2017-08-09 2023-10-03 Merus N.V. Antibodies that bind EGFR and cMET
US11780925B2 (en) 2017-03-31 2023-10-10 Merus N.V. ErbB-2 and ErbB3 binding bispecific antibodies for use in the treatment of cells that have an NRG1 fusion gene
US11939389B2 (en) 2018-06-13 2024-03-26 Novartis Ag BCMA chimeric antigen receptors and uses thereof
US11939394B2 (en) 2015-10-23 2024-03-26 Merus N.V. Binding molecules that inhibit cancer growth
WO2024069165A1 (en) * 2022-09-27 2024-04-04 Coding Bio Limited Cll1 binding molecules

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004009618A2 (en) 2002-07-18 2004-01-29 Crucell Holland B.V. Recombinant production of mixtures of antibodies
USRE47770E1 (en) 2002-07-18 2019-12-17 Merus N.V. Recombinant production of mixtures of antibodies
EP3721902A1 (en) * 2013-03-14 2020-10-14 The Scripps Research Institute Targeting agent antibody conjugates and uses thereof
CA2907429A1 (en) * 2013-07-04 2015-01-08 Rheinisch-Westfaelische Technische Hochschule Aachen A new fusion protein to target and treat acute myloid leukemia cells
WO2016130986A1 (en) * 2015-02-13 2016-08-18 Sorrento Therapeutics, Inc. Antibody therapeutics that bind ctla4
AU2016225993B2 (en) * 2015-03-04 2020-09-24 Yuhan Corporation Antibody therapeutics that bind CD47
EP3464365A1 (en) * 2016-06-01 2019-04-10 Xencor, Inc. Bispecific antibodies that bind cd123 and cd3
AU2018219887A1 (en) * 2017-02-08 2019-08-22 Dragonfly Therapeutics, Inc. Multi-specific binding proteins for activation of natural killer cells and therapeutic uses thereof to treat cancer
CA3235295A1 (en) 2017-02-20 2018-08-23 Dragonfly Therapeutics, Inc. Proteins binding her2, nkg2d and cd16
BR112019017256A2 (en) * 2017-02-20 2020-04-14 Dragonfly Therapeutics Inc gd2, nkg2d and cd16 binding proteins
KR20200010429A (en) * 2017-05-23 2020-01-30 드래곤플라이 쎄라퓨틱스, 인크. Proteins Bind to NKG2D, CD16 and Tumor-associated Antigens
CN111278455A (en) * 2017-05-23 2020-06-12 蜻蜓疗法股份有限公司 Proteins that bind NKG2D, CD16 and tumor-associated antigens
CA3068929A1 (en) * 2017-07-06 2019-01-10 Merus N.V. Binding molecules that modulate a biological activity expressed by a cell
EP3681532A4 (en) * 2017-09-14 2021-09-01 Dragonfly Therapeutics, Inc. Proteins binding nkg2d, cd16, and c-type lectin-like molecule-1 (cll-1)
PE20220278A1 (en) 2018-02-08 2022-02-25 Dragonfly Therapeutics Inc VARIABLE DOMAINS OF ANTIBODIES TARGETING THE NKG2D RECEPTOR
CN112584851A (en) * 2018-06-22 2021-03-30 科优基因公司 Novel interleukin-15 (IL-15) fusion proteins and uses thereof
KR20210081346A (en) * 2018-10-19 2021-07-01 리전츠 오브 더 유니버시티 오브 미네소타 NK Involving Molecules and Methods of Using Same
CN114929745A (en) * 2019-10-17 2022-08-19 明尼苏达大学董事会 CLEC12A antibody fragment sequences and methods
US20230111279A1 (en) * 2021-04-26 2023-04-13 Millennium Pharmaceuticals, Inc. Anti-clec12a antibodies and uses thereof
WO2023002390A1 (en) * 2021-07-20 2023-01-26 Abl Bio Inc. Anti-cll-1 antibodies and uses thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004009618A2 (en) * 2002-07-18 2004-01-29 Crucell Holland B.V. Recombinant production of mixtures of antibodies
WO2005118635A2 (en) * 2004-06-03 2005-12-15 Novimmune S.A. Anti-cd3 antibodies and methods of use thereof
WO2009051974A1 (en) * 2007-10-17 2009-04-23 Nuvelo, Inc. Antibodes to cll-1
WO2011028952A1 (en) * 2009-09-02 2011-03-10 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens

Family Cites Families (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8308235D0 (en) 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4801687A (en) 1986-10-27 1989-01-31 Bioprobe International, Inc. Monoclonal antibody purification process using protein A
NZ226694A (en) 1987-10-28 1994-04-27 Oncogen Human immunoglobulin produced by recombinant dna techniques
US5151504A (en) 1989-11-17 1992-09-29 E. R. Squibb & Sons, Inc. Method for purification of monoclonal antibodies
GB9022543D0 (en) 1990-10-17 1990-11-28 Wellcome Found Antibody production
CA2111858A1 (en) 1991-07-15 1993-02-04 James S. Crowe Production of antibodies
US5457035A (en) 1993-07-23 1995-10-10 Immunex Corporation Cytokine which is a ligand for OX40
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
EP0979281B1 (en) 1997-05-02 2005-07-20 Genentech, Inc. A method for making multispecific antibodies having heteromultimeric and common components
US20020062010A1 (en) 1997-05-02 2002-05-23 Genentech, Inc. Method for making multispecific antibodies having heteromultimeric and common components
CN100457914C (en) 1999-04-15 2009-02-04 荷兰克鲁塞尔公司 Recombinant protein production in human cell using sequences encoding adenovirus E1 protein
EP1806407B1 (en) 2001-07-04 2010-05-05 Chromagenics B.V. DNA sequences having anti-repressor activity
US20050130224A1 (en) 2002-05-31 2005-06-16 Celestar Lexico- Sciences, Inc. Interaction predicting device
WO2004050850A2 (en) * 2002-12-02 2004-06-17 Abgenix, Inc. Antibodies directed to phospholipase a2 and uses thereof
CA2512647C (en) 2003-01-07 2013-10-08 Symphogen A/S Method for manufacturing recombinant polyclonal proteins
US20100069614A1 (en) 2008-06-27 2010-03-18 Merus B.V. Antibody producing non-human mammals
EP2322547A1 (en) 2003-06-25 2011-05-18 Crucell Holland B.V. Myeloid cell-specific lectin
CN1984931B (en) * 2004-06-03 2012-11-28 诺维莫尼公司 Anti-CD3 antibodies and methods of use thereof
SE527196C2 (en) 2004-07-08 2006-01-17 Chemel Ab SIRE flow-through detector
AU2005282700A1 (en) 2004-09-02 2006-03-16 Genentech, Inc. Heteromultimeric molecules
US20060171929A1 (en) 2005-01-31 2006-08-03 The University Of Washington Regulation of dendritic cell functions by the DCAL-2 receptor
AU2006230413B8 (en) 2005-03-31 2011-01-20 Xencor, Inc Fc variants with optimized properties
EP3050963B1 (en) 2005-03-31 2019-09-18 Chugai Seiyaku Kabushiki Kaisha Process for production of polypeptide by regulation of assembly
JP2008537941A (en) * 2005-03-31 2008-10-02 ゼンコー・インコーポレイテッド Fc variants with optimized properties
GB2429517B (en) 2005-07-15 2010-10-06 Viridian Concepts Ltd Solar collector devices
WO2007047901A2 (en) 2005-10-18 2007-04-26 Lacy Kolo Credit fraud prevention systems and methods
JP2009541275A (en) 2006-06-22 2009-11-26 ノボ・ノルデイスク・エー/エス Production of bispecific antibodies
WO2008027236A2 (en) 2006-08-30 2008-03-06 Genentech, Inc. Multispecific antibodies
US8290739B2 (en) 2006-10-20 2012-10-16 Amfit, Inc. Method for determining relative mobility of regions of an object
CN101802015B (en) 2007-03-29 2015-05-06 根马布股份公司 Bispecific antibodies and methods for production thereof
EP2185701A4 (en) 2007-08-15 2011-03-02 Amunix Operating Inc Compositions and methods for modifying properties of biologically active polypeptides
US20090162359A1 (en) 2007-12-21 2009-06-25 Christian Klein Bivalent, bispecific antibodies
US8242247B2 (en) 2007-12-21 2012-08-14 Hoffmann-La Roche Inc. Bivalent, bispecific antibodies
US9266967B2 (en) 2007-12-21 2016-02-23 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
MX350962B (en) 2008-01-07 2017-09-27 Amgen Inc Method for making antibody fc-heterodimeric molecules using electrostatic steering effects.
CA2715043C (en) 2008-02-05 2021-02-16 Zymeworks Inc. Methods for determining correlated residues in a protein or other biopolymer using molecular dynamics
PL2147594T3 (en) 2008-06-27 2014-04-30 Merus Nv Antibody producing non-human mammals
JP2012515540A (en) 2009-01-26 2012-07-12 ゲンマブ エー/エス Methods for producing antibody mixtures
SG174378A1 (en) 2009-03-20 2011-10-28 Genentech Inc Bispecific anti-her antibodies
CA2759233C (en) 2009-04-27 2019-07-16 Oncomed Pharmaceuticals, Inc. Method for making heteromultimeric molecules
KR102010827B1 (en) * 2009-06-26 2019-08-14 리제너론 파마슈티칼스 인코포레이티드 Readily isolated bispecific antibodies with native immunoglobulin format
ES2603559T5 (en) 2010-02-08 2021-02-22 Regeneron Pharma Mouse common light chain
CA2797981C (en) 2010-05-14 2019-04-23 Rinat Neuroscience Corporation Heterodimeric proteins and methods for producing and purifying them
JP2013537416A (en) * 2010-08-13 2013-10-03 メディミューン リミテッド Monomer polypeptide containing mutant Fc region and method of use
RU2608640C2 (en) 2010-08-16 2017-01-23 Новиммун С.А. Methods for generation of multispecific and multivalent antibodies
MX352929B (en) 2010-11-05 2017-12-13 Zymeworks Inc Stable heterodimeric antibody design with mutations in the fc domain.
EA028804B1 (en) 2011-03-25 2018-01-31 Гленмарк Фармасьютикалс С.А. Hetero-dimeric immunoglobulins
CA2791109C (en) 2011-09-26 2021-02-16 Merus B.V. Generation of binding molecules
LT2838918T (en) 2012-04-20 2019-09-10 Merus N.V. Methods and means for the production of heterodimeric ig-like molecules
CA2889681C (en) 2012-09-27 2023-04-11 Merus B.V. Bispecific igg antibodies as t cell engagers
MX2018000344A (en) 2015-07-10 2018-03-14 Merus Nv Human cd3 binding antibody.

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004009618A2 (en) * 2002-07-18 2004-01-29 Crucell Holland B.V. Recombinant production of mixtures of antibodies
WO2005118635A2 (en) * 2004-06-03 2005-12-15 Novimmune S.A. Anti-cd3 antibodies and methods of use thereof
WO2009051974A1 (en) * 2007-10-17 2009-04-23 Nuvelo, Inc. Antibodes to cll-1
WO2011028952A1 (en) * 2009-09-02 2011-03-10 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
ANNA VAN RHENEN ET AL: "The novel AML stem cell associated antigen CLL-1 aids in discrimination between normal and leukemic stem cells", BLOOD, AMERICAN SOCIETY OF HEMATOLOGY, US, vol. 110, no. 7, 1 October 2007 (2007-10-01), pages 2659 - 2666, XP002632479, ISSN: 0006-4971, [retrieved on 20070703], DOI: 10.1182/BLOOD-2007-03-083048 *
BAEUERLE PATRICK A ET AL: "Bispecific T-cell engaging antibodies for cancer therapy", CANCER RESEARCH, AACR, US PHILADELPHIA, PA, vol. 69, no. 12, 15 June 2009 (2009-06-15), pages 4941 - 4944, XP002665118, ISSN: 1538-7445, [retrieved on 20090609], DOI: 10.1158/0008-5472.CAN-09-0547 *
N.N.: "Merus Press release", 7 January 2013 (2013-01-07), XP055094673, Retrieved from the Internet <URL:http://www.merus.nl/uploads/images/archive/20130107_Merus PR first candidate_FINAL.pdf> [retrieved on 20140102], DOI: 10.1093/annonc/mdq179 *
N.N.: "Press Release Merus Presents Preclinical Data on its Novel Bispecific Antibody MCLA-­-117 at EHA 2013 -­- Clinical Candidate Designed for the Treatment of Acute Myeloid Leukemia (AML) -­-", 17 June 2013 (2013-06-17), XP055094670, Retrieved from the Internet <URL:http://www.merus.nl/uploads/docs/Merus PR EHA 17 June 2013 final clean.pdf> [retrieved on 20140102] *
X. ZHAO ET AL: "Targeting C-type lectin-like molecule-1 for antibody-mediated immunotherapy in acute myeloid leukemia", HAEMATOLOGICA, vol. 95, no. 1, 31 July 2009 (2009-07-31), pages 71 - 78, XP055094666, ISSN: 0390-6078, DOI: 10.3324/haematol.2009.009811 *

Cited By (110)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11168344B2 (en) 2005-03-31 2021-11-09 Chugai Seiyaku Kabushiki Kaisha Methods for producing polypeptides by regulating polypeptide association
US10011858B2 (en) 2005-03-31 2018-07-03 Chugai Seiyaku Kabushiki Kaisha Methods for producing polypeptides by regulating polypeptide association
US11046784B2 (en) 2006-03-31 2021-06-29 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
US11230697B2 (en) 2006-09-01 2022-01-25 Therapeutic Human Polyclonals Inc. Enhanced expression of human or humanized immunoglobulin in non-human transgenic animals
US9828429B2 (en) 2007-09-26 2017-11-28 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US11248053B2 (en) 2007-09-26 2022-02-15 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US11066483B2 (en) 2010-11-30 2021-07-20 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
US10337045B2 (en) 2012-04-20 2019-07-02 Merus N.V. Methods and means for the production of Ig-like molecules
US10329596B2 (en) 2012-04-20 2019-06-25 Merus N.V. Methods and means for the production of Ig-like molecules
US10752929B2 (en) 2012-04-20 2020-08-25 Merus N.V. Methods and means for the production of ig-like molecules
US9758805B2 (en) 2012-04-20 2017-09-12 Merus N.V. Methods and means for the production of Ig-like molecules
US10358492B2 (en) 2012-09-27 2019-07-23 Merus N.V. Bispecific IgG antibodies as T cell engagers
US10988537B2 (en) 2013-02-01 2021-04-27 Regeneren Pharmaceuticals, Inc. Antibodies comprising chimeric constant domains
US10106610B2 (en) 2013-02-01 2018-10-23 Regeneron Pharmaceuticals, Inc. Antibodies comprising chimeric constant domains
US11124576B2 (en) 2013-09-27 2021-09-21 Chungai Seiyaku Kabushiki Kaisha Method for producing polypeptide heteromultimer
US11820825B2 (en) 2014-02-28 2023-11-21 Merus N.V. Methods of treating a subject having an EGFR-positive and/or ErbB-3-positive tumor
US11279770B2 (en) 2014-02-28 2022-03-22 Merus N.V. Antibody that binds ErbB-2 and ErbB-3
US10844127B2 (en) 2014-02-28 2020-11-24 Merus N.V. Antibodies that bind EGFR and ErbB3
US10550193B2 (en) 2014-03-19 2020-02-04 Regeneron Pharmaceuticals, Inc. Methods and antibody compositions for tumor treatment
US11434300B2 (en) 2014-03-19 2022-09-06 Regeneron Pharmaceuticals, Inc. Methods and antibody compositions for tumor treatment
US10568947B2 (en) 2014-07-21 2020-02-25 Novartis Ag Treatment of cancer using a CLL-1 chimeric antigen receptor
US10227412B2 (en) 2014-09-12 2019-03-12 Genentech, Inc. Anti-CLL-1 antibodies and immunoconjugates
US11084877B2 (en) 2014-09-12 2021-08-10 Genentech, Inc. Anti-CLL-1 antibodies and immunoconjugates
US10239947B2 (en) 2014-09-12 2019-03-26 Genentech, Inc. Anti-CLL-1 antibodies and immunoconjugates
US10266597B2 (en) 2014-09-12 2019-04-23 Genentech, Inc. Anti-CLL-1 antibodies and immunoconjugates
US9975966B2 (en) 2014-09-26 2018-05-22 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing theraputic agent
WO2016047722A1 (en) * 2014-09-26 2016-03-31 中外製薬株式会社 Cytotoxicity-inducing therapeutic agent
JP7054402B2 (en) 2014-09-26 2022-04-13 中外製薬株式会社 Cytotoxicity-inducing therapeutic agent
JP5941230B1 (en) * 2014-09-26 2016-06-29 中外製薬株式会社 Cell injury inducing treatment
JP5941235B1 (en) * 2014-09-26 2016-06-29 中外製薬株式会社 Cell injury inducing treatment
JP2020141709A (en) * 2014-09-26 2020-09-10 中外製薬株式会社 Cytotoxic induction therapeutic agent
US11001643B2 (en) 2014-09-26 2021-05-11 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
JP2016166174A (en) * 2014-09-26 2016-09-15 中外製薬株式会社 Cytotoxicity-inducing therapeutic agent
US10662244B2 (en) 2014-11-17 2020-05-26 Regeneron Pharmaceuticals, Inc. Methods for tumor treatment using CD3XCD20 bispecific antibody
JP7164949B2 (en) 2014-11-20 2022-11-02 エフ.ホフマン-ラ ロシュ アーゲー Common Light Chains and Methods of Use
US11613587B2 (en) 2014-11-20 2023-03-28 Hoffmann-La Roche Inc. Combination therapy of T cell activating bispecific antigen binding molecules and PD-1 axis binding antagonists
WO2016079081A1 (en) * 2014-11-20 2016-05-26 F. Hoffmann-La Roche Ag Common light chains and methods of use
RU2747011C2 (en) * 2014-11-20 2021-04-23 Ф.Хоффманн-Ля Рош Аг General light chains and methods of their application
EP3747905A1 (en) * 2014-11-20 2020-12-09 F. Hoffmann-La Roche AG Common light chains and methods of use
JP2018504092A (en) * 2014-11-20 2018-02-15 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Common light chain and methods of use
US10781262B2 (en) 2014-11-20 2020-09-22 Hoffmann-La Roche Inc. Combination therapy of T cell activating bispecific antigen binding molecules and PD-1 axis binding antagonists
CN107207609A (en) * 2014-11-20 2017-09-26 豪夫迈·罗氏有限公司 Common light chain and application method
CN107207609B (en) * 2014-11-20 2022-07-19 豪夫迈·罗氏有限公司 Common light chains and methods of use
US11518807B2 (en) 2015-03-30 2022-12-06 Regeneron Pharmaceuticals, Inc. Heavy chain constant regions with reduced binding to Fc gamma receptors
US10556952B2 (en) 2015-03-30 2020-02-11 Regeneron Pharmaceuticals, Inc. Heavy chain constant regions with reduced binding to Fc gamma receptors
US11142587B2 (en) 2015-04-01 2021-10-12 Chugai Seiyaku Kabushiki Kaisha Method for producing polypeptide hetero-oligomer
JP6996983B2 (en) 2015-06-16 2022-02-21 ジェネンテック, インコーポレイテッド Anti-CLL-1 antibody and how to use
US11466087B2 (en) 2015-06-16 2022-10-11 Genentech, Inc. Anti-CLL-1 antibodies and methods of use
WO2016205200A1 (en) * 2015-06-16 2016-12-22 Genentech, Inc. Anti-cll-1 antibodies and methods of use
CN107847568A (en) * 2015-06-16 2018-03-27 豪夫迈·罗氏有限公司 The anti-antibody of CLL 1 and application method
JP2018522541A (en) * 2015-06-16 2018-08-16 ジェネンテック, インコーポレイテッド Anti-CLL-1 antibody and method of use
US10501545B2 (en) 2015-06-16 2019-12-10 Genentech, Inc. Anti-CLL-1 antibodies and methods of use
US9914777B2 (en) 2015-07-10 2018-03-13 Merus N.V. Human CD3 binding antibody
CN108026174B (en) * 2015-07-10 2023-02-17 美勒斯公司 Human CD3 binding antibodies
WO2017010874A1 (en) 2015-07-10 2017-01-19 Merus N.V. Human cd3 binding antibody
US11739148B2 (en) 2015-07-10 2023-08-29 Merus N.V. Human CD3 binding antibody
EP3757129A1 (en) 2015-07-10 2020-12-30 Merus N.V. Human cd3 binding antibody
AU2016293942B2 (en) * 2015-07-10 2022-06-16 Merus N.V. Human CD3 binding antibody
JP7379446B2 (en) 2015-07-10 2023-11-14 メルス ナムローゼ フェンノートシャップ human CD3 binding antibody
CN108026174A (en) * 2015-07-10 2018-05-11 美勒斯公司 People's CD3 binding antibodies
EP3345928A1 (en) 2015-07-10 2018-07-11 Merus N.V. Human cd3 binding antibody
JP7010811B2 (en) 2015-07-10 2022-02-10 メルス ナムローゼ フェンノートシャップ Human CD3 binding antibody
US10266593B2 (en) 2015-07-10 2019-04-23 Merus N.V. Human CD3 binding antibody
JP2022023228A (en) * 2015-07-10 2022-02-07 メルス ナムローゼ フェンノートシャップ Human CD3 binding antibody
JP2018520169A (en) * 2015-07-10 2018-07-26 メルス ナムローゼ フェンノートシャップ Human CD3 binding antibody
WO2017053856A1 (en) * 2015-09-23 2017-03-30 Regeneron Pharmaceuticals, Inc. Optimized anti-cd3 bispecific antibodies and uses thereof
CN108290951A (en) * 2015-09-23 2018-07-17 瑞泽恩制药公司 Optimize AntiCD3 McAb bispecific antibody and its purposes
JP2022064886A (en) * 2015-09-23 2022-04-26 リジェネロン・ファーマシューティカルズ・インコーポレイテッド Optimized anti-cd3 bispecific antibodies and uses thereof
JP2018537516A (en) * 2015-09-23 2018-12-20 リジェネロン・ファーマシューティカルズ・インコーポレイテッドRegeneron Pharmaceuticals, Inc. Optimized anti-CD3 bispecific antibody and use thereof
CN108290951B (en) * 2015-09-23 2022-04-01 瑞泽恩制药公司 Optimized anti-CD 3 bispecific antibodies and uses thereof
JP7023231B2 (en) 2015-09-23 2022-02-21 リジェネロン・ファーマシューティカルズ・インコーポレイテッド Optimized anti-CD3 bispecific antibody and its use
US11939394B2 (en) 2015-10-23 2024-03-26 Merus N.V. Binding molecules that inhibit cancer growth
CN108347906A (en) * 2015-10-29 2018-07-31 豪夫迈·罗氏有限公司 Transgene rabbit with common light chain
CN113897371A (en) * 2015-10-29 2022-01-07 豪夫迈·罗氏有限公司 Transgenic rabbits with common light chain
JP7085992B2 (en) 2015-10-29 2022-06-17 エフ.ホフマン-ラ ロシュ アーゲー Transgenic rabbits with a common light chain
WO2017072208A1 (en) * 2015-10-29 2017-05-04 F. Hoffmann-La Roche Ag Transgenic rabbit with common light chain
JP2018537968A (en) * 2015-10-29 2018-12-27 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Transgenic rabbit with a common light chain
EP3851457A1 (en) 2016-01-21 2021-07-21 Novartis AG Multispecific molecules targeting cll-1
WO2017125897A1 (en) 2016-01-21 2017-07-27 Novartis Ag Multispecific molecules targeting cll-1
US11072666B2 (en) 2016-03-14 2021-07-27 Chugai Seiyaku Kabushiki Kaisha Cell injury inducing therapeutic drug for use in cancer therapy
EP3515503A4 (en) * 2016-09-23 2020-09-23 The Regents Of The University Of Michigan Engineered lymphocytes
WO2018057915A1 (en) 2016-09-23 2018-03-29 The Regents Of The University Of Michigan Engineered lymphocytes
US11780925B2 (en) 2017-03-31 2023-10-10 Merus N.V. ErbB-2 and ErbB3 binding bispecific antibodies for use in the treatment of cells that have an NRG1 fusion gene
US11753470B2 (en) 2017-07-06 2023-09-12 Merus N.V. Bispecific anti PD1-anti TIM3 antibodies
US11732043B2 (en) 2017-07-06 2023-08-22 Merus N.V. Antibodies that modulate a biological activity expressed by a cell
US11773170B2 (en) 2017-08-09 2023-10-03 Merus N.V. Antibodies that bind EGFR and cMET
WO2019070047A1 (en) 2017-10-06 2019-04-11 小野薬品工業株式会社 Bispecific antibody
US11161911B2 (en) 2017-10-23 2021-11-02 Go Therapeutics, Inc. Anti-glyco-MUC1 antibodies and their uses
WO2019108065A1 (en) 2017-12-01 2019-06-06 Merus N.V. Use of bispecific antibody and il-15 for combination therapy
WO2019156199A1 (en) 2018-02-09 2019-08-15 小野薬品工業株式会社 Bispecific antibody
WO2019190327A2 (en) 2018-03-30 2019-10-03 Merus N.V. Multivalent antibody
US11952424B2 (en) 2018-03-30 2024-04-09 Merus N.V. Multivalent antibody
US11939389B2 (en) 2018-06-13 2024-03-26 Novartis Ag BCMA chimeric antigen receptors and uses thereof
US11952428B2 (en) 2018-06-13 2024-04-09 Novartis Ag BCMA chimeric antigen receptors and uses thereof
US11590223B2 (en) 2018-08-31 2023-02-28 Regeneron Pharmaceuticals, Inc. Dosing strategy that mitigates cytokine release syndrome for therapeutic antibodies
WO2020086328A1 (en) * 2018-10-25 2020-04-30 The Medical College Of Wisconsin, Inc. Targeting clptm1l for treatment and prevention of cancer
US11873338B2 (en) 2018-12-20 2024-01-16 Merus N.V. CLEC12AxCD3 bispecific antibodies and methods for the treatment of disease
WO2020130829A1 (en) 2018-12-20 2020-06-25 Merus N.V. Clec12axcd3 bispecific antibodies and methods for the treatment of disease
WO2020141974A1 (en) 2018-12-31 2020-07-09 Merus N.V. Truncated multivalent multimers
WO2020204708A1 (en) 2019-03-29 2020-10-08 Merus N.V. Cd3 binding molecules
WO2020204152A1 (en) 2019-04-04 2020-10-08 小野薬品工業株式会社 Bispecific antibody
WO2020227073A1 (en) * 2019-05-04 2020-11-12 Inhibrx, Inc. Clec12a-binding polypeptides and uses thereof
WO2021006199A1 (en) 2019-07-05 2021-01-14 小野薬品工業株式会社 Treatment of hematologic cancer with pd-1/cd3 dual specificity protein
WO2021020416A1 (en) 2019-07-30 2021-02-04 小野薬品工業株式会社 Bispecific antibody
WO2021025140A1 (en) 2019-08-08 2021-02-11 小野薬品工業株式会社 Dual-specific protein
WO2021133167A1 (en) 2019-12-24 2021-07-01 Merus N.V. Tgf-beta-rii binding proteins
EP4269433A2 (en) 2019-12-24 2023-11-01 Merus N.V. Tgf-beta-rii binding proteins
WO2022031935A1 (en) 2020-08-05 2022-02-10 Dragonfly Therapeutics, Inc. Antibodies targeting egfr and use thereof
WO2023107954A1 (en) 2021-12-08 2023-06-15 Dragonfly Therapeutics, Inc. Antibodies targeting 5t4 and uses thereof
WO2024069165A1 (en) * 2022-09-27 2024-04-04 Coding Bio Limited Cll1 binding molecules

Also Published As

Publication number Publication date
BR112015006824A2 (en) 2017-07-04
KR20150076178A (en) 2015-07-06
CN105051066B (en) 2019-08-06
SI2900694T1 (en) 2018-12-31
JP2019068803A (en) 2019-05-09
HRP20181717T1 (en) 2018-12-28
CA2889681C (en) 2023-04-11
IL237945A0 (en) 2015-05-31
AU2013324527A1 (en) 2015-05-14
ES2692951T3 (en) 2018-12-05
DK2900694T3 (en) 2018-11-19
AU2013324527B9 (en) 2018-07-26
US10358492B2 (en) 2019-07-23
PL2900694T3 (en) 2018-12-31
NZ630563A (en) 2017-04-28
ZA201502835B (en) 2016-01-27
EP2900694A1 (en) 2015-08-05
EP3470431A1 (en) 2019-04-17
AU2013324527B2 (en) 2018-05-24
TR201815768T4 (en) 2018-11-21
SG10201705787VA (en) 2017-08-30
CN110066338B (en) 2024-04-09
US20190352393A1 (en) 2019-11-21
JP6471095B2 (en) 2019-02-20
CY1120976T1 (en) 2020-05-29
EA201590640A1 (en) 2015-11-30
KR102390177B1 (en) 2022-04-26
CN105051066A (en) 2015-11-11
JP2015532278A (en) 2015-11-09
MX2015003885A (en) 2015-07-06
US20140120096A1 (en) 2014-05-01
AU2018204173B2 (en) 2020-05-07
EP2900694B1 (en) 2018-09-12
HK1211966A1 (en) 2016-06-03
CA2889681A1 (en) 2014-04-03
RS57910B1 (en) 2019-01-31
SG11201502451QA (en) 2015-05-28
AU2018204173A1 (en) 2018-06-28
CN110066338A (en) 2019-07-30
HUE042040T2 (en) 2019-06-28
IL237945B (en) 2022-04-01
PT2900694T (en) 2018-11-13
LT2900694T (en) 2018-11-12

Similar Documents

Publication Publication Date Title
AU2018204173B2 (en) Bispecific IgG antibodies as T cell engagers
US20210047426A1 (en) Bi-Specific Monovalent Diabodies That are Capable of Binding CD123 and CD3, and Uses Thereof
TWI718098B (en) Tri-specific binding molecules and methods of use thereof
KR20170137067A (en) Anti-PVRIG antibody and methods of use
JP7115758B2 (en) Tandem diabodies for CD16A-directed NK cell engagement
US20200062858A1 (en) Trispecific antigen binding proteins
JP2024504820A (en) Anti-TNFR2 humanized antibody and its use
WO2020186111A2 (en) Vista-binding antibodies and uses thereof
EA040393B1 (en) BISPECIFIC IgG ANTIBODIES AS T-CELL RECRUITERS
TW202413419A (en) Natural killer (nk) cell engagers binding to nkp46 and bcma variants with fc-engineering
WO2023275621A1 (en) Anti-tigit and anti-pvrig in monotherapy and combination treatments
TW202207977A (en) Compositions comprising a t cell redirection therapeutic and a vla-4 adhesion pathway inhibitor
JP2022525261A (en) Vista-binding antibody and its use
OA20146A (en) Bi-specific monovalent diabodies that are capable of binding to GPA33 and CD3, and uses thereof.

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201380061615.9

Country of ref document: CN

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

Ref document number: 13777349

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 237945

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2015534421

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/A/2015/003885

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112015006824

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: 201590640

Country of ref document: EA

Ref document number: 2013777349

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2889681

Country of ref document: CA

Ref document number: 20157010993

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2013324527

Country of ref document: AU

Date of ref document: 20130927

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112015006824

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20150326