WO2011147834A1 - Antibodies against cd19 and uses thereof - Google Patents

Antibodies against cd19 and uses thereof Download PDF

Info

Publication number
WO2011147834A1
WO2011147834A1 PCT/EP2011/058482 EP2011058482W WO2011147834A1 WO 2011147834 A1 WO2011147834 A1 WO 2011147834A1 EP 2011058482 W EP2011058482 W EP 2011058482W WO 2011147834 A1 WO2011147834 A1 WO 2011147834A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
antibody
region
chain variable
variable domain
Prior art date
Application number
PCT/EP2011/058482
Other languages
French (fr)
Inventor
Nikolaos Dimoudis
Ekkehard Moessner
Original Assignee
Roche Glycart Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Roche Glycart Ag filed Critical Roche Glycart Ag
Publication of WO2011147834A1 publication Critical patent/WO2011147834A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • 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
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation
    • 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
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • 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
    • C07K2317/565Complementarity determining region [CDR]
    • 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
    • C07K2317/567Framework region [FR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/72Increased effector function due to an Fc-modification
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/77Internalization into the cell
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present invention relates to antibodies against human CD 19 (CD 19 antibody), such antibodies that do not bind complement factor Clq, methods for their production, pharmaceutical compositions containing said antibodies, and uses thereof.
  • Human CD 19 is a 95 kDa transmembrane protein (B-cell coreceptor) exclusively expressed on B-cells and on follicular dendritic cells. CD 19 is found in association with CD21 and CD81. CD 19 and CD21 are required for normal B-cell differentiation (Carter, R.H., et al, Immunol. Res. 26 (2002) 45-54). Antibodies against CD19 have been used in several clinical trials (see e. g. Hekman, A., et al., Cancer Immunol. Immunother. 32 (191) 364-372; Vlasfeld, L.T., et al., Cancer Immunol. Immunother.
  • Antibodies against CD19 are e.g. mentioned in WO 2004106381, WO 2005/012493, WO 2006/089133, WO 2007/002223, WO 2006/133450, WO 2006/121852, WO 2003/048209, US 7,109,304, US 2006/0233791, US 2006/0280738, US 2006/0263357, US 2006/0257398, EP 1648512, EP 1629012, US 2008/0138336, WO 2008/022152 and in Bruenke, J., et al, Br. J.
  • Antibody against CD 19 can have inhibitory or stimulating effects on B-cell activation. Binding of CD 19 antibodies to mitogen-stimulated B-cells inhibits the subsequent rise in Ca 2+ and the resulting activation and proliferation of these cells and B-cell proliferation and differentiation can either be inhibited or enhanced by CD 19 antibody depending on the mitogenic stimulus used and the degree of crosslinking by the antibody.
  • antibodies against CD 19 which are useful as a therapeutic agent for treatment of autoimmune, rheumatoid arthritis, lupus, psoriasis, and bone diseases and for tumor treatment.
  • CD ⁇ antibodies and their uses are reported in WO 2007/002223.
  • WO 2006/133450 anti-CD19 antibody therapy for the transplantation is reported.
  • the invention relates to an antibody binding to human CD 19 and being characterized in binding to the same CD 19 epitope to which monoclonal antibody
  • Antibody 381 binds.
  • Antibody 381 is characterized by comprising as heavy chain variable region SEQ ID NO: 1 and as light chain variable region SEQ ID NO: 5.
  • Antibody 381 comprises as CDRs a CDRHl region of SEQ ID NO: 2, a CDRH2 region of SEQ ID NO: 3 and a CDRH3 region of SEQ ID NO: 4, and a CDRLl region of SEQ ID NO: 20, a CDRL2 region of SEQ ID NO: 6 and a CDRL3 region of SEQ
  • antibody 396 and the other mentioned antibodies, light chains, heavy chains and CDRs according to the invention are humanized variants of antibody 381 and of its light and heavy chains and CDRs.
  • the antibody according to the invention is characterized in being of human IgGl isotype comprising mutations L234A, which is alanine instead of leucine at amino acid position 234, and L235A, which is alanine instead of leucine at amino acid position 235.
  • the antibody according to the invention is characterized in being of human IgG4 isotype with or without mutation S228P.
  • the antibody according to the invention contains in one embodiment an Fc part derived from human origin.
  • the antibody according to the invention is characterized in that the heavy chain variable domain comprises as CDRs a) a CDRH1 region of SEQ ID NO: 2, a CDRH2 region of SEQ ID NO: 3 and a CDRH3 region of SEQ ID NO: 4,
  • the antibody according to the invention is characterized in that the light chain variable domain comprises as CDRs a) a CDRLl region of SEQ ID NO: 20, a CDRL2 region of SEQ ID NO: 6 and a CDRL3 region of SEQ ID NO:7,
  • the antibody according to the invention is characterized in that the heavy chain variable domain and the light chain variable domain comprises as CDRs a) a CDRH 1 region of SEQ ID NO: 2, a CDRLL? region of SEQ ID NO: 3 and a CDRH3 region of SEQ ID NO: 4, and a CDRLl region of SEQ ID NO: 20, a CDRL2 region of SEQ ID NO: 6 and a CDRL3 region of SEQ ID NO:7, or b) a CDRH1 region of SEQ ID NO: 9, a CDRH2 region of SEQ ID NO: 10 and a CDRH3 region of SEQ ID NO: 11, and a CDRL1 region of SEQ ID NO: 13, a CDRL2 region of SEQ ID NO: 14 a CDRL3 region of SEQ ID NO: 15.
  • CDRs a) a CDRH 1 region of SEQ ID NO: 2, a CDRLL? region of SEQ ID NO: 3 and a CDRH3 region of SEQ ID NO: 4, and
  • the antibody according to the invention is characterized in that the antibody comprises as heavy chain variable domain a) the heavy chain variable domain of SEQ ID NO: 1 ,
  • the heavy chain variable domain of SEQ ID NO: 8 including mutations T28A, VI 1L, Y91F, and T108S, and optionally mutation K12V.
  • the antibody according to the invention is characterized in that the antibody comprises as light chain variable domain a) the light chain variable domain of SEQ ID NO: 5
  • the antibody according to the invention is characterized in that the antibody comprises as heavy chain variable domain and as light chain variable domain a) the heavy chain variable domain of SEQ ID NO:l and the light chain variable domain of SEQ ID NO:5, b) the heavy chain variable domain of SEQ ID NO: 8 and the light chain variable domain of SEQ ID NO: 12,
  • the antibody according to the invention is characterized in that the heavy chain variable domain comprises in addition to mutation T28A,
  • the antibody according to the invention is characterized by the above mentioned amino acid sequences or amino acid sequence fragments and properties.
  • the antibody according to the invention in one embodiment comprises an Fc part derived from human origin.
  • the antibody according to the invention is of human IgGl or IgG4 isotype.
  • the antibody according to the invention is in a specific embodiment of IgGl isotype modified in the hinge region at about amino acid position 216-240, especially at about amino acid position 220-240, between CHI and CH2 and/or in the second inter-domain region at about amino acid position 327-331 between CH2 and CH3.
  • the antibody is of human IgGl isotype comprising mutations L234A (alanine instead of leucine at amino acid position 234) and L235A.
  • the antibody according to the invention is in one embodiment of human IgG4 isotype with or without mutation S228P.
  • the antibody is in one embodiment a monoclonal antibody and, in addition, a chimeric antibody (human constant chain), or a humanized antibody. In a specific embodiment the antibody is a humanized antibody.
  • the antibody is in one embodiment further characterized by an affinity of 10 ⁇ 8 M (K D ) or less, especially of about 10 ⁇ 9 M to 10 ⁇ 13 M in binding to human CD 19.
  • an antibody according to the invention binds also to mouse CD 19 and chimpanzee CD 19.
  • the invention further provides methods for the recombinant production of such antibodies.
  • the invention further provides a nucleic acid encoding a heavy chain variable domain of an antibody binding to human CD 19, characterized in that said nucleic acid encodes a) the heavy chain variable domain of SEQ ID NO: 1 ,
  • the heavy chain variable domain of SEQ ID NO: 8 including mutations T28A, VI 1L, Y91F, and T108S, and optionally mutation K12V.
  • the invention further provides a nucleic acid encoding a light chain variable domain of an antibody binding to human CD 19, characterized in that said nucleic acid encodes a) the light chain variable domain of SEQ ID NO: 5
  • the invention further provides a nucleic acid encoding a heavy chain variable domain and a light chain domain of an antibody binding to human CD 19, characterized in that said nucleic acid encodes a) the heavy chain variable domain of SEQ ID NO:l and the light chain variable domain of SEQ ID NO:5,
  • the antibody according to the invention blocks B-cell activation without causing B-cell depletion.
  • Such an antibody is antibody 381.
  • the antibody according to the invention blocks B-cell activation and causes in addition B-cell depletion in a whole blood B-cell depletion assay.
  • Such an antibody is antibody 396.
  • the invention further comprises the use of an antibody according to the invention, for the treatment of inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases.
  • the invention further comprises the use of an antibody according to the invention for the manufacture of a medicament for the treatment of diseases, in one embodiment of inflammatory, autoimmune, lupus, psoriasis, or bone diseases.
  • the invention further comprises a method for the manufacture of a medicament for the treatment of diseases, in one embodiment of inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases, characterized in comprising an antibody according to the invention.
  • the antibodies according to the invention have properties causing a benefit for a patient suffering from a disease associated with pathologic increase of B-cells.
  • Antibodies according to the invention provide for the reduction of tumor growth and a significant prolongation of the time to progression.
  • the antibodies according to the invention are characterized by the above mentioned properties .
  • the invention further provides methods for treating inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases, comprising administering to a patient diagnosed as having such disease (and therefore being in need of such a therapy) an antibody against CD 19 according to the invention.
  • the antibody may be administered alone, in a pharmaceutical composition, or alternatively in combination with other medicaments for treating inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases.
  • the antibody is administered in a pharmaceutically effective amount.
  • the invention further comprises the use of an antibody according to the invention for the treatment of inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis or bone diseases, and for the manufacture of a pharmaceutical composition comprising an antibody according to the invention.
  • the invention comprises a method for the manufacture of a pharmaceutical composition comprising an antibody according to the invention.
  • the invention further comprises an antibody according to the invention for the treatment of inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases.
  • the invention further comprises a pharmaceutical composition containing an antibody according to the invention, optionally together with a buffer and/or an adjuvant useful for the formulation of antibodies for pharmaceutical purposes.
  • the invention further comprises a pharmaceutical composition comprising an antibody according to the invention.
  • the invention further provides pharmaceutical compositions comprising such antibodies in a pharmaceutically acceptable carrier.
  • the pharmaceutical composition may be included in an article of manufacture or kit.
  • the invention further provides the use of an antibody according to the invention for the manufacture of a pharmaceutical composition for the treatment of cancer or inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases.
  • the antibody is used in a pharmaceutically effective amount.
  • the invention further comprises the use of an antibody according to the invention for the manufacture of a pharmaceutical composition for the treatment of inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases.
  • the antibody is used in a pharmaceutically effective amount.
  • the invention further comprises a method for the production of a recombinant antibody according to the invention, characterized by expressing a nucleic acid encoding an antibody binding to CD 19 according to the invention in a CHO cell and recovering said antibody from said cell.
  • the invention further comprises a process for the production of an antibody against CD 19 according to the invention, comprising the steps of transforming a host cell, in one embodiment a CHO cell.
  • Daudi target cells (geometric mean vs. antibody concentration).
  • Figure 2 CD 19 target binding of anti-CD 19 antibodies comprising constant domains using Daudi target cells (geometric mean vs. antibody concentration).
  • Figure 3 ADCC assay in vitro with anti-CD 19 antibody constant region variants with Raji cells.
  • Figure 4 B-cell depletion in whole blood assay using anti-CD 19 antibodies.
  • acceptor human framework for the purposes herein is a framework comprising the amino acid sequence of a light chain variable domain (VL) framework or a heavy chain variable domain (VH) framework derived from a human immunoglobulin framework or a human consensus framework, as defined below.
  • An acceptor human framework "derived from” a human immunoglobulin framework or a human consensus framework may comprise the same amino acid sequence thereof, or it may contain amino acid sequence changes. In some embodiments, the number of amino acid changes are 10 or less, 9 or less, 8 or less, 7 or less, 6 or less, 5 or less, 4 or less, 3 or less, or 2 or less.
  • the VL acceptor human framework is identical in sequence to the VL human immunoglobulin framework sequence or human consensus framework sequence.
  • Binding affinity refers to intrinsic binding affinity which reflects a 1 : 1 interaction between members of a binding pair (e.g., antibody and antigen).
  • the affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (K D ). Affinity can be measured by common methods known in the art, including those described herein. Specific illustrative and exemplary embodiments for measuring binding affinity are described in the following.
  • an antibody that binds to CD 19 refers to an antibody that is capable of binding CD 19 with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent in targeting CD 19.
  • the extent of binding of an anti-CD 19 antibody to an unrelated, non-CD 19 protein is less than about 10 % of the binding of the antibody to CD 19 as measured, by Surface Plasmon Resonance.
  • an antibody that binds to CD 19 has a dissociation constant (K D ) of 10 "8 M or less, e.g. from 10 "8 M to 10 "13 M, e.g., from 10 "9 M to 10 "13 M).
  • antibody herein is used in the broadest sense and encompasses various antibody structures, including but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they exhibit the desired antigen-binding activity.
  • An "antibody fragment” refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds. Examples of antibody fragments include but are not limited to Fv, Fab, Fab', Fab'-SH, F(ab') 2 ; diabodies; linear antibodies; single-chain antibody molecules (e.g. scFv); and multispecific antibodies formed from antibody fragments.
  • B-cell depletion refers to a B-cell depletion experiment, wherein human whole blood samples (after erythrocyte removal) were incubated for 24 hours with an antibody according to the invention. B-cell depletion was measured by counting the CD 19 positive cells in a flow cytometer relative to the CD3 positive cells as a reference and with an antibody concentration of 5 ng/ml.
  • the humanized variant antibody 396 shows a depletion potency of human B-cells of 28 % compared to antibody 381, which shows no detectable depletion.
  • Constructs that are comprised of the heavy chain variants HA, HB, or HD show a B-cell depleting behavior
  • constructs having the chimeric heavy chain (either together with the chimeric light chain, or the humanized light chain LE), or the heavy chain variants HE, and HF show a strongly reduced activity.
  • variants HE and HF show B-cell depletion activity at the same low level as the parental antibody, or even less.
  • blocking B-cell activation refers to the inhibitory activity of an antibody according to the invention on the proliferation of human B-cells, measured by FACS and SU-DHL4 cells (DSMZ ACC 495) as cell lines in this assay. Blocking is found, if at an concentration of 1 ⁇ g/ml of the antibody 30 % and more inhibition of proliferation was found compared to the CD 19 negative cell line Plasmacytoma RPMI8226 (ATCC CCL 155).
  • chimeric antibody refers to an antibody in which a portion of the heavy and/or light chain is derived from a particular source or species, while the remainder of the heavy and/or light chain is derived from a different source or species.
  • the "class" of an antibody refers to the type of constant domain or constant region possessed by its heavy chain.
  • the heavy chain constant domains that correspond to the different classes of immunoglobulins are called a, ⁇ , ⁇ , ⁇ , and ⁇ , respectively.
  • the antibody according to the invention is in one embodiment characterized by being of human subclass IgGl with mutations PVA236, L234A/L235A, and/or GLPSS331, or of subclass IgG4.
  • the antibody is characterized by being of any IgG class, in one embodiment being IgGl or IgG4, containing at least one mutation in E233, L234, L235, G236, D270, N297, E318, K320, K322, A327, A330, P331 and/or P329 (numbering according to EU index).
  • the IgGl mutations PVA236, L234A/L235A and/or GLPSS331 are especially suited. It is further in one embodiment that the antibody of IgG4 subclass contains the mutation S228P, or the mutation
  • the antibody according to the invention therefore is in one embodiment an antibody of human subclass IgGl, containing one or more mutation(s) selected from PVA236, GLPSS331 and/or L234A/L235A (numbering according to EU index of Kabat).
  • the present invention refers to an antibody that binds CD 19 and does not bind complement factor Clq, and/or Fc receptor. In one embodiment of the invention, these antibodies do not elicit the complement dependent cytotoxicity (CDC) and/or antibody-dependent cellular cytotoxicity (ADCC). In one embodiment this antibody is characterized in that it binds CD 19, contains an Fc part derived from human origin and does not bind complement factor Clq. In a specific embodiment this antibody is a human or humanized monoclonal antibody. Such an antibody is useful for the treatment of inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases.
  • CDC complement dependent cytotoxicity
  • ADCC antibody-dependent cellular cytotoxicity
  • Antibody effector functions refer to those biological activities attributable to the Fc region of an antibody, which vary with the antibody isotype. Examples of antibody effector functions include: Clq binding and complement dependent cytotoxicity (CDC); Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g. B-cell receptor); and B-cell activation.
  • the antibodies according to the invention contain as Fc part, in one embodiment an Fc part derived from human origin and preferably all other parts of the human constant regions.
  • the Fc part of an antibody is directly involved in complement activation, Clq binding, C3 activation and Fc receptor binding. While the influence of an antibody on the complement system is dependent on certain conditions, binding to Clq is caused by defined binding sites in the Fc part. Such binding sites are known in the state of the art and described e.g. by Lukas, T.J., et al, J. Immunol. 127 (1981) 2555-2560; Brunhouse, R., and Cebra, J.J., Mol. Immunol.
  • binding sites are e.g. L234, L235, D270, N297, E318, K320, K322, P331 and P329
  • Antibodies of subclass IgGl, IgG2 and IgG3 usually show complement activation, Clq binding and C3 activation, whereas IgG4 do not activate the complement system, do not bind Clq and do not activate C3.
  • Fc part derived from human origin and does not bind human complement factor Clq and/or human Fey receptor on NK cells denotes a Fc part which is either a Fc part of a human antibody of the subclass IgG4 or a Fc part of a human antibody of the subclass IgGl, IgG2 or IgG3 which is modified in such a way that no Clq binding, C3 activation and/or FcR binding as defined below can be detected.
  • An "Fc part of an antibody” is a term well known to the skilled artisan and defined on the basis of papain cleavage of antibodies.
  • the Fc part is a human Fc part and especially preferred either from human IgG4 subclass, in a specific embodiment mutated in the hinge region (e.g. S228P and/or L235E) or a mutated Fc part from human IgGl subclass.
  • Fc parts comprising heavy chain constant regions selected from the regions shown in SEQ ID NO: 14 and 15 or included in SEQ ID NO:
  • an antibody according to the present invention binds CD 19 and does not bind complement factor Clq, and/or Fc receptor. In one embodiment of the invention, these antibodies do not elicit the complement dependent cytotoxicity (CDC) and/or antibody-dependent cellular cytotoxicity (ADCC). In one embodiment this antibody is characterized in that it binds CD 19, contains an Fc part derived from human origin and does not bind complement factor Clq. In a specific embodiment this antibody is a human or humanized monoclonal antibody.
  • the effector functions mediated by the Fc part of the antibody Fc region refer to effector functions that operate after the binding of an antibody to an antigen (these functions involve the activation of the complement cascade and/or cell activation by an Fc receptor).
  • the function of the complement cascade can be assessed by the CH50 assay.
  • Sheep red cells sensitized with anti-red cell antibodies (EA) are added to test serum to activate the classical pathway resulting in hemolysis.
  • the volume of serum needed to lyse 50 % of the red cells determines the CH50 unit.
  • the AP-CH50 measures the alternative and the terminal pathways. The procedure is similar except that rabbit red cells are used.
  • the alternative pathway is activated upon addition of test serum. Clq and two serine proteases, Clr and Cls, form the complex CI, the first component of the complement dependent cytotoxicity (CDC) pathway.
  • CDC complement dependent cytotoxicity
  • complement-dependent cytotoxicity refers to lysis of CD 19 expressing human endothelial cells by the antibody according to the invention in the presence of complement.
  • CDC is measured in one embodiment by the treatment of CD 19 expressing human endothelial cells with an antibody according to the invention in the presence of complement.
  • the cells are in one embodiment labeled with calcein.
  • CDC is found if the antibody induces lysis of 20 % or more of the target cells at a concentration of 30 ⁇ g/ml.
  • Binding to the complement factor Clq can be measured in an ELISA assay. In such an assay in principle an ELISA plate is coated with concentration ranges of the antibody, to which purified human Clq or human serum is added.
  • Clq binding is detected by an antibody directed against Clq followed by a peroxidase-labeled conjugate. Detection of binding (maximal binding B max ) is measured as optical density at 405 nm (OD405) for peroxidase substrate ABTS ® (2,2' -azino-di-[3-ethylbenzthiazoline-6-sulfonate (6)]).
  • the present invention in one embodiment refers to an antibody, characterized in that non-binding of the antibody to complement factor Clq refers to such an ELISA assay measurement wherein the maximal binding (B max ) of Clq to an antibody according to the invention at a concentration of 10 ⁇ g/ml of the antibody is 20 % or lower of B max observed with antibody 381 comprising human IgGl constant region of SEQ ID NO: 26 and SEQ ID NO: 27, in one embodiment
  • an antibody according to the invention shows a reduced activation of complement factor C3 in an ELISA assay.
  • the assay is performed in the same manner as the Clq assay.
  • an ELISA plate is coated with concentration ranges of the antibody, to which human serum is added.
  • the present invention refers in one embodiment to an antibody, characterized in that non-binding of the antibody to complement factor C3 refers to such an ELISA assay measurement wherein the maximal binding (B max ) of C3 to the antibody at a concentration of 10 ⁇ g/ml of the antibody is 10 % of B max of antibody 381 comprising human IgGl constant region of SEQ ID NO: 26 and SEQ ID NO: 27, in one embodiment 5 % or lower.
  • ADCC antibody-dependent cellular cytotoxicity
  • the labeled cells are incubated with effector cells and the supernatant is analyzed for released 51 Cr. Controls include the incubation of the target endothelial cells with effector cells but without the antibody.
  • the capacity of the antibodies to induce the initial steps mediating ADCC was investigated by measuring their binding to Fey receptors expressing cells, such as cells, recombinantly expressing FcyRI and/or FcyRIIA or NK cells (expressing essentially FcyRIIIA). Preferably binding to FcyR on NK cells is measured.
  • Fc receptor binding effector functions can be mediated by the interaction of the Fc region of an antibody with Fc receptors (FcRs), which are specialized cell surface receptors on hematopoietic cells.
  • Fc receptors belong to the immunoglobulin superfamily, and have been shown to mediate both the removal of antibody-coated pathogens by phagocytosis of immune complexes, and the lysis of erythrocytes and various other cellular targets (e.g. tumor cells) coated with the corresponding antibody, via antibody dependent cell mediated cytotoxicity (ADCC) (Van de Winkel, J.G. and Anderson, C.L., J. Leukoc. Biol. 49 (1991) 511-524).
  • ADCC antibody dependent cell mediated cytotoxicity
  • FcRs are defined by their specificity for immunoglobulin isotypes; Fc receptors for IgG antibodies are referred to as FcyR, for IgE as FcyR, for IgA as FcyR and so on. Fc receptor binding is described e.g. in Ravetch, J.V., and Kinet, J.P., Annu. Rev.
  • FcyR cross-linking of receptors for the Fc domain of IgG antibodies
  • - FcyRI (CD64) binds monomeric IgG with high affinity and is expressed on macrophages, monocytes, neutrophils and eosinophils.
  • Modification in IgG of at least one of E233-G236, P238, D265, N297, A327 and P329 reduce binding to FcyRI.
  • FcyRIIA is found on many cells involved in killing (e.g. macrophages, monocytes, neutrophils) and seems able to activate the killing process.
  • FcyRIIB seems to play a role in inhibitory processes and is found on B-cells, macrophages and on mast cells and eosinophils. On B-cells it seems to function to suppress further immunoglobulin production and isotype switching to, for example, the IgE class.
  • FcyRIIB acts to inhibit phagocytosis as mediated through FcyRIIA.
  • eosinophils and mast cells the eosinophils and mast cells the
  • B-form may help to suppress activation of these cells through IgE binding to its separate receptor.
  • Reduced binding for FcyRIIA is found e.g. for IgG mutation of at least one of E233-G236, P238, D265, N297, A327, P329, D270, Q295, A327, R292, and K414.
  • - FcyRIII (CD 16) binds IgG with medium to low affinity and exists as two types.
  • FcyRIIIA is found on NK cells, macrophages, eosinophils and some monocytes and T cells and mediates ADCC.
  • FcyRIIIB is highly expressed on neutrophils.
  • Reduced binding to FcyRIIIA is found e.g. for mutation of at least one of E233- G236, P238, D265, N297, A327, P329, D270, Q295, A327, S239, E269, E293, Y296, V303, A327, K338 and D376.
  • Fc receptor when used herein refers to activation receptors characterized by the presence of a cytoplasmatic ITAM sequence associated with the receptor (see e.g. Ravetch, J.V., and Bolland, S., Annu. Rev. Immunol. 19 (2001) 275-290).
  • cytoplasmatic ITAM sequence associated with the receptor
  • Such receptors are FcyRI, FcyRIIA and FcyRIIIA.
  • the antibodies according to the invention in one embodiment show a reduced binding to Fey receptors, especially to FcylllA.
  • no binding of FcyR means that in an antibody concentration of 10 ⁇ g/ml the binding of an antibody according to the invention to NK cells is 10 % or less of the binding found for anti- OX40L antibody LC.001 as reported in WO 2006/029879.
  • IgG4 shows reduced FcR binding
  • antibodies of other IgG subclasses show strong binding.
  • an antibody according to the invention is of IgGl or IgG2 subclass and comprises mutation PVA236, GLPSS331, and/or L234A/L235A.
  • An antibody according to the invention of IgG4 subclass comprises in one embodiment mutation L235E.
  • IgG4 mutations are S228P or L235E and S228P.
  • Cell-mediated effector functions of monoclonal antibodies can be enhanced by engineering their oligosaccharide component as described in Umana, P., et al., Nature Biotechnol. 17 (1999) 176-180; and US 6,602,684.
  • IgGl type antibodies the most commonly used antibodies in cancer immunotherapy, are glycoproteins that have a conserved N-linked glycosylation site at Asn297 in each CH2 domain.
  • ADCC antibody dependent cellular cytotoxicity
  • Glycosylation of human IgGl or IgG3 occurs at Asn297 as core fucosylated biantennary complex oligosaccharide glycosylation terminated with up to 2 Gal residues.
  • These structures are designated as GO, Gl (a 1,6 or a 1,3) or G2 glycan residues, depending from the amount of terminal Gal residues (Raju, T.S., Bioprocess Int. 1 (2003) 44-53).
  • CHO type glycosylation of antibody Fc parts is e.g. described by Routier, F.H., Glycoconjugate J. 14 (1997) 201-207.
  • the "partially- or non- fucosylated anti-CD 19 antibody" according to the invention can be expressed in a glycomodified host cell engineered to express at least one nucleic acid encoding a polypeptide having GnTIII activity in an amount sufficient to partially fucosylate the oligosaccharides in the Fc region.
  • the polypeptide having GnTIII activity is a fusion polypeptide.
  • al,6- fucosyltransferase activity of the host cell can be decreased or eliminated according to US 6,946,292 to generate glycomodified host cells.
  • the amount of antibody fucosylation can be predetermined e.g. either by fermentation conditions (e.g. fermentation time), or by combination of at least two antibodies with different fucosylation amount.
  • a partially- or non- fucosylated anti-CD 19 antibody refers to an anti-CD 19 antibody showing a fucosylation at Asn297 of 50 % or lower compared with antibody 381 comprising human IgGl constant region of SEQ ID NO:26 and SEQ ID NO:27. Such an antibody is useful for the treatment of cancer diseases.
  • the partially- or non-fucosylated anti-CD 19 antibody according to the invention can be produced in a host cell by a method comprising: (a) culturing a host cell engineered to express at least one polynucleotide encoding a fusion polypeptide having GnTIII activity under conditions which permit the production of said antibody with partial fucosylation of the oligosaccharides present on the Fc region of said antibody; and (b) isolating said antibody.
  • the polypeptide having GnTIII activity is a fusion polypeptide, especially comprising the catalytic domain of GnTIII and the Golgi localization domain of a heterologous Golgi resident polypeptide selected from the group consisting of the localization domain of mannosidase II, the localization domain of B(l,2)-N- acetylglucosaminyltransferase I ("GnTI”), the localization domain of mannosidase I, the localization domain of B(l,2)-N-acetylglucosaminyltransferase II (“GnTII”), and the localization domain of al-6 core fucosyltransferase.
  • GnTI the localization domain of mannosidase II
  • GnTII the localization domain of B(l,2)-N-acetylglucosaminyltransferase II
  • al-6 core fucosyltransferase a heterologous Golgi resident poly
  • the Golgi localization domain is from mannosidase II or GnTI.
  • the invention is directed to a method for modifying the glycosylation profile of an anti- CD ⁇ antibody by using such method.
  • An "effective amount" of an agent e.g., a pharmaceutical formulation, refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result.
  • Fc region herein is used to define a C-terminal region of an immunoglobulin heavy chain that contains at least a portion of the constant region.
  • the term includes native sequence Fc regions and variant Fc regions.
  • a human IgG heavy chain Fc region extends from Cys226, or from Pro230, to the carboxyl-terminus of the heavy chain.
  • the C-terminal lysine (Lys447) of the Fc region may or may not be present.
  • EU numbering system also called the EU index, as described in Kabat, E.A., et al, Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, MD (1991).
  • FR Framework or "FR” refers to variable domain residues other than hypervariable region (HVR) residues.
  • the FR of a variable domain generally consists of four FR domains: FR1, FR2, FR3, and FR4. Accordingly, the CDR and FR sequences generally appear in the following sequence in VH (or VL): FR1-CDR1(L1)-FR2- CDR2(L2)-FR3-CDR3(L3)-FR4.
  • full length antibody “intact antibody,” and “whole antibody” are used herein interchangeably to refer to an antibody having a structure substantially similar to a native antibody structure or having heavy chains that contain an Fc region as defined herein.
  • host cell refers to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells.
  • Host cells include “transformants” and “transformed cells,” which include the primary transformed cell and progeny derived therefrom without regard to the number of passages. Progeny may not be completely identical in nucleic acid content to a parent cell, but may contain mutations. Mutant progeny that have the same function or biological activity as screened or selected for in the originally transformed cell are included herein.
  • a "human consensus framework” is a framework which represents the most commonly occurring amino acid residues in a selection of human immunoglobulin VL or VH framework sequences.
  • the selection of human immunoglobulin VL or VH sequences is from a subgroup of variable domain sequences.
  • the subgroup of sequences is a subgroup as in Kabat, E.A., et al., Sequences of Proteins of Immunological Interest, fifth edition, NIH Publication
  • the subgroup is subgroup kappa I as in Kabat, E.A., et al, supra.
  • the subgroup is subgroup III as in Kabat, E.A., et al, supra.
  • a "humanized" antibody refers to a chimeric antibody comprising amino acid residues from non-human CDRs and amino acid residues from human FRs.
  • a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDRs correspond to those of a non-human antibody, and all or substantially all of the FRs correspond to those of a human antibody.
  • a humanized antibody optionally may comprise at least a portion of an antibody constant region derived from a human antibody.
  • a "humanized form" of an antibody, e.g., a non-human antibody refers to an antibody that has undergone humanization.
  • CDR complementarity determining regions
  • an “immunoconjugate” is an antibody conjugated to one or more heterologous molecule(s).
  • mammals include, but are not limited to, domesticated animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats).
  • domesticated animals e.g., cows, sheep, cats, dogs, and horses
  • primates e.g., humans and non-human primates such as monkeys
  • rabbits e.g., mice and rats
  • rodents e.g., mice and rats.
  • the individual or subject is a human.
  • An "isolated" antibody is one which has been separated from a component of its natural environment.
  • an antibody is purified to greater than 95 % or 99 % purity as determined by, for example, electrophoretic (e.g., SDS- PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatographic (e.g., ion exchange or reverse phase HPLC).
  • electrophoretic e.g., SDS- PAGE, isoelectric focusing (IEF), capillary electrophoresis
  • chromatographic e.g., ion exchange or reverse phase HPLC
  • nucleic acid refers to a nucleic acid molecule that has been separated from a component of its natural environment.
  • An isolated nucleic acid includes a nucleic acid molecule contained in cells that ordinarily contain the nucleic acid molecule, but the nucleic acid molecule is present extrachromosomally or at a chromosomal location that is different from its natural chromosomal location.
  • isolated nucleic acid encoding an anti-CD 19 antibody refers to one or more nucleic acid molecules encoding antibody heavy and light chains (or fragments thereof), including such nucleic acid molecule(s) in a single vector or separate vectors, and such nucleic acid molecule(s) present at one or more locations in a host cell.
  • monoclonal antibody refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical and/or bind the same epitope, except for possible variant antibodies, e.g., containing naturally occurring mutations or arising during production of a monoclonal antibody preparation, such variants generally being present in minor amounts.
  • polyclonal antibody preparations typically include different antibodies directed against different determinants (epitopes)
  • each monoclonal antibody of a monoclonal antibody preparation is directed against a single determinant on an antigen.
  • the modifier "monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by a variety of techniques, including but not limited to the hybridoma method, recombinant DNA methods, phage-display methods, and methods utilizing transgenic animals containing all or part of the human immunoglobulin loci, such methods and other exemplary methods for making monoclonal antibodies being described herein.
  • naked antibody refers to an antibody that is not conjugated to a heterologous moiety.
  • the naked antibody may be present in a pharmaceutical formulation.
  • Native antibodies refer to naturally occurring immunoglobulin molecules with varying structures.
  • native IgG antibodies are heterotetrameric glycoproteins of about 150,000 Daltons, composed of two identical light chains and two identical heavy chains that are disulfide-bonded. From N- to C-terminus, each heavy chain has a variable region (VH), also called a variable heavy domain or a heavy chain variable domain, followed by three constant domains (CHI, CH2, and CH3).
  • VH variable region
  • CHI variable heavy domain
  • CH2 constant domains
  • CL constant light
  • the light chain of an antibody may be assigned to one of two types, called kappa ( ⁇ ) and lambda ( ⁇ ), based on the amino acid sequence of its constant domain.
  • package insert is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, combination therapy, contraindications and/or warnings concerning the use of such therapeutic products.
  • Percent (%) amino acid sequence identity with respect to a reference polypeptide sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the reference polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.
  • % amino acid sequence identity values are generated using the sequence comparison computer program ALIGN-2.
  • the ALIGN -2 sequence comparison computer program was authored by Genentech, Inc., and the source code has been filed with user documentation in the U.S. Copyright Office, Washington D.C., 20559, where it is registered under U.S. Copyright Registration
  • the ALIGN-2 program is publicly available from Genentech, Inc., South San Francisco, California, or may be compiled from the source code.
  • the ALIGN-2 program should be compiled for use on a UNIX operating system, including digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary.
  • % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B is calculated as follows:
  • pharmaceutical formulation refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
  • a “pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject.
  • a pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
  • CD 19 refers to human B-lymphocyte antigen CD 19 (alternative name(s) are: Differentiation antigen CD 19, B-lymphocyte surface antigen B4, T-cell surface antigen Leu-12; UniProtKB P15391).
  • treatment refers to clinical intervention in an attempt to alter the natural course of the individual being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include, but are not limited to, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, decreasing the rate of disease progression, amelioration or palliation of the disease state.
  • antibodies of the invention are used to delay development of a disease or to slow the progression of a disease.
  • variable region refers to the domain of an antibody heavy or light chain that is involved in binding the antibody to antigen.
  • the variable domains of the heavy chain and light chain (VH and VL, respectively) of a native antibody generally have similar structures, with each domain comprising four conserved framework regions (FRs) and three hypervariable regions (HVRs).
  • FRs conserved framework regions
  • HVRs hypervariable regions
  • antibodies that bind a particular antigen may be isolated using a VH or VL domain from an antibody that binds the antigen to screen a library of complementary VL or VH domains, respectively. See, e.g., Potolano, S., et al, J. Immunol. 150 (1993) 880-887; Clackson, T., et al, Nature 352 (1991) 624-628.
  • vector refers to a nucleic acid molecule capable of propagating another nucleic acid to which it is linked.
  • the term includes the vector as a self-replicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced.
  • Certain vectors are capable of directing the expression of nucleic acids to which they are operatively linked. Such vectors are referred to herein as "expression vectors.”
  • compositions and methods in one aspect, is based on antibodies that bind to CD 19.
  • Antibodies of the invention are useful, e.g., for the diagnosis or treatment of inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases.
  • an anti-CD 19 antibody is a monoclonal antibody, including a chimeric or humanized antibody.
  • an anti-CD19 antibody is an antibody fragment, e.g., an Fv, Fab, Fab', scFv, diabody, or F(ab') 2 fragment.
  • the antibody is a full length antibody, e.g., an intact IgGl antibody or other antibody class or isotype as defined herein.
  • an anti-CD 19 antibody according to any of the above embodiments may incorporate any of the features, singly or in combination, as described in the sections below: 1. Antibody Affinity
  • an antibody provided herein has a dissociation constant (K D ) of 10 "8 M or less, e.g. from 10 "8 M to 10 "13 M, e.g., from 10 "9 M to 10 "13 M.
  • K D dissociation constant
  • K D is measured using surface plasmon resonance assays using a BIACORE ® -2000 or a BIACORE ® -3000 (BIAcore, Inc.,
  • CM5 chips Piscataway, NJ
  • immobilized antigen CM5 chips at 25 °C with immobilized antigen CM5 chips at ⁇ 10 response units (RU).
  • carboxymethylated dextran biosensor chips CM5, BIACORE, Inc.
  • EDC N-ethyl-N'-(3-dimethylaminopropyl)-carbodiimide hydrochloride
  • NHS N-hydroxysuccinimide
  • an antibody provided herein is an antibody fragment.
  • Antibody fragments include, but are not limited to, Fab, Fab', Fab'-SH, F(ab') 2 , Fv, and scFv fragments, and other fragments described below.
  • Fab fragment antigen binding protein
  • Fab' fragment antigen binding protein
  • Fab'-SH fragment antigen binding protein
  • Fv fragment antigen binding protein
  • scFv fragments fragment antigen binding protein fragments
  • other fragments described below For a review of certain antibody fragments, see Hudson, et al., Nat. Med. 9 (2003) 129-134.
  • scFv fragments see, e.g., Pluckthun, in The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., (Springer-Verlag, New York), pp. 269-315 (1994); see also WO 93/16185; and US 5,571,894 and US 5,587,458.
  • Diabodies are antibody fragments with two antigen-binding sites that may be bivalent or bispecific. See, for example, EP 404,097; WO 1993/01161; Hudson, et al, Nat. Med. 9 (2003) 129-134; and Holliger, P., et al, Proc. Natl. Acad. Sci. USA 90 (1993) 6444-6448. Triabodies and tetrabodies are also described in Hudson, et al, Nat. Med. 9 (2003) 129-134.
  • Single-domain antibodies are antibody fragments comprising all or a portion of the heavy chain variable domain or all or a portion of the light chain variable domain of an antibody.
  • a single-domain antibody is a human single-domain antibody (Domantis, Inc., Waltham, MA; see, e.g., US 6,248,516 Bl).
  • Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as production by recombinant host cells (e.g. E. coli or phage), as described herein.
  • recombinant host cells e.g. E. coli or phage
  • an antibody provided herein is a multispecific antibody, e.g. a bispecific antibody.
  • Multispecific antibodies are monoclonal antibodies that have binding specificities for at least two different sites.
  • one of the binding specificities is for CD 19 and the other is for any other antigen.
  • bispecific antibodies may bind to two different epitopes of CD 19.
  • Bispecific antibodies can be prepared as full length antibodies or antibody fragments.
  • Techniques for making multispecific antibodies include, but are not limited to, recombinant co-expression of two immunoglobulin heavy chain-light chain pairs having different specificities (see Milstein, C, and Cuello, Nature 305 (1983) 537- 540), WO 93/08829, and Traunecker, A., et al, EMBO J. 10 (1991) 3655-3659), and "knob-in-hole” engineering (see, e.g., US 5,731,168).
  • Multi-specific antibodies may also be made by engineering electrostatic steering effects for making antibody Fc-heterodimeric molecules (WO 2009/089004A1); cross-linking two or more antibodies or fragments (see, e.g., US 4,676,980, and Brennan, M., et al, Science, 229 (1985) 81-83); using leucine zippers to produce bi-specific antibodies (see, e.g., Kostelny, S.A., et al, J. Immunol, 148(5) (1992) 1547-1553); using "diabody” technology for making bispecific antibody fragments (see, e.g., Hollinger, et al, Proc. Natl. Acad. Sci.
  • the antibody or fragment herein also includes a "Dual Acting FAb” or “DAF” comprising an antigen binding site that binds to CD 19 as well as another, different antigen (see, US 2008/0069820, for example).
  • the antibody or fragment herein also includes multispecific antibodies described in
  • WO 2009/080251 WO 2009/080252, WO 2009/080253, WO 2009/080254, WO 2010/112193, WO 2010/115589, WO 2010/136172, WO 2010/145792, and WO 2010/145793.
  • an antibody provided herein is altered to increase or decrease the extent to which the antibody is glycosylated.
  • Addition or deletion of glycosylation sites to an antibody may be conveniently accomplished by altering the amino acid sequence such that one or more glycosylation sites is created or removed.
  • the carbohydrate attached thereto may be altered.
  • Native antibodies produced by mammalian cells typically comprise a branched, biantennary oligosaccharide that is generally attached by an N-linkage to Asn297 of the CH2 domain of the Fc region. See, e.g., Wright, et al, TIBTECH 15 (1997) 26-32.
  • the oligosaccharide may include various carbohydrates, e.g., mannose, N-acetyl glucosamine (GlcNAc), galactose, and sialic acid, as well as a fucose attached to a GlcNAc in the "stem" of the biantennary oligosaccharide structure.
  • modifications of the oligosaccharide in an antibody of the invention may be made in order to create antibody variants with certain improved properties.
  • the amount of fucose is determined by calculating the average amount of fucose within the sugar chain at Asn297, relative to the sum of all glycostructures attached to Asn 297 (e.g. complex, hybrid and high mannose structures) as measured by
  • MALDI-TOF mass spectrometry as described in WO 2008/077546, for example.
  • Asn297 refers to the asparagine residue located at about position 297 in the Fc region (Eu numbering of Fc region residues); however, Asn297 may also be located about ⁇ 3 amino acids upstream or downstream of position 297, i.e., between positions 294 and 300, due to minor sequence variations in antibodies.
  • Such fucosylation variants may have improved ADCC function. See, e.g.,
  • WO 2002/031140 Okazaki,. et al, J. Mol. Biol. 336 (2004) 1239-1249; Yamane- Ohnuki, N., et al, Biotech. Bioeng. 87 (2004) 614-622).
  • Examples of cell lines capable of producing defucosylated antibodies include Led 3 CHO cells deficient in protein fucosylation (Ripka, J., et al, Arch. Biochem. Biophys. 249 (1986) 533-
  • knockout cell lines such as alpha- 1,6- fucosyltransferase gene, FUT8, knockout CHO cells (see, e.g., Yamane-Ohnuki,
  • Antibodies variants are further provided with bisected oligosaccharides, e.g., in which a biantennary oligosaccharide attached to the Fc region of the antibody is bisected by GlcNAc. Such antibody variants may have reduced fucosylation and/or improved ADCC function. Examples of such antibody variants are described, e.g., in WO 2003/011878, US 6,602,684, and US 2005/0123546. Antibody variants with at least one galactose residue in the oligosaccharide attached to the Fc region are also provided. Such antibody variants may have improved CDC function. Such antibody variants are described, e.g., in WO 1997/30087, WO 1998/58964, and
  • one or more amino acid modifications may be introduced into the Fc region of an antibody provided herein, thereby generating an Fc region variant.
  • the Fc region variant may comprise a human Fc region sequence (e.g., a human IgGl , IgG2, IgG3 or IgG4 Fc region) comprising an amino acid modification (e.g. a substitution) at one or more amino acid positions.
  • the invention contemplates an antibody variant that possesses some but not all effector functions, which make it a desirable candidate for applications in which the half life of the antibody in vivo is important yet certain effector functions (such as complement and ADCC) are unnecessary or deleterious.
  • In vitro and/or in vivo cytotoxicity assays can be conducted to confirm the reduction/depletion of CDC and/or ADCC activities.
  • Fc receptor (FcR) binding assays can be conducted to ensure that the antibody lacks FcyR binding (hence likely lacking ADCC activity), but retains FcRn binding ability.
  • NK cells express FcyRIII only, whereas monocytes express FcyRI, FcyRII and FcyRIII.
  • FcR expression on hematopoietic cells is summarized in Table 3 on page 464 of Ravetch, J.V., and Kinet, J.P., Annu. Rev. Immunol. 9 (1991) 457-492.
  • Non-limiting examples of in vitro assays to assess ADCC activity of a molecule of interest is described in US 5,500,362 (see, e.g. Hellstrom, I., et al. Proc. Natl. Acad. Sci.
  • non-radioactive assays methods may be employed (see, for example, ACTITM non-radioactive cytotoxicity assay for flow cytometry (CellTechnology, Inc. Mountain View, CA; and CytoTox 96® non-radioactive cytotoxicity assay
  • ADCC activity of the molecule of interest may be assessed in vivo, e.g., in an animal model such as that disclosed in Clynes, R., et al., Proc. Natl. Acad. Sci. USA 95 (1998) 652-656.
  • Clq binding assays may also be carried out to confirm that the antibody is unable to bind C lq and hence lacks CDC activity. See, e.g., C lq and C3c binding ELISA in WO 2006/029879 and WO 2005/100402.
  • a CDC assay may be performed (see, for example, Gazzano-Santoro, H., et al, J. Immunol. Methods 202 (1997) 163-171 ; Cragg, M.S., et al, Blood 101 (2003) 1045-1052; and Cragg, M.S., and M.J., Glennie,
  • FcRn binding and in vivo clearance/half life determinations can also be performed using methods known in the art (see, e.g., Petkova, SB. et al, Int. Immunol. 18 (2006) 1759-1769).
  • Antibodies with reduced effector function include those with substitution of one or more of Fc region residues 238, 265, 269, 270, 297, 327 and 329 (US 6,737,056).
  • Fc mutants include Fc mutants with substitutions at two or more of amino acid positions 265, 269, 270, 297 and 327, including the so-called "DANA" Fc mutant with substitution of residues 265 and 297 to alanine (US 7,332,581).
  • an antibody variant comprises an Fc region with one or more amino acid substitutions which improve ADCC, e.g., substitutions at positions 298, 333, and/or 334 of the Fc region (EU numbering of residues).
  • alterations are made in the Fc region that result in altered (i.e., either improved or diminished) Clq binding and/or Complement Dependent
  • Cytotoxicity e.g., as described in US 6,194,551, WO 99/51642, and Idusogie E.E., et al. J. Immunol. 164 (2000) 4178-4184.
  • FcRn neonatal Fc receptor
  • Fc region residues 238, 256, 265, 272, 286, 303, 305, 307, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 413, 424 or 434, e.g., substitution of Fc region residue 434 (US 7,371,826).
  • cysteine engineered antibodies e.g., "thioMAbs”
  • one or more residues of an antibody are substituted with cysteine residues.
  • the substituted residues occur at accessible sites of the antibody.
  • reactive thiol groups are thereby positioned at accessible sites of the antibody and may be used to conjugate the antibody to other moieties, such as drug moieties or linker-drug moieties, to create an immunoconjugate, as described further herein.
  • any one or more of the following residues may be substituted with cysteine: V205 (Kabat numbering) of the light chain; Al 18 (EU numbering) of the heavy chain; and S400 (EU numbering) of the heavy chain Fc region.
  • Cysteine engineered antibodies may be generated as described, e.g., in US 7,521,541.
  • an antibody provided herein may be further modified to contain additional non-proteinaceous moieties that are known in the art and readily available.
  • the moieties suitable for derivatization of the antibody include but are not limited to water soluble polymers.
  • water soluble polymers include, but are not limited to, polyethylene glycol (PEG), copolymers of ethylene glycol/propylene glycol, carboxymethylcellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone, poly-1, 3-dioxolane, poly-l,3,6-trioxane, ethylene/maleic anhydride copolymer, polyaminoacids (either homopolymers or random copolymers), and dextran or poly(n-vinyl pyrrolidone)poly ethylene glycol, propropylene glycol homopolymers, prolypropylene oxide/ethylene oxide co-polymers, polyoxyethylated polyols (e.g.
  • Polyethylene glycol propionaldehyde may have advantages in manufacturing due to its stability in water.
  • the polymer may be of any molecular weight, and may be branched or unbranched.
  • the number of polymers attached to the antibody may vary, and if more than one polymer is attached, they can be the same or different molecules. In general, the number and/or type of polymers used for derivatization can be determined based on considerations including, but not limited to, the particular properties or functions of the antibody to be improved, whether the antibody derivative will be used in a therapy under defined conditions, etc.
  • conjugates of an antibody and non-proteinaceous moiety that may be selectively heated by exposure to radiation are provided.
  • the non-proteinaceous moiety is a carbon nanotube (Kam, N.W., et al, Proc. Natl. Acad. Sci. USA 102 (2005) 11600-11605).
  • the radiation may be of any wavelength, and includes, but is not limited to, wavelengths that do not harm ordinary cells, but which heat the non-proteinaceous moiety to a temperature at which cells proximal to the antibody-non-proteinaceous moiety are killed.
  • Antibodies may be produced using recombinant methods and compositions, e.g., as described in US 4,816,567.
  • isolated nucleic acid encoding an anti-CD 19 antibody described herein is provided.
  • Such nucleic acid may encode an amino acid sequence comprising the VL and/or an amino acid sequence comprising the VH of the antibody (e.g., the light and/or heavy chains of the antibody).
  • one or more vectors e.g., expression vectors
  • a host cell comprising such nucleic acid is provided.
  • a host cell comprises (e.g., has been transformed with): (1) a vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and an amino acid sequence comprising the VH of the antibody, or (2) a first vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and a second vector comprising a nucleic acid that encodes an amino acid sequence comprising the VH of the antibody.
  • the host cell is eukaryotic, e.g. a Chinese Hamster Ovary (CHO) cell or lymphoid cell (e.g., Y0, NSO, Sp20 cell).
  • a method of making an anti-CD 19 antibody comprises culturing a host cell comprising a nucleic acid encoding the antibody, as provided above, under conditions suitable for expression of the antibody, and optionally recovering the antibody from the host cell (or host cell culture medium).
  • nucleic acid encoding an antibody e.g., as described above, is isolated and inserted into one or more vectors for further cloning and/or expression in a host cell.
  • nucleic acid may be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the antibody).
  • Suitable host cells for cloning or expression of antibody-encoding vectors include prokaryotic or eukaryotic cells described herein.
  • antibodies may be produced in bacteria, in particular when glycosylation and Fc effector function are not needed.
  • For expression of antibody fragments and polypeptides in bacteria see, e.g., US 5,648,237, US 5,789,199, and US 5,840,523. (See also Charlton, K.A., Methods in Molecular Biology, Vol. 248 (B.K.C. Lo, ed., Humana Press, Totowa, NJ, 2004), pp. 245-254, describing expression of antibody fragments in E. coli.)
  • the antibody may be isolated from the bacterial cell paste in a soluble fraction and can be further purified.
  • eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for antibody-encoding vectors, including fungi and yeast strains whose glycosylation pathways have been "humanized” resulting in the production of an antibody with a partially or fully human glycosylation pattern. See Gerngross, Nat. Biotech. 22 (2004) 1409-1414, and Li et al, Nat. Biotech. 24 (2006) 210-215.
  • Suitable host cells for the expression of glycosylated antibody are also derived from multicellular organisms (invertebrates and vertebrates). Examples of invertebrate cells include plant and insect cells. Numerous baculoviral strains have been identified which may be used in conjunction with insect cells, particularly for transfection of Spodoptera frugiperda cells.
  • Plant cell cultures can also be utilized as hosts. See, e.g., US 5,959,177, US 6,040,498, US 6,420,548, US 7,125,978, and US 6,417,429 (describing PLANTIBODIESTM technology for producing antibodies in transgenic plants).
  • Vertebrate cells may also be used as hosts.
  • mammalian cell lines that are adapted to grow in suspension may be useful.
  • Other examples of useful mammalian host cell lines are monkey kidney CVl line transformed by SV40 (COS-7); human embryonic kidney line (293 or 293 cells as described, e.g., in Graham. F.L., et al, J. Gen Virol. 36 (1977) 59-74; baby hamster kidney cells (BHK); mouse Sertoli cells (TM4 cells as described, e.g., in Mather, J.P., Biol.
  • MRC 5 cells MRC 5 cells
  • FS4 cells Other useful mammalian host cell lines include Chinese hamster ovary (CHO) cells, including DHFR " CHO cells (Urlaub, G., et al, Proc. Natl. Acad. Sci. USA 77 (1980) 4216-4220); and myeloma cell lines such as Y0, NS0 and Sp2/0.
  • CHO Chinese hamster ovary
  • DHFR CHO cells
  • myeloma cell lines such as Y0, NS0 and Sp2/0.
  • any of the anti-CD 19 antibodies provided herein is useful for detecting the presence of CD 19 in a biological sample.
  • the term "detecting" as used herein encompasses quantitative or qualitative detection.
  • a biological sample comprises a cell or tissue, such as tumor tissue.
  • an anti-CD 19 antibody for use in a method of diagnosis or detection is provided.
  • a method of detecting the presence of CD 19 in a biological sample comprises contacting the biological sample with an anti-CD 19 antibody as described herein under conditions permissive for binding of the anti-CD 19 antibody to CD 19, and detecting whether a complex is formed between the anti- CD 19 antibody and CD 19.
  • Such method may be an in vitro or in vivo method.
  • an anti-CD 19 antibody is used to select subjects eligible for therapy with an anti-CD 19 antibody, e.g. where CD 19 is a biomarker for selection of patients.
  • Exemplary disorders that may be diagnosed using an antibody of the invention are inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases.
  • labeled anti-CD 19 antibodies include, but are not limited to, labels or moieties that are detected directly (such as fluorescent, chromophoric, electron-dense, chemiluminescent, and radioactive labels), as well as moieties, such as enzymes or ligands, that are detected indirectly, e.g., through an enzymatic reaction or molecular interaction.
  • labels or moieties that are detected directly such as fluorescent, chromophoric, electron-dense, chemiluminescent, and radioactive labels
  • moieties such as enzymes or ligands
  • radioisotopes P, C, I, H, and I include, but are not limited to, the radioisotopes P, C, I, H, and I, fluorophores such as rare earth chelates or fluorescein and its derivatives, rhodamine and its derivatives, dansyl, umbelliferone, luceriferases, e.g., firefly luciferase and bacterial luciferase (US 4,737,456), luciferin, 2,3- dihydrophthalazinediones, horseradish peroxidase (HRP), alkaline phosphatase, ⁇ - galactosidase, glucoamylase, lysozyme, saccharide oxidases, e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase, heterocyclic oxidases such as uricase and xanthine oxid
  • compositions of an anti-CD 19 antibody as described herein are prepared by mixing such antibody having the desired degree of purity with one or more optional pharmaceutically acceptable carriers (Remington's Pharmaceutical Sciences 16 th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions.
  • Pharmaceutically acceptable carriers are generally nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyl dimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinyl pyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arg
  • sHASEGP soluble neutral-active hyaluronidase glycoproteins
  • rhuPH20 HYLENEX®, Baxter International, Inc.
  • Certain exemplary sHASEGPs and methods of use, including rhuPH20, are described in US 2005/0260186 and US 2006/0104968.
  • a sHASEGP is combined with one or more additional glycosaminoglycanases such as chondroitinases.
  • additional glycosaminoglycanases such as chondroitinases.
  • Exemplary lyophilized antibody formulations are described in US 6,267,958.
  • Aqueous antibody formulations include those described in US 6, 171 ,586 and WO 2006/044908, the latter formulations including a histidine-acetate buffer.
  • the formulation herein may also contain more than one active ingredients as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other.
  • active ingredients are suitably present in combination in amounts that are effective for the purpose intended.
  • Active ingredients may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methyl methacrylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • Sustained-release preparations may be prepared. Suitable examples of sustained- release preparations include semi permeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g. films, or microcapsules.
  • the formulations to be used for in vivo administration are generally sterile.
  • Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes.
  • anti-CD 19 antibodies Any of the anti-CD 19 antibodies provided herein may be used in therapeutic methods.
  • an anti-CD 19 antibody as reported herein for use as a medicament is provided.
  • an anti-CD 19 antibody as reported herein for use in treating inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases is provided.
  • an anti-CD 19 antibody as reported herein for use in a method of treatment is provided.
  • the invention provides an anti-CD 19 antibody for use in a method of treating an individual having inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone disease comprising administering to the individual an effective amount of the anti-CD 19 antibody as reported herein.
  • the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., as described below.
  • the invention provides an anti-CD 19 antibody for use in blocking B- cell activation without causing B-cell depletion.
  • the invention provides an anti-CD 19 antibody for use in a method of blocking B-cell activation without causing B-cell depletion in an individual comprising administering to the individual an effective of the anti-CD 19 antibody as reported herein.
  • An "individual" according to any of the above embodiments is in one embodiment a human.
  • the invention provides for the use of an anti-CD 19 antibody as reported herein in the manufacture or preparation of a medicament.
  • the medicament is for treatment of inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone disease.
  • the medicament is for use in a method of treating inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone disease comprising administering to an individual having inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone disease an effective amount of the medicament.
  • the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., as described below.
  • the medicament is for blocking B-cell activation without causing B-cell depletion.
  • the medicament is for use in a method blocking B-cell activation without causing B-cell depletion in an individual comprising administering to the individual an amount effective of the medicament.
  • An "individual" according to any of the above embodiments may be a human.
  • the invention provides a method for treating an inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone disease.
  • the method comprises administering to an individual having such inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone disease an effective amount of an anti-CD 19 antibody as reported herein.
  • the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, as described below.
  • An "individual" according to any of the above embodiments may be a human.
  • the invention provides a method for blocking B-cell activation without causing B-cell depletion in an individual.
  • the method comprises administering to the individual an effective amount of an anti-CD 19 antibody as reported herein.
  • an "individual" is a human.
  • the invention provides pharmaceutical formulations comprising any of the anti-CD19 antibodies provided herein, e.g., for use in any of the above therapeutic methods.
  • a pharmaceutical formulation comprises any of the anti-CD 19 antibodies provided herein and a pharmaceutically acceptable carrier.
  • a pharmaceutical formulation comprises any of the anti-CD 19 antibodies provided herein and at least one additional therapeutic agent, e.g., as described below.
  • Antibodies of the invention can be used either alone or in combination with other agents in a therapy.
  • an antibody of the invention may be co- administered with at least one additional therapeutic agent.
  • Such combination therapies noted above encompass combined administration (where two or more therapeutic agents are included in the same or separate formulations), and separate administration, in which case, administration of the antibody of the invention can occur prior to, simultaneously, and/or following, administration of the additional therapeutic agent and/or adjuvant.
  • Antibodies of the invention can also be used in combination with radiation therapy.
  • An antibody of the invention can be administered by any suitable means, including parenteral, intrapulmonary, and intranasal, and, if desired for local treatment, intralesional administration.
  • Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. Dosing can be by any suitable route, e.g. by injections, such as intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic.
  • Various dosing schedules including but not limited to single or multiple administrations over various time- points, bolus administration, and pulse infusion are contemplated herein.
  • Antibodies of the invention would be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • the antibody need not be, but is optionally formulated with one or more agents currently used to prevent or treat the disorder in question. The effective amount of such other agents depends on the amount of antibody present in the formulation, the type of disorder or treatment, and other factors discussed above.
  • an antibody of the invention when used alone or in combination with one or more other additional therapeutic agents, will depend on the type of disease to be treated, the type of antibody, the severity and course of the disease, whether the antibody is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody, and the discretion of the attending physician.
  • the antibody is suitably administered to the patient at one time or over a series of treatments.
  • about 1 ⁇ g/kg to 15 mg/kg (e.g. 0.1 mg/kg - 10 mg/kg) of antibody can be an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion.
  • One typical daily dosage might range from about 1 ⁇ g/kg to 100 mg/kg or more, depending on the factors mentioned above.
  • the treatment would generally be sustained until a desired suppression of disease symptoms occurs.
  • One exemplary dosage of the antibody would be in the range from about 0.05 mg/kg to about 10 mg/kg.
  • one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 4.0 mg/kg or 10 mg/kg (or any combination thereof) may be administered to the patient.
  • Such doses may be administered intermittently, e.g. every week or every three weeks (e.g. such that the patient receives from about two to about twenty, or e.g. about six doses of the antibody).
  • An initial higher loading dose, followed by one or more lower doses may be administered.
  • An exemplary dosing regimen comprises administering about 4 to 10 mg/kg.
  • other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
  • an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of the disorders described above comprises a container and a label or package insert on or associated with the container.
  • Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • At least one active agent in the composition is an antibody of the invention.
  • the label or package insert indicates that the composition is used for treating the condition of choice.
  • the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises an antibody of the invention; and (b) a second container with a composition contained therein, wherein the composition comprises a further therapeutic agent.
  • the article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the compositions can be used to treat a particular condition.
  • the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate -buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • BWFI bacteriostatic water for injection
  • phosphate -buffered saline such as bacteriostatic water for injection (BWFI), phosphate -buffered saline,
  • any of the above articles of manufacture may include an immunoconjugate of the invention in place of or in addition to an anti-CD 19 antibody.
  • SEQ ID NO: 28 Human IgGl (Afroamerican Allotype)
  • CD HB is a heavy chain variable region comprising T28A mutation in SEQ ID NO: 8; T28A means that amino acid T of position 28 of SEQ ID NO: 8 is replaced by amino acid A; etc.
  • CD HB comprises CDRHl of SEQ ID NO: 9, CDRH2 of SEQ ID NO: 10, and CDRH3 of SEQ ID NO: 11.
  • CD LE is alight chain variable region comprising P80S and F83L mutation in SEQ ID NO: 12.
  • P80S means that amino acid P of position 80 of SEQ ID NO: 12 is replaced by amino acid S; etc.
  • CD LE comprises CDRL1 of SEQ ID NO: 20, CDRL2 of SEQ ID NO: 14, and CDRL3 of SEQ ID NO: 15.
  • Mab396 and all CD HX and CD LX regions are humanized versions of Mab381 with analog nomenclature as CD HB and CD LE.
  • mice were immunized six times and boosted with CD19-transfected HEK293 cells (mean receptor density 35,000 per cell). The immune response was monitored by testing serum samples with a CD 19 - cell-ELISA on CD 19- transfected NIH-3T3 cells. Spleen cells from mice with sufficient titers of anti- CD ⁇ immunoglobulin were used for immortalization by fusion with mouse myeloma cell line P3X63 Ag8.653. Three fusions were carried out and hybridoma supernatants screened by cell-ELISA on CD19-transfected NIH-3T3 cells and FACS binding assay using Daudi (CD19 + ) and CD 19 " cells for anti-CD 19 specific antibodies.
  • the positive hybridoma 8C3, producing Mab381 was selected for the further evaluation based on the combination of the assays and properties.
  • Mab381 was selected as preferred antibody due to its biochemical and functional properties described in the following.
  • the functional properties of the antibody in biochemical and cellular assays are described in examples 2-8.
  • a property of the hybridoma 8C3 and antibody Mab381, derived from this hybridoma, as well as for the humanized version Mab396 and the other humanized light and heavy chain versions, for the selection was their inhibitory activity on activated primary B-cells.
  • Purified antibody from in-vitro production of the 8C3 hybridoma was used for characterization in different biochemical and cellular assays (see below).
  • the hybridoma 8C3 was cloned by single cell FACS sorting.
  • a cell ELISA was applied for screening of hybridomas, and to identify those hybridomas that secrete antibodies against human-CD 19.
  • NIH3T3 cells transfected with human-CD 19 were used as positive cells; untransfected NIH3T3 cells were used as negative control cells.
  • the OD ratio between transfected and untransfected NIH3T3 cells was quantified.
  • - Culture Medium DMEM high glucose (4.5 mg/ml), 10 % FCS, Na-Pyruvate, NEAA, Glutamin
  • Daudi was used. Daudi cells were incubated with different concentrations of the purified Mab381 and in a second step with a goat-anti-mouse labeled antibody. Quantification of the binding was performed by FACS analysis; the EC50 value for the binding potency was calculated.
  • Daudi Human Burkitt Lymphoma ATCC-Nr.
  • CCL-213 Human Burkitt Lymphoma
  • RPMI1640 supplemented with 10 % FCS, 2 mM L- Glutamine, 100 ⁇ NEAA, 1 mM Na-Pyruvate
  • CD 19 is a part of the B-cell co-receptor. On the cellular level stimulation of the B- cell coreceptor is possible with an anti-human-IgM antibody. The activated CD 19 can be detected with an anti-phospho-CD19 (Tyr531) antibody.
  • the Ramos cell line human B-cell lymphoma was used and the quantification of the phosphorylated CD 19 was done in the cell extracts by Western-Blot- Analysis with a Lumi-ImagerTM.
  • RAMOS ATCC CRL 1596, human, Caucasian, lymphoma, Burkitt
  • Antibody Mab381 (1 ⁇ / ⁇ 1 and 10 ⁇ / ⁇ 1) induces inhibition of anti-IgM stimulated phospho Tyr531-CD19 levels. Level of inhibition reaches non- stimulated control.
  • RAMOS ATCC CRL 1596, human, Caucasian, lymphoma, Burkitt cell line.
  • the suspensions were then incubated at 37 °C for 15 min. up to 18 hours in the continuous presence of antibody, so that the dissociation rate was negligible, and that the antigen was always saturated.
  • Other samples treated with antibody were kept at 0 °C to evaluate the level of antigen expression on non-modulated cells.
  • Mab381 shows an internalization rate of 57 % after 3 hours.
  • CD 19 as a member of the B-cell-co-receptor may play a role for proliferation of B-cells.
  • the inhibitory activity of Mab381 is determined for the proliferation of human B-cell lines with different expression levels of CD 19 and CD20 (see below). Cells were incubated with antibodies for 3 days. Viable cells after this incubation period were analyzed and calculated by FACS (FACSArrayTM BD
  • Proliferation-inhibitory activity of Mab381 was tested using primary human B- cells after stimulation with a combination of an anti-IgM-antibody and h-IL4.
  • the primary B-cells were isolated from Leucapheresis material from different healthy donors applying a negative selection process (depletion of non-B-cells); purity of the B-cells used in these experiments was >90 %.
  • the cells were incubated with antibodies for 7 days; proliferation rate was measured using a BrdU assay.
  • the antibody Mab381 shows a significant inhibition of proliferation of primary B- cells from three donors stimulated with anti-IgM and IL-4 (table 4).
  • Flex StationTM is benchtop scanning fluorometer with integrated fluid transfer workstation; the machine monitors the wells over a time course, during which compounds can be added robotically. Responses were measured as peak fluorescence intensity minus basal fluorescent intensity.
  • the antibody Mab381 inhibits Ca 2+ Influx by an EC50 of 0.37 nM (56 ng/ml, with a maximal inhibition of 50 %) and the antibody
  • Mab396 inhibits Ca 2+ Influx by an EC50 of 0.20 nM (30 ng/ml, with a maximal inhibition of 50 %).
  • the murine monoclonal antibody Mab381 expressed by a hybridoma cell line was cloned to determine the protein sequence of the antibody.
  • the isotype of the murine antibody was immunologically identified as IgGl and kappa.
  • canonical loop structures are determined by the type of residues present at the so-called canonical positions. These positions lie (partially) outside of the CDR regions, and have to be kept functionally equivalent in the final construct in order to retain the CDR conformation of the parental (donor) antibody.
  • the human germ- line sequence IGHVl-69*06 (IMGT Ac No L22583; old nomenclature: DP88) was chosen as the acceptor for the heavy chain and sequence IGKV1-9 (IMGT Acc No. Z00013) was chosen for the light chain.
  • CDR-L2 L34 A 7 aa Class 1 Determination of canonical classes of CDR loops CDR residues CDR length
  • the canonical structure class is: 1-2-2-1-1. (H1-H2-L1-L2-L3)
  • the potential acceptor sequence was selected based on high overall homology and the presence of the right canonical residues already in the acceptor sequence.
  • the humanized constructs are labeled CD LA, CD LB, CD LC, CD LD, and CD LE for the light chain, and CD HA, CD HB, CD HC, CD HD, CD HE, and CD HF for the heavy chain.
  • Binding of humanized antibodies compared to murine antibody Daudi target cells were incubated with the antibodies and binding was quantified using a fluorescently labeled secondary antibody and subsequent analysis via flow cytometry (FACS).
  • FACS flow cytometry
  • the EC50 values of the parental murine antibody and the humanized variants are essentially unchanged.
  • the different signal amplitude of the murine antibody compared to the humanized ones is due to a different secondary antibody used for detection of human or murine Fc.
  • Binding to Clq (competence for CDC)
  • the binding property of the antibody to the Clq protein of the human complement system is an indicator for its CDC potency.
  • Different variants of the humanized Mab381 (such as Mab396) produced in HEK293 or CHO cells were tested by Clq- ELISA. Result: The humanized antibody Mab396 did not show any binding activity to the human Clq protein; therefore the expectation is that it will not trigger CDC.
  • the expression system comprises the MPSV promoter system for transient expression and is described in tables 6 and 7.
  • the ADCC potency of the humanized antibody 396 was investigated.
  • the ADCC was performed using the Raji cell line and a high affinity Fcgamma-Receptor expressing cell line as effector cells.
  • the read out for killing of Raji cells was LDH release. It was found, that the antibody does not show any ADCC.
  • Mab396 (wt Fc region) and Mab396 with mutations L234A/L235A (LALA-Fc region) did not mediate significant amount of ADCC. Only by using very high concentrations of up to 300 ⁇ g/ml a moderate ADCC effect (15 %) has been observed.
  • Mab396 with low fucosylation (Mab396-g2, see WO 99/54342 and example 13) shows already ADCC mediated cell killing at lower concentrations. Results are shown in Figure 3.
  • Whole blood (B-cells) depletion assays Goal was to evaluate the depletion potency of the antibody 381 and its humanized variants in human B-cells in- vitro.
  • Whole Blood samples (after erythrocyte removal) from different healthy donors were incubated for 24 hours with the antibodies; B-cell depletion was calculated by the ratio: B-cells (CD 19 positive cells): T-cells (CD3 positive cells) with FACS analysis.
  • a human/mouse chimeric variant of Mab381 antibody, the construct with chimeric heavy chain and light chain LB, as well as light chain LE combined with heavy chain HF do not mediate significant B-cell depletion.
  • the other humanized variants do induce B-cell depletion.
  • Figures 4 and 5 show the results of a B-cell depletion experiment, where human whole blood samples were incubated for 24 hours with the indicated antibody samples (ch: chimeric; LBHch: LB and heavy chain chimeric 8C3 Mab381)).
  • B- cell depletion was measured by counting the CD 19 positive cells stained with a labeled anti-CD20 antibody in a flow cytometer (after erythrocyte removal by cell lysis) relative to the CD3 positive cells as a reference.
  • Constructs that are comprised of the heavy chain variants HA, HB, or HD show a B-cell depleting behavior
  • constructs having the chimeric heavy chain (either together with the chimeric light chain, or the humanized light chain LE), or the heavy chain variants HE, and HF show a strongly reduced activity.
  • variants HE and HF show B-cell depletion activity at the same low level as the parental antibody, or even less.
  • the humanized variant antibody 396 shows a depletion potency of human B-cells of 28 %. This depletion potency is achieved at a concentration of 5 ng/ml and remains with higher concentrations.

Abstract

The invention provides anti-CD 19 antibodies and methods of using the same. An isolated antibody that binds to CD 19, characterized in binding to the same CD 19 epitope to which monoclonal antibody 381 binds, which is characterized by comprising as heavy chain variable region SEQ ID NO: 1 and as light chain variable region SEQ ID NO: 5. The antibodies are useful as a therapeutic agent for treatment of autoimmune, rheumatoid arthritis, lupus, psoriasis, and bone diseases, as well as for tumor treatment.

Description

Antibodies against CD19 and uses thereof Field of the Invention
The present invention relates to antibodies against human CD 19 (CD 19 antibody), such antibodies that do not bind complement factor Clq, methods for their production, pharmaceutical compositions containing said antibodies, and uses thereof.
Background of the Invention
Human CD 19 is a 95 kDa transmembrane protein (B-cell coreceptor) exclusively expressed on B-cells and on follicular dendritic cells. CD 19 is found in association with CD21 and CD81. CD 19 and CD21 are required for normal B-cell differentiation (Carter, R.H., et al, Immunol. Res. 26 (2002) 45-54). Antibodies against CD19 have been used in several clinical trials (see e. g. Hekman, A., et al., Cancer Immunol. Immunother. 32 (191) 364-372; Vlasfeld, L.T., et al., Cancer Immunol. Immunother. 40 (1995) 37-47; Corny, R.M., et al., J. Immunother. Emphasis Tumor Immunol. 18 (1995) 231-241; Manzke, O., et al., Int. J. Cancer 91 (2001) 516-522).
Antibodies against CD19 are e.g. mentioned in WO 2004106381, WO 2005/012493, WO 2006/089133, WO 2007/002223, WO 2006/133450, WO 2006/121852, WO 2003/048209, US 7,109,304, US 2006/0233791, US 2006/0280738, US 2006/0263357, US 2006/0257398, EP 1648512, EP 1629012, US 2008/0138336, WO 2008/022152 and in Bruenke, J., et al, Br. J.
Hematol. 130 (2005) 218-228; Vallera, D.A., et al, Cancer Biother. Radiopharm. 19 (2004) 11-23; Ghetie, M.A., et al, Blood 104 (2004) 178-183; Lang, P., et al, Blood 103 (2004) 3982-3985; Loeffier, A., et al, Blood 95 (2000) 2098-2103; Le Gall, F., et al, FEBS Lett. 453 (1999) 164-168; Li, Q., et al, Cancer Immunol. Immunother. 47 (1998) 121-130; Eberl, G., et al, Clin. Exp. Immunol. 114 (1998)
173-178; Pietersz, G.A., et al, Cancer Immunol. Immunother. 41 (1995) 53-60; Myers, D.E., et al, Leuk Lymphoma. 18 (1995) 93-102; Bejcek, B.E., et al, Cancer Res. 55 (1995) 2346-2351; Hagen, I.A., et al, Blood.. 85 (1995) 3208-3212; Vlasfeld, L.T., et al, Cancer Immunol. Immunother. 40 (1995) 37-47; Rhodes, E.G. et al, Bone Marrow Transplant. 10 (1992) 485-489; Zola, H., et al, Immunol.
Cell Biol. 69 (1991) 411-422 ; Watanabe, M., et al, Cancer Res. 50 (1990) 3245- 3248; Uckun, F.M., et al, Blood 71 (1988) 13-29; Pezzutto, A., et al; J Immunol. 138 (1987) 2793-2799. Monoclonal antibody SJ25-C1 is commercially available (Product No. 4737, Sigma-Aldrich Co. USA, SEQ ID NO: 21 to 24). Antibodies with increased affinity to the FcyRIIIa are mentioned in WO 2008/022152.
Antibody against CD 19 can have inhibitory or stimulating effects on B-cell activation. Binding of CD 19 antibodies to mitogen-stimulated B-cells inhibits the subsequent rise in Ca2+ and the resulting activation and proliferation of these cells and B-cell proliferation and differentiation can either be inhibited or enhanced by CD 19 antibody depending on the mitogenic stimulus used and the degree of crosslinking by the antibody.
Herein are provided antibodies against CD 19 which are useful as a therapeutic agent for treatment of autoimmune, rheumatoid arthritis, lupus, psoriasis, and bone diseases and for tumor treatment.
In WO 2004/106381 pharmaceutical compositions comprising bispecific anti-CD3, anti-CD 19 antibody constructs for the treatment of B-cell related disorders are reported. Anti-CD 19 antibodies are reported in WO 2005/012493. In WO 2006/089133 anti-CD 19 antibodies and uses in oncology are reported. Anti-
CD^ antibodies and their uses are reported in WO 2007/002223. In WO 2006/133450 anti-CD19 antibody therapy for the transplantation is reported.
Summary of the invention
The invention relates to an antibody binding to human CD 19 and being characterized in binding to the same CD 19 epitope to which monoclonal antibody
381 binds. Antibody 381 is characterized by comprising as heavy chain variable region SEQ ID NO: 1 and as light chain variable region SEQ ID NO: 5. Antibody 381 comprises as CDRs a CDRHl region of SEQ ID NO: 2, a CDRH2 region of SEQ ID NO: 3 and a CDRH3 region of SEQ ID NO: 4, and a CDRLl region of SEQ ID NO: 20, a CDRL2 region of SEQ ID NO: 6 and a CDRL3 region of SEQ
ID NO: 7. As can be seen from table 1 and example 10, antibody 396 and the other mentioned antibodies, light chains, heavy chains and CDRs according to the invention are humanized variants of antibody 381 and of its light and heavy chains and CDRs. In one embodiment the antibody according to the invention is characterized in being of human IgGl isotype comprising mutations L234A, which is alanine instead of leucine at amino acid position 234, and L235A, which is alanine instead of leucine at amino acid position 235. In one embodiment the antibody according to the invention is characterized in being of human IgG4 isotype with or without mutation S228P.
The antibody according to the invention contains in one embodiment an Fc part derived from human origin.
In one embodiment the antibody according to the invention is characterized in that the heavy chain variable domain comprises as CDRs a) a CDRH1 region of SEQ ID NO: 2, a CDRH2 region of SEQ ID NO: 3 and a CDRH3 region of SEQ ID NO: 4,
b) a CDRH1 region of SEQ ID NO: 9, a CDRH2 region of SEQ ID NO: 10 and a CDRH3 region of SEQ ID NO: 1 1 ,
c) a CDRH1 region of SEQ ID NO: 9, a CDRH2 region of SEQ ID NO: 16 and a CDRH3 region of SEQ ID NO: 1 1 , or
d) a CDRH1 region of SEQ ID NO: 9, a CDRH2 region of SEQ ID NO: 17 and a CDRH3 region of SEQ ID NO: 11.
In one embodiment the antibody according to the invention is characterized in that the light chain variable domain comprises as CDRs a) a CDRLl region of SEQ ID NO: 20, a CDRL2 region of SEQ ID NO: 6 and a CDRL3 region of SEQ ID NO:7,
b) a CDRLl region of SEQ ID NO: 13, a CDRL2 region of SEQ ID NO: 14 a CDRL3 region of SEQ ID NO: 15,
c) a CDRLl region of SEQ ID NO: 13, a CDRL2 region of SEQ ID NO: 18 and a CDRL3 region of SEQ ID NO : 15 ,
d) a CDRLl region of SEQ ID NO: 19, a CDRL2 region of SEQ ID NO: 14 and a CDRL3 region of SEQ ID NO: 15, or
e) a CDRLl region of SEQ ID NO:20, a CDRL2 region of SEQ ID NO: 14 and a CDRL3 region of SEQ ID NO: 15.
In one embodiment the antibody according to the invention is characterized in that the heavy chain variable domain and the light chain variable domain comprises as CDRs a) a CDRH 1 region of SEQ ID NO: 2, a CDRLL? region of SEQ ID NO: 3 and a CDRH3 region of SEQ ID NO: 4, and a CDRLl region of SEQ ID NO: 20, a CDRL2 region of SEQ ID NO: 6 and a CDRL3 region of SEQ ID NO:7, or b) a CDRH1 region of SEQ ID NO: 9, a CDRH2 region of SEQ ID NO: 10 and a CDRH3 region of SEQ ID NO: 11, and a CDRL1 region of SEQ ID NO: 13, a CDRL2 region of SEQ ID NO: 14 a CDRL3 region of SEQ ID NO: 15.
In one embodiment the antibody according to the invention is characterized in that the antibody comprises as heavy chain variable domain a) the heavy chain variable domain of SEQ ID NO: 1 ,
b) the heavy chain variable domain of SEQ ID NO: 8,
c) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A, d) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A and as CDRH2 SEQ ID NO: 16 instead of SEQ ID NO: 10,
e) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A and as CDRH2 SEQ ID NO: 17 instead of SEQ ID NO: 10,
f) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A, and mutation VI 1L and optionally mutation K12V, or
g) the heavy chain variable domain of SEQ ID NO: 8 including mutations T28A, VI 1L, Y91F, and T108S, and optionally mutation K12V.
In one embodiment the antibody according to the invention is characterized in that the antibody comprises as light chain variable domain a) the light chain variable domain of SEQ ID NO: 5
b) the light chain variable domain of SEQ ID NO: 12,
c) the light chain variable domain of SEQ ID NO: 12 including as CDRL2 SEQ ID NO : 18 instead of SEQ ID NO : 14
d) the light chain variable domain of SEQ ID NO: 12 including as CDRLl SEQ ID NO : 19 instead of SEQ ID NO : 13 ,
e) the light chain variable domain of SEQ ID NO: 12 including as CDRLl SEQ ID NO:20 instead of SEQ ID NO: 13, or
f) the light chain variable domain of SEQ ID NO: 12 including mutations P80S and F83L and as CDRLl SEQ ID NO: 20 instead of SEQ ID NO: 13.
In one embodiment the antibody according to the invention is characterized in that the antibody comprises as heavy chain variable domain and as light chain variable domain a) the heavy chain variable domain of SEQ ID NO:l and the light chain variable domain of SEQ ID NO:5, b) the heavy chain variable domain of SEQ ID NO: 8 and the light chain variable domain of SEQ ID NO: 12,
c) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A and the light chain variable domain of SEQ ID NO: 12,
d) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A and as CDRH2 SEQ ID NO: 16 or 17, and the light chain variable domain of SEQ ID NO: 12,
e) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A and as CDRH2 SEQ ID NO: 16 or 17, and the light chain variable domain of SEQ ID NO: 12, and as CDRL1 SEQ ID NO: 19 or 20,
f) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A and as CDRH2 SEQ ID NO: 16 or 17, and the light chain variable domain of SEQ ID NO: 12, and as CDRL2 SEQ ID NO: 18, or
g) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A and as CDRH2 SEQ ID NO: 16 or 17, and the light chain variable domain of SEQ ID NO: 12 including mutations P80S and F83L, and as CDRLl SEQ ID NO: 20.
In one specific embodiment the antibody according to the invention is characterized in that the heavy chain variable domain comprises in addition to mutation T28A,
a) mutation VI 1L and optionally mutation K12V, or
b) mutations VI 1L, Y91F and T108S, and optionally mutation K12V.
In one embodiment the antibody according to the invention is characterized by the above mentioned amino acid sequences or amino acid sequence fragments and properties. The antibody according to the invention in one embodiment comprises an Fc part derived from human origin. In a specific embodiment the antibody according to the invention is of human IgGl or IgG4 isotype. The antibody according to the invention is in a specific embodiment of IgGl isotype modified in the hinge region at about amino acid position 216-240, especially at about amino acid position 220-240, between CHI and CH2 and/or in the second inter-domain region at about amino acid position 327-331 between CH2 and CH3.
In one embodiment the antibody is of human IgGl isotype comprising mutations L234A (alanine instead of leucine at amino acid position 234) and L235A. The antibody according to the invention is in one embodiment of human IgG4 isotype with or without mutation S228P.
The antibody is in one embodiment a monoclonal antibody and, in addition, a chimeric antibody (human constant chain), or a humanized antibody. In a specific embodiment the antibody is a humanized antibody.
The antibody is in one embodiment further characterized by an affinity of 10~8 M (KD) or less, especially of about 10~9 M to 10~13 M in binding to human CD 19. In one embodiment an antibody according to the invention binds also to mouse CD 19 and chimpanzee CD 19.
The invention further provides methods for the recombinant production of such antibodies.
The invention further provides a nucleic acid encoding a heavy chain variable domain of an antibody binding to human CD 19, characterized in that said nucleic acid encodes a) the heavy chain variable domain of SEQ ID NO: 1 ,
b) the heavy chain variable domain of SEQ ID NO: 8,
c) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A, d) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A and as CDRH2 SEQ ID NO: 16
e) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A and as CDRH2 SEQ ID NO: 17,
f) the heavy chain variable domain of SEQ ID NO: 8 including mutations T28A, VI 1L and optionally mutation K12V, or
g) the heavy chain variable domain of SEQ ID NO: 8 including mutations T28A, VI 1L, Y91F, and T108S, and optionally mutation K12V.
The invention further provides a nucleic acid encoding a light chain variable domain of an antibody binding to human CD 19, characterized in that said nucleic acid encodes a) the light chain variable domain of SEQ ID NO: 5
b) the light chain variable domain of SEQ ID NO: 12,
c) the light chain variable domain of SEQ ID NO: 12 including as CDRL2 SEQ ID NO : 18 instead of SEQ ID NO : 14 d) the light chain variable domain of SEQ ID NO: 12 including as CDRL1 SEQ ID NO : 19 instead of SEQ ID NO: 13, or
e) the light chain variable domain of SEQ ID NO: 12 including as CDRL1 SEQ ID NO: 20 instead of SEQ ID NO: 13.
f) the light chain variable domain of SEQ ID NO: 12 including mutations P80S and F83L and as CDRL1 SEQ ID NO: 19 and as CDRL2 SEQ ID NO: 14.
The invention further provides a nucleic acid encoding a heavy chain variable domain and a light chain domain of an antibody binding to human CD 19, characterized in that said nucleic acid encodes a) the heavy chain variable domain of SEQ ID NO:l and the light chain variable domain of SEQ ID NO:5,
b) the heavy chain variable domain of SEQ ID NO: 8 and the light chain variable domain of SEQ ID NO: 12,
c) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A and the light chain variable domain of SEQ ID NO: 12,
d) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A and as CDRH2 SEQ ID NO: 16 or 17, and the light chain variable domain of SEQ ID NO: 12,
e) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A and as CDRH2 SEQ ID NO: 16 or 17, and the light chain variable domain of SEQ ID NO: 12, and as CDRL1 SEQ ID NO: 19 or 20,
f) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A and as CDRH2 SEQ ID NO: 16 or 17, and the light chain variable domain of SEQ ID NO: 12, and as CDRL2 SEQ ID NO: 18, or
g) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A and as CDRH2 SEQ ID NO: 16 or 17, and the light chain variable domain of SEQ ID NO: 12 including mutations P80S and F83L, and as CDRLl SEQ ID NO: 20.
In one embodiment the antibody according to the invention blocks B-cell activation without causing B-cell depletion. Such an antibody is antibody 381. In a further embodiment the antibody according to the invention blocks B-cell activation and causes in addition B-cell depletion in a whole blood B-cell depletion assay. Such an antibody is antibody 396. The invention further comprises the use of an antibody according to the invention, for the treatment of inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases. The invention further comprises the use of an antibody according to the invention for the manufacture of a medicament for the treatment of diseases, in one embodiment of inflammatory, autoimmune, lupus, psoriasis, or bone diseases. The invention further comprises a method for the manufacture of a medicament for the treatment of diseases, in one embodiment of inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases, characterized in comprising an antibody according to the invention. The antibodies according to the invention have properties causing a benefit for a patient suffering from a disease associated with pathologic increase of B-cells.
Antibodies according to the invention provide for the reduction of tumor growth and a significant prolongation of the time to progression.
The antibodies according to the invention are characterized by the above mentioned properties .
The invention further provides methods for treating inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases, comprising administering to a patient diagnosed as having such disease (and therefore being in need of such a therapy) an antibody against CD 19 according to the invention. The antibody may be administered alone, in a pharmaceutical composition, or alternatively in combination with other medicaments for treating inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases. The antibody is administered in a pharmaceutically effective amount.
The invention further comprises the use of an antibody according to the invention for the treatment of inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis or bone diseases, and for the manufacture of a pharmaceutical composition comprising an antibody according to the invention. In addition, the invention comprises a method for the manufacture of a pharmaceutical composition comprising an antibody according to the invention. The invention further comprises an antibody according to the invention for the treatment of inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases. The invention further comprises a pharmaceutical composition containing an antibody according to the invention, optionally together with a buffer and/or an adjuvant useful for the formulation of antibodies for pharmaceutical purposes.
The invention further comprises a pharmaceutical composition comprising an antibody according to the invention.
The invention further provides pharmaceutical compositions comprising such antibodies in a pharmaceutically acceptable carrier. In one embodiment, the pharmaceutical composition may be included in an article of manufacture or kit. The invention further provides the use of an antibody according to the invention for the manufacture of a pharmaceutical composition for the treatment of cancer or inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases. The antibody is used in a pharmaceutically effective amount.
The invention further comprises the use of an antibody according to the invention for the manufacture of a pharmaceutical composition for the treatment of inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases.
The antibody is used in a pharmaceutically effective amount.
The invention further comprises a method for the production of a recombinant antibody according to the invention, characterized by expressing a nucleic acid encoding an antibody binding to CD 19 according to the invention in a CHO cell and recovering said antibody from said cell.
The invention further comprises a process for the production of an antibody against CD 19 according to the invention, comprising the steps of transforming a host cell, in one embodiment a CHO cell.
Description of the figures Figure 1: CD 19 target binding of anti-CD 19 antibody Fab fragments using
Daudi target cells (geometric mean vs. antibody concentration). Figure 2: CD 19 target binding of anti-CD 19 antibodies comprising constant domains using Daudi target cells (geometric mean vs. antibody concentration).
Figure 3: ADCC assay in vitro with anti-CD 19 antibody constant region variants with Raji cells. Figure 4: B-cell depletion in whole blood assay using anti-CD 19 antibodies.
Figure 5: B-cell depletion in whole blood assay using anti-CD 19 antibodies. Detailed description of embodiments of the invention I. Definitions
An "acceptor human framework" for the purposes herein is a framework comprising the amino acid sequence of a light chain variable domain (VL) framework or a heavy chain variable domain (VH) framework derived from a human immunoglobulin framework or a human consensus framework, as defined below. An acceptor human framework "derived from" a human immunoglobulin framework or a human consensus framework may comprise the same amino acid sequence thereof, or it may contain amino acid sequence changes. In some embodiments, the number of amino acid changes are 10 or less, 9 or less, 8 or less, 7 or less, 6 or less, 5 or less, 4 or less, 3 or less, or 2 or less. In some embodiments, the VL acceptor human framework is identical in sequence to the VL human immunoglobulin framework sequence or human consensus framework sequence.
"Affinity" refers to the strength of the sum total of non-covalent interactions between a single binding site of a molecule (e.g., an antibody) and its binding partner (e.g., an antigen). Unless indicated otherwise, as used herein, "binding affinity" refers to intrinsic binding affinity which reflects a 1 : 1 interaction between members of a binding pair (e.g., antibody and antigen). The affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (KD). Affinity can be measured by common methods known in the art, including those described herein. Specific illustrative and exemplary embodiments for measuring binding affinity are described in the following.
The terms "anti-CD 19 antibody" and "an antibody that binds to CD 19" refer to an antibody that is capable of binding CD 19 with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent in targeting CD 19. In one embodiment, the extent of binding of an anti-CD 19 antibody to an unrelated, non-CD 19 protein is less than about 10 % of the binding of the antibody to CD 19 as measured, by Surface Plasmon Resonance. In certain embodiments, an antibody that binds to CD 19 has a dissociation constant (KD) of 10"8 M or less, e.g. from 10"8 M to 10"13 M, e.g., from 10"9 M to 10"13 M). The term "antibody" herein is used in the broadest sense and encompasses various antibody structures, including but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they exhibit the desired antigen-binding activity. An "antibody fragment" refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds. Examples of antibody fragments include but are not limited to Fv, Fab, Fab', Fab'-SH, F(ab')2; diabodies; linear antibodies; single-chain antibody molecules (e.g. scFv); and multispecific antibodies formed from antibody fragments.
The term "without causing B-cell depletion" refers to a B-cell depletion experiment, wherein human whole blood samples (after erythrocyte removal) were incubated for 24 hours with an antibody according to the invention. B-cell depletion was measured by counting the CD 19 positive cells in a flow cytometer relative to the CD3 positive cells as a reference and with an antibody concentration of 5 ng/ml. The humanized variant antibody 396 shows a depletion potency of human B-cells of 28 % compared to antibody 381, which shows no detectable depletion. Constructs that are comprised of the heavy chain variants HA, HB, or HD show a B-cell depleting behavior, whereas constructs having the chimeric heavy chain (either together with the chimeric light chain, or the humanized light chain LE), or the heavy chain variants HE, and HF show a strongly reduced activity. In both experiments, variants HE and HF show B-cell depletion activity at the same low level as the parental antibody, or even less.
The term "blocking B-cell activation" refers to the inhibitory activity of an antibody according to the invention on the proliferation of human B-cells, measured by FACS and SU-DHL4 cells (DSMZ ACC 495) as cell lines in this assay. Blocking is found, if at an concentration of 1 μg/ml of the antibody 30 % and more inhibition of proliferation was found compared to the CD 19 negative cell line Plasmacytoma RPMI8226 (ATCC CCL 155). The term "chimeric" antibody refers to an antibody in which a portion of the heavy and/or light chain is derived from a particular source or species, while the remainder of the heavy and/or light chain is derived from a different source or species. The "class" of an antibody refers to the type of constant domain or constant region possessed by its heavy chain. There are five major classes of antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgGi, IgG2, IgG3, IgG4, IgAls and IgA2. The heavy chain constant domains that correspond to the different classes of immunoglobulins are called a, δ, ε, γ, and μ, respectively.
The antibody according to the invention is in one embodiment characterized by being of human subclass IgGl with mutations PVA236, L234A/L235A, and/or GLPSS331, or of subclass IgG4. In a further embodiment of the invention, the antibody is characterized by being of any IgG class, in one embodiment being IgGl or IgG4, containing at least one mutation in E233, L234, L235, G236, D270, N297, E318, K320, K322, A327, A330, P331 and/or P329 (numbering according to EU index). Especially suited are the IgGl mutations PVA236, L234A/L235A and/or GLPSS331 as well as the IgG4 mutation L235E. It is further in one embodiment that the antibody of IgG4 subclass contains the mutation S228P, or the mutation
S228P and L235E (Angal, S., et al, Mol. Immunol. 30 (1993) 105-108).
The antibody according to the invention therefore is in one embodiment an antibody of human subclass IgGl, containing one or more mutation(s) selected from PVA236, GLPSS331 and/or L234A/L235A (numbering according to EU index of Kabat).
The present invention refers to an antibody that binds CD 19 and does not bind complement factor Clq, and/or Fc receptor. In one embodiment of the invention, these antibodies do not elicit the complement dependent cytotoxicity (CDC) and/or antibody-dependent cellular cytotoxicity (ADCC). In one embodiment this antibody is characterized in that it binds CD 19, contains an Fc part derived from human origin and does not bind complement factor Clq. In a specific embodiment this antibody is a human or humanized monoclonal antibody. Such an antibody is useful for the treatment of inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases. "Effector functions" refer to those biological activities attributable to the Fc region of an antibody, which vary with the antibody isotype. Examples of antibody effector functions include: Clq binding and complement dependent cytotoxicity (CDC); Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g. B-cell receptor); and B-cell activation.
The antibodies according to the invention contain as Fc part, in one embodiment an Fc part derived from human origin and preferably all other parts of the human constant regions. The Fc part of an antibody is directly involved in complement activation, Clq binding, C3 activation and Fc receptor binding. While the influence of an antibody on the complement system is dependent on certain conditions, binding to Clq is caused by defined binding sites in the Fc part. Such binding sites are known in the state of the art and described e.g. by Lukas, T.J., et al, J. Immunol. 127 (1981) 2555-2560; Brunhouse, R., and Cebra, J.J., Mol. Immunol.
16 (1979) 907-917; Burton, D.R., et al, Nature 288 (1980) 338-344; Thommesen, J.E., et al, Mol. Immunol. 37 (2000) 995-1004; Idusogie, E.E., et al, J. Immunol. 164 (2000) 4178-4184; Hezareh, M., et al, J. Virol. 75 (2001) 12161-12168; Morgan, A., et al, Immunology 86 (1995) 319-324; and EP 0 307 434. Such binding sites are e.g. L234, L235, D270, N297, E318, K320, K322, P331 and P329
(numbering according to EU index of Kabat, see below). Antibodies of subclass IgGl, IgG2 and IgG3 usually show complement activation, Clq binding and C3 activation, whereas IgG4 do not activate the complement system, do not bind Clq and do not activate C3. As used herein the term "Fc part derived from human origin and does not bind human complement factor Clq and/or human Fey receptor on NK cells" denotes a Fc part which is either a Fc part of a human antibody of the subclass IgG4 or a Fc part of a human antibody of the subclass IgGl, IgG2 or IgG3 which is modified in such a way that no Clq binding, C3 activation and/or FcR binding as defined below can be detected. An "Fc part of an antibody" is a term well known to the skilled artisan and defined on the basis of papain cleavage of antibodies. In one embodiment the Fc part is a human Fc part and especially preferred either from human IgG4 subclass, in a specific embodiment mutated in the hinge region (e.g. S228P and/or L235E) or a mutated Fc part from human IgGl subclass. Mostly preferred are Fc parts comprising heavy chain constant regions selected from the regions shown in SEQ ID NO: 14 and 15 or included in SEQ ID
NO:58, 59, 60, SEQ ID NO: 14 with mutations L234A and L235A or SEQ ID NO: 15 with mutation S228P or mutations S228P and L235E.
In one embodiment of the invention an antibody according to the present invention binds CD 19 and does not bind complement factor Clq, and/or Fc receptor. In one embodiment of the invention, these antibodies do not elicit the complement dependent cytotoxicity (CDC) and/or antibody-dependent cellular cytotoxicity (ADCC). In one embodiment this antibody is characterized in that it binds CD 19, contains an Fc part derived from human origin and does not bind complement factor Clq. In a specific embodiment this antibody is a human or humanized monoclonal antibody. The effector functions mediated by the Fc part of the antibody Fc region refer to effector functions that operate after the binding of an antibody to an antigen (these functions involve the activation of the complement cascade and/or cell activation by an Fc receptor).
The function of the complement cascade can be assessed by the CH50 assay. Sheep red cells sensitized with anti-red cell antibodies (EA) are added to test serum to activate the classical pathway resulting in hemolysis. The volume of serum needed to lyse 50 % of the red cells determines the CH50 unit. The AP-CH50 measures the alternative and the terminal pathways. The procedure is similar except that rabbit red cells are used. The alternative pathway is activated upon addition of test serum. Clq and two serine proteases, Clr and Cls, form the complex CI, the first component of the complement dependent cytotoxicity (CDC) pathway. To activate the complement cascade Clq binds to at least two molecules of IgGl or one molecule of IgM, attached to the antigenic target (Ward, E.S. and Ghetie, V., Ther. Immunol. 2 (1995) 77-94). Burton, D.R., (Mol. Immunol. 22 (1985) 161-206) described that the heavy chain region comprising amino acid residues 318 to 337 is being involved in complement fixation. Duncan, A.R. and Winter, G. (Nature 332 (1988) 738-740) reported using site directed mutagenesis that Glu318, Lys320 and Lys322 form the binding site to Clq. The role of Glu318, Lys320 and Lys 322 residues in the binding of Clq was confirmed by the ability of a short synthetic peptide containing these residues to inhibit complement mediated lysis.
The term "complement-dependent cytotoxicity (CDC)" refers to lysis of CD 19 expressing human endothelial cells by the antibody according to the invention in the presence of complement. CDC is measured in one embodiment by the treatment of CD 19 expressing human endothelial cells with an antibody according to the invention in the presence of complement. The cells are in one embodiment labeled with calcein. CDC is found if the antibody induces lysis of 20 % or more of the target cells at a concentration of 30 μg/ml. Binding to the complement factor Clq can be measured in an ELISA assay. In such an assay in principle an ELISA plate is coated with concentration ranges of the antibody, to which purified human Clq or human serum is added. Clq binding is detected by an antibody directed against Clq followed by a peroxidase-labeled conjugate. Detection of binding (maximal binding Bmax) is measured as optical density at 405 nm (OD405) for peroxidase substrate ABTS® (2,2' -azino-di-[3-ethylbenzthiazoline-6-sulfonate (6)]). Accordingly the present invention in one embodiment refers to an antibody, characterized in that non-binding of the antibody to complement factor Clq refers to such an ELISA assay measurement wherein the maximal binding (Bmax) of Clq to an antibody according to the invention at a concentration of 10 μg/ml of the antibody is 20 % or lower of Bmax observed with antibody 381 comprising human IgGl constant region of SEQ ID NO: 26 and SEQ ID NO: 27, in one embodiment
10 % or lower.
In one embodiment an antibody according to the invention shows a reduced activation of complement factor C3 in an ELISA assay. The assay is performed in the same manner as the Clq assay. In such an assay in principle an ELISA plate is coated with concentration ranges of the antibody, to which human serum is added.
C3 binding is detected by an antibody directed against C3 followed by a peroxidase-labeled conjugate. Detection of binding (maximal binding Bmax) is measured as optical density at 405 nm (OD405) for peroxidase substrate ABTS®. Accordingly the present invention refers in one embodiment to an antibody, characterized in that non-binding of the antibody to complement factor C3 refers to such an ELISA assay measurement wherein the maximal binding (Bmax) of C3 to the antibody at a concentration of 10 μg/ml of the antibody is 10 % of Bmax of antibody 381 comprising human IgGl constant region of SEQ ID NO: 26 and SEQ ID NO: 27, in one embodiment 5 % or lower. The term "antibody-dependent cellular cytotoxicity (ADCC)" is a function mediated by Fc receptor binding and refers to lysis of CD 19 expressing target cells by an antibody according to the invention in the presence of effector cells. ADCC is measured in one embodiment by the treatment of a preparation of CD 19 expressing erythroid cells (e.g. K562 cells expressing recombinant human CD 19) with an antibody according to the invention in the presence of effector cells such as freshly isolated PBMC (peripheral blood mononuclear cells) or purified effector cells from buffy coats, like monocytes or NK (natural killer) cells. Target cells are labeled with 51Cr and subsequently incubated with the antibodies. The labeled cells are incubated with effector cells and the supernatant is analyzed for released 51Cr. Controls include the incubation of the target endothelial cells with effector cells but without the antibody. The capacity of the antibodies to induce the initial steps mediating ADCC was investigated by measuring their binding to Fey receptors expressing cells, such as cells, recombinantly expressing FcyRI and/or FcyRIIA or NK cells (expressing essentially FcyRIIIA). Preferably binding to FcyR on NK cells is measured. Fc receptor binding effector functions can be mediated by the interaction of the Fc region of an antibody with Fc receptors (FcRs), which are specialized cell surface receptors on hematopoietic cells. Fc receptors belong to the immunoglobulin superfamily, and have been shown to mediate both the removal of antibody-coated pathogens by phagocytosis of immune complexes, and the lysis of erythrocytes and various other cellular targets (e.g. tumor cells) coated with the corresponding antibody, via antibody dependent cell mediated cytotoxicity (ADCC) (Van de Winkel, J.G. and Anderson, C.L., J. Leukoc. Biol. 49 (1991) 511-524). FcRs are defined by their specificity for immunoglobulin isotypes; Fc receptors for IgG antibodies are referred to as FcyR, for IgE as FcyR, for IgA as FcyR and so on. Fc receptor binding is described e.g. in Ravetch, J.V., and Kinet, J.P., Annu. Rev.
Immunol. 9 (1991) 457-492; Capel, P.J., et al, Immunomethods 4 (1994) 25-34; de Haas, M., et al, J. Lab. Clin. Med. 126 (1995) 330-341; and Gessner, J.E., et al, Ann. Hematol. 76 (1998) 231-248.
Cross-linking of receptors for the Fc domain of IgG antibodies (FcyR) triggers a wide variety of effector functions including phagocytosis, antibody-dependent cellular cytotoxicity, and release of inflammatory mediators, as well as immune complex clearance and regulation of antibody production. In humans, three classes of FcyR have been characterized, which are:
- FcyRI (CD64) binds monomeric IgG with high affinity and is expressed on macrophages, monocytes, neutrophils and eosinophils. Modification in IgG of at least one of E233-G236, P238, D265, N297, A327 and P329 reduce binding to FcyRI. IgG2 residues at positions 233-236, substituted into IgGl and IgG4, reduced binding to FcyRI by 103-fold and eliminated the human monocyte response to antibody-sensitized red blood cells (Armour, K.L., et al, Eur. J. Immunol. 29 (1999) 2613-2624).
- FcyRII (CD32) binds complexed IgG with medium to low affinity and is widely expressed. These receptors can be divided into two important types, FcyRIIA and FcyRIIB. FcyRIIA is found on many cells involved in killing (e.g. macrophages, monocytes, neutrophils) and seems able to activate the killing process. FcyRIIB seems to play a role in inhibitory processes and is found on B-cells, macrophages and on mast cells and eosinophils. On B-cells it seems to function to suppress further immunoglobulin production and isotype switching to, for example, the IgE class. On macrophages, FcyRIIB acts to inhibit phagocytosis as mediated through FcyRIIA. On eosinophils and mast cells the
B-form may help to suppress activation of these cells through IgE binding to its separate receptor. Reduced binding for FcyRIIA is found e.g. for IgG mutation of at least one of E233-G236, P238, D265, N297, A327, P329, D270, Q295, A327, R292, and K414. - FcyRIII (CD 16) binds IgG with medium to low affinity and exists as two types.
FcyRIIIA is found on NK cells, macrophages, eosinophils and some monocytes and T cells and mediates ADCC. FcyRIIIB is highly expressed on neutrophils. Reduced binding to FcyRIIIA is found e.g. for mutation of at least one of E233- G236, P238, D265, N297, A327, P329, D270, Q295, A327, S239, E269, E293, Y296, V303, A327, K338 and D376.
Mapping of the binding sites on human IgGl for Fc receptors, the above mentioned mutation sites and methods for measuring binding to FcyRI and FcyRIIA are described in Shields, R.L., et al. J. Biol. Chem. 276 (2001) 6591-6604.
The term "Fc receptor" when used herein refers to activation receptors characterized by the presence of a cytoplasmatic ITAM sequence associated with the receptor (see e.g. Ravetch, J.V., and Bolland, S., Annu. Rev. Immunol. 19 (2001) 275-290). Such receptors are FcyRI, FcyRIIA and FcyRIIIA. The antibodies according to the invention in one embodiment show a reduced binding to Fey receptors, especially to FcylllA. In one embodiment the term "no binding of FcyR" means that in an antibody concentration of 10 μg/ml the binding of an antibody according to the invention to NK cells is 10 % or less of the binding found for anti- OX40L antibody LC.001 as reported in WO 2006/029879.
While IgG4 shows reduced FcR binding, antibodies of other IgG subclasses show strong binding. However Pro238, Asp265, Asp270, Asn297 (loss of Fc carbohydrate), Pro329 and 234, 235, 236 and 237 Ile253, Ser254, Lys288 , Thr307,
Gln311, Asn434, and His435 are residues which provide if altered also reduce FcR binding (Shields, R.L., et al. J. Biol. Chem. 276 (2001) 6591-6604; Lund, J., et al, FASEB J. 9 (1995) 115-119; Morgan, A., et al, Immunology 86 (1995) 319-324; and EP 0 307 434). In one embodiment an antibody according to the invention is of IgGl or IgG2 subclass and comprises mutation PVA236, GLPSS331, and/or L234A/L235A. An antibody according to the invention of IgG4 subclass comprises in one embodiment mutation L235E. In one embodiment further specific IgG4 mutations are S228P or L235E and S228P. Cell-mediated effector functions of monoclonal antibodies can be enhanced by engineering their oligosaccharide component as described in Umana, P., et al., Nature Biotechnol. 17 (1999) 176-180; and US 6,602,684. IgGl type antibodies, the most commonly used antibodies in cancer immunotherapy, are glycoproteins that have a conserved N-linked glycosylation site at Asn297 in each CH2 domain. The two complex biantennary oligosaccharides attached to Asn297 are buried between the CH2 domains, forming extensive contacts with the polypeptide backbone, and their presence is essential for the antibody to mediate effector functions such as antibody dependent cellular cytotoxicity (ADCC) (Lifely, M.R., et al, Glycobiology 5 (1995) 813-822; Jefferis, R., et al, Immunol. Rev. 163 (1998) 59-76; Wright, A. and Morrison, S.L., Trends Biotechnol. 15 (1997) 26-32).
Umana, P., et al, Nature Biotechnol. 17 (1999) 176-180 and WO 99/54342 showed that overexpression in Chinese hamster ovary (CHO) cells of B(l,4)-N- acetylglucosaminyltransferase III ("GnTIII"), a glycosyltransferase catalyzing the formation of bisected oligosaccharides, significantly increases the in vitro ADCC activity of antibodies. Alterations in the composition of the N297 carbohydrate or its elimination affect also binding to Fc binding to FcyR and CI q (Umana, P., et al, Nature Biotechnol. 17 (1999) 176-180; Davies, J., et al, Biotechnol. Bioeng. 74 (2001) 288-294; Mimura, Y., et al, J. Biol. Chem. 276 (2001) 45539-45547; Radaev, S., et al, J. Biol. Chem. 276 (2001) 16478-16483; Shields, R.L., et al, J. Biol. Chem. 276 (2001) 6591-6604; Shields, R.L., et al, J. Biol. Chem. 277 (2002) 26733-26740; Simmons, L.C., et al, J. Immunol. Methods 263 (2002) 133-147).
Glycosylation of human IgGl or IgG3 occurs at Asn297 as core fucosylated biantennary complex oligosaccharide glycosylation terminated with up to 2 Gal residues. These structures are designated as GO, Gl (a 1,6 or a 1,3) or G2 glycan residues, depending from the amount of terminal Gal residues (Raju, T.S., Bioprocess Int. 1 (2003) 44-53). CHO type glycosylation of antibody Fc parts is e.g. described by Routier, F.H., Glycoconjugate J. 14 (1997) 201-207.
The "partially- or non- fucosylated anti-CD 19 antibody" according to the invention can be expressed in a glycomodified host cell engineered to express at least one nucleic acid encoding a polypeptide having GnTIII activity in an amount sufficient to partially fucosylate the oligosaccharides in the Fc region. In one embodiment, the polypeptide having GnTIII activity is a fusion polypeptide. Alternatively al,6- fucosyltransferase activity of the host cell can be decreased or eliminated according to US 6,946,292 to generate glycomodified host cells. The amount of antibody fucosylation can be predetermined e.g. either by fermentation conditions (e.g. fermentation time), or by combination of at least two antibodies with different fucosylation amount. Measurement of the fucosylation is performed by Liquid chromatography-mass spectrometry (LC-MS). The term "a partially- or non- fucosylated anti-CD 19 antibody" according to the invention when used herein refers to an anti-CD 19 antibody showing a fucosylation at Asn297 of 50 % or lower compared with antibody 381 comprising human IgGl constant region of SEQ ID NO:26 and SEQ ID NO:27. Such an antibody is useful for the treatment of cancer diseases.
The partially- or non-fucosylated anti-CD 19 antibody according to the invention can be produced in a host cell by a method comprising: (a) culturing a host cell engineered to express at least one polynucleotide encoding a fusion polypeptide having GnTIII activity under conditions which permit the production of said antibody with partial fucosylation of the oligosaccharides present on the Fc region of said antibody; and (b) isolating said antibody. In one embodiment, the polypeptide having GnTIII activity is a fusion polypeptide, especially comprising the catalytic domain of GnTIII and the Golgi localization domain of a heterologous Golgi resident polypeptide selected from the group consisting of the localization domain of mannosidase II, the localization domain of B(l,2)-N- acetylglucosaminyltransferase I ("GnTI"), the localization domain of mannosidase I, the localization domain of B(l,2)-N-acetylglucosaminyltransferase II ("GnTII"), and the localization domain of al-6 core fucosyltransferase. In one embodiment the Golgi localization domain is from mannosidase II or GnTI. In a further aspect, the invention is directed to a method for modifying the glycosylation profile of an anti- CD^ antibody by using such method. An "effective amount" of an agent, e.g., a pharmaceutical formulation, refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result.
The term "Fc region" herein is used to define a C-terminal region of an immunoglobulin heavy chain that contains at least a portion of the constant region. The term includes native sequence Fc regions and variant Fc regions. In one embodiment, a human IgG heavy chain Fc region extends from Cys226, or from Pro230, to the carboxyl-terminus of the heavy chain. However, the C-terminal lysine (Lys447) of the Fc region may or may not be present. Unless otherwise specified herein, numbering of amino acid residues in the Fc region or constant region is according to the EU numbering system, also called the EU index, as described in Kabat, E.A., et al, Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, MD (1991).
"Framework" or "FR" refers to variable domain residues other than hypervariable region (HVR) residues. The FR of a variable domain generally consists of four FR domains: FR1, FR2, FR3, and FR4. Accordingly, the CDR and FR sequences generally appear in the following sequence in VH (or VL): FR1-CDR1(L1)-FR2- CDR2(L2)-FR3-CDR3(L3)-FR4.
The terms "full length antibody," "intact antibody," and "whole antibody" are used herein interchangeably to refer to an antibody having a structure substantially similar to a native antibody structure or having heavy chains that contain an Fc region as defined herein.
The terms "host cell," "host cell line," and "host cell culture" are used interchangeably and refer to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells. Host cells include "transformants" and "transformed cells," which include the primary transformed cell and progeny derived therefrom without regard to the number of passages. Progeny may not be completely identical in nucleic acid content to a parent cell, but may contain mutations. Mutant progeny that have the same function or biological activity as screened or selected for in the originally transformed cell are included herein. A "human consensus framework" is a framework which represents the most commonly occurring amino acid residues in a selection of human immunoglobulin VL or VH framework sequences. Generally, the selection of human immunoglobulin VL or VH sequences is from a subgroup of variable domain sequences. Generally, the subgroup of sequences is a subgroup as in Kabat, E.A., et al., Sequences of Proteins of Immunological Interest, fifth edition, NIH Publication
91-3242, Bethesda, MD (1991), Vol. 1-3. In one embodiment, for the VL, the subgroup is subgroup kappa I as in Kabat, E.A., et al, supra. In one embodiment, for the VH, the subgroup is subgroup III as in Kabat, E.A., et al, supra. A "humanized" antibody refers to a chimeric antibody comprising amino acid residues from non-human CDRs and amino acid residues from human FRs. In certain embodiments, a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDRs correspond to those of a non-human antibody, and all or substantially all of the FRs correspond to those of a human antibody. A humanized antibody optionally may comprise at least a portion of an antibody constant region derived from a human antibody. A "humanized form" of an antibody, e.g., a non-human antibody, refers to an antibody that has undergone humanization. The term "complementarity determining regions" or "CDR," as used herein refers to each of the regions of an antibody variable domain which are hypervariable in sequence and/or form structurally defined loops ("hypervariable loops) (Kabat, E.A., et al, Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, MD (1991).) With the exception of CDR1 in VH, CDRs generally comprise the amino acid residues that form the hypervariable loops. Unless otherwise indicated, CDR residues and other residues in the variable domain (e.g., FR residues) are numbered herein according to Kabat, E.A., et al, supra.
An "immunoconjugate" is an antibody conjugated to one or more heterologous molecule(s).
An "individual" or "subject" is a mammal. Mammals include, but are not limited to, domesticated animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats). In certain embodiments, the individual or subject is a human. An "isolated" antibody is one which has been separated from a component of its natural environment. In some embodiments, an antibody is purified to greater than 95 % or 99 % purity as determined by, for example, electrophoretic (e.g., SDS- PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatographic (e.g., ion exchange or reverse phase HPLC). For review of methods for assessment of antibody purity, see, e.g., Flatman, S., et al, J. Chromatogr. B 848 (2007) 79-87.
An "isolated" nucleic acid refers to a nucleic acid molecule that has been separated from a component of its natural environment. An isolated nucleic acid includes a nucleic acid molecule contained in cells that ordinarily contain the nucleic acid molecule, but the nucleic acid molecule is present extrachromosomally or at a chromosomal location that is different from its natural chromosomal location.
"Isolated nucleic acid encoding an anti-CD 19 antibody" refers to one or more nucleic acid molecules encoding antibody heavy and light chains (or fragments thereof), including such nucleic acid molecule(s) in a single vector or separate vectors, and such nucleic acid molecule(s) present at one or more locations in a host cell.
The term "monoclonal antibody" as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical and/or bind the same epitope, except for possible variant antibodies, e.g., containing naturally occurring mutations or arising during production of a monoclonal antibody preparation, such variants generally being present in minor amounts. In contrast to polyclonal antibody preparations, which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody of a monoclonal antibody preparation is directed against a single determinant on an antigen. Thus, the modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the present invention may be made by a variety of techniques, including but not limited to the hybridoma method, recombinant DNA methods, phage-display methods, and methods utilizing transgenic animals containing all or part of the human immunoglobulin loci, such methods and other exemplary methods for making monoclonal antibodies being described herein.
A "naked antibody" refers to an antibody that is not conjugated to a heterologous moiety. The naked antibody may be present in a pharmaceutical formulation.
"Native antibodies" refer to naturally occurring immunoglobulin molecules with varying structures. For example, native IgG antibodies are heterotetrameric glycoproteins of about 150,000 Daltons, composed of two identical light chains and two identical heavy chains that are disulfide-bonded. From N- to C-terminus, each heavy chain has a variable region (VH), also called a variable heavy domain or a heavy chain variable domain, followed by three constant domains (CHI, CH2, and CH3). Similarly, from N- to C-terminus, each light chain has a variable region (VL), also called a variable light domain or a light chain variable domain, followed by a constant light (CL) domain. The light chain of an antibody may be assigned to one of two types, called kappa (κ) and lambda (λ), based on the amino acid sequence of its constant domain. The term "package insert" is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, combination therapy, contraindications and/or warnings concerning the use of such therapeutic products.
"Percent (%) amino acid sequence identity" with respect to a reference polypeptide sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the reference polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. For purposes herein, however, % amino acid sequence identity values are generated using the sequence comparison computer program ALIGN-2. The ALIGN -2 sequence comparison computer program was authored by Genentech, Inc., and the source code has been filed with user documentation in the U.S. Copyright Office, Washington D.C., 20559, where it is registered under U.S. Copyright Registration
No. TXU510087. The ALIGN-2 program is publicly available from Genentech, Inc., South San Francisco, California, or may be compiled from the source code. The ALIGN-2 program should be compiled for use on a UNIX operating system, including digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary.
In situations where ALIGN-2 is employed for amino acid sequence comparisons, the % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given amino acid sequence A that has or comprises a certain % amino acid sequence identity to, with, or against a given amino acid sequence B) is calculated as follows:
100 times the fraction X/Y where X is the number of amino acid residues scored as identical matches by the sequence alignment program ALIGN-2 in that program's alignment of A and B, and where Y is the total number of amino acid residues in B. It will be appreciated that where the length of amino acid sequence A is not equal to the length of amino acid sequence B, the % amino acid sequence identity of A to B will not equal the % amino acid sequence identity of B to A. Unless specifically stated otherwise, all % amino acid sequence identity values used herein are obtained as described in the immediately preceding paragraph using the ALIGN-2 computer program.
The term "pharmaceutical formulation" refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
A "pharmaceutically acceptable carrier" refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject., a pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
The term "CD 19," as used herein, refers to human B-lymphocyte antigen CD 19 (alternative name(s) are: Differentiation antigen CD 19, B-lymphocyte surface antigen B4, T-cell surface antigen Leu-12; UniProtKB P15391).
As used herein, "treatment" (and grammatical variations thereof such as "treat" or "treating") refers to clinical intervention in an attempt to alter the natural course of the individual being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include, but are not limited to, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, decreasing the rate of disease progression, amelioration or palliation of the disease state. In some embodiments, antibodies of the invention are used to delay development of a disease or to slow the progression of a disease. The term "variable region" or "variable domain" refers to the domain of an antibody heavy or light chain that is involved in binding the antibody to antigen. The variable domains of the heavy chain and light chain (VH and VL, respectively) of a native antibody generally have similar structures, with each domain comprising four conserved framework regions (FRs) and three hypervariable regions (HVRs). (See, e.g., Kindt, et al, Kuby Immunology, 6th ed., W.H. Freeman and Co., page 91 (2007)). A single VH or VL domain may be sufficient to confer antigen-binding specificity. Furthermore, antibodies that bind a particular antigen may be isolated using a VH or VL domain from an antibody that binds the antigen to screen a library of complementary VL or VH domains, respectively. See, e.g., Potolano, S., et al, J. Immunol. 150 (1993) 880-887; Clackson, T., et al, Nature 352 (1991) 624-628.
The term "vector," as used herein, refers to a nucleic acid molecule capable of propagating another nucleic acid to which it is linked. The term includes the vector as a self-replicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced. Certain vectors are capable of directing the expression of nucleic acids to which they are operatively linked. Such vectors are referred to herein as "expression vectors."
II. Compositions and methods In one aspect, the invention is based on antibodies that bind to CD 19. Antibodies of the invention are useful, e.g., for the diagnosis or treatment of inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases.
A. Exemplary Anti-CD 19 Antibodies
In a further aspect of the invention, an anti-CD 19 antibody according to any of the above embodiments is a monoclonal antibody, including a chimeric or humanized antibody. In one embodiment, an anti-CD19 antibody is an antibody fragment, e.g., an Fv, Fab, Fab', scFv, diabody, or F(ab')2 fragment. In another embodiment, the antibody is a full length antibody, e.g., an intact IgGl antibody or other antibody class or isotype as defined herein. In a further aspect, an anti-CD 19 antibody according to any of the above embodiments may incorporate any of the features, singly or in combination, as described in the sections below: 1. Antibody Affinity
In certain embodiments, an antibody provided herein has a dissociation constant (KD) of 10"8 M or less, e.g. from 10"8 M to 10"13 M, e.g., from 10"9 M to 10"13 M.
According to another embodiment, KD is measured using surface plasmon resonance assays using a BIACORE®-2000 or a BIACORE ®-3000 (BIAcore, Inc.,
Piscataway, NJ) at 25 °C with immobilized antigen CM5 chips at ~10 response units (RU). Briefly, carboxymethylated dextran biosensor chips (CM5, BIACORE, Inc.) are activated with N-ethyl-N'-(3-dimethylaminopropyl)-carbodiimide hydrochloride (EDC) and N-hydroxysuccinimide (NHS) according to the supplier's instructions. Antigen is diluted with 10 mM sodium acetate, pH 4.8, to 5 μg/ml
(-0.2 μΜ) before injection at a flow rate of 5 μΐ/minute to achieve approximately 10 response units (RU) of coupled protein. Following the injection of antigen, 1 M ethanolamine is injected to block unreacted groups. For kinetics measurements, two-fold serial dilutions of Fab (0.78 nM to 500 nM) are injected in PBS with 0.05 % polysorbate 20 (TWEEN-20™) surfactant (PBST) at 25 °C at a flow rate of approximately 25 μΐ/min. Association rates (kon) and dissociation rates (k0ff) are calculated using a simple one-to-one Langmuir binding model (BIACORE Evaluation Software version 3.2) by simultaneously fitting the association and dissociation sensorgrams. The equilibrium dissociation constant (KD) is calculated as the ratio koff/kon. See, e.g., Chen, Y., et al, J. Mol. Biol. 293 (1999) 865-881. If the on-rate exceeds 10^ M~l s~l by the surface plasmon resonance assay above, then the on-rate can be determined by using a fluorescent quenching technique that measures the increase or decrease in fluorescence emission intensity (excitation = 295 nm; emission = 340 nm, 16 nm band-pass) at 25 °C of a 20 nM anti-antigen antibody (Fab form) in PBS, pH 7.2, in the presence of increasing concentrations of antigen as measured in a spectrometer, such as a stop-flow equipped spectrophotometer (Aviv Instruments) or a 8000-series SLM-AMINCO ™ spectrophotometer (ThermoSpectronic) with a stirred cuvette.
2. Antibody Fragments In certain embodiments, an antibody provided herein is an antibody fragment.
Antibody fragments include, but are not limited to, Fab, Fab', Fab'-SH, F(ab')2, Fv, and scFv fragments, and other fragments described below. For a review of certain antibody fragments, see Hudson, et al., Nat. Med. 9 (2003) 129-134. For a review of scFv fragments, see, e.g., Pluckthun, in The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., (Springer-Verlag, New York), pp. 269-315 (1994); see also WO 93/16185; and US 5,571,894 and US 5,587,458. For discussion of Fab and F(ab')2 fragments comprising salvage receptor binding epitope residues and having increased in vivo half-life, see US 5,869,046.
Diabodies are antibody fragments with two antigen-binding sites that may be bivalent or bispecific. See, for example, EP 404,097; WO 1993/01161; Hudson, et al, Nat. Med. 9 (2003) 129-134; and Holliger, P., et al, Proc. Natl. Acad. Sci. USA 90 (1993) 6444-6448. Triabodies and tetrabodies are also described in Hudson, et al, Nat. Med. 9 (2003) 129-134.
Single-domain antibodies are antibody fragments comprising all or a portion of the heavy chain variable domain or all or a portion of the light chain variable domain of an antibody. In certain embodiments, a single-domain antibody is a human single-domain antibody (Domantis, Inc., Waltham, MA; see, e.g., US 6,248,516 Bl).
Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as production by recombinant host cells (e.g. E. coli or phage), as described herein.
3. Multispecific Antibodies In certain embodiments, an antibody provided herein is a multispecific antibody, e.g. a bispecific antibody. Multispecific antibodies are monoclonal antibodies that have binding specificities for at least two different sites. In certain embodiments, one of the binding specificities is for CD 19 and the other is for any other antigen. In certain embodiments, bispecific antibodies may bind to two different epitopes of CD 19. Bispecific antibodies can be prepared as full length antibodies or antibody fragments.
Techniques for making multispecific antibodies include, but are not limited to, recombinant co-expression of two immunoglobulin heavy chain-light chain pairs having different specificities (see Milstein, C, and Cuello, Nature 305 (1983) 537- 540), WO 93/08829, and Traunecker, A., et al, EMBO J. 10 (1991) 3655-3659), and "knob-in-hole" engineering (see, e.g., US 5,731,168). Multi-specific antibodies may also be made by engineering electrostatic steering effects for making antibody Fc-heterodimeric molecules (WO 2009/089004A1); cross-linking two or more antibodies or fragments (see, e.g., US 4,676,980, and Brennan, M., et al, Science, 229 (1985) 81-83); using leucine zippers to produce bi-specific antibodies (see, e.g., Kostelny, S.A., et al, J. Immunol, 148(5) (1992) 1547-1553); using "diabody" technology for making bispecific antibody fragments (see, e.g., Hollinger, et al, Proc. Natl. Acad. Sci. USA, 90 (1993) 6444-6448); and using single-chain Fv (sFv) dimers (see, e.g. Gruber, M., et al, J. Immunol, 152 (1994) 5368-5374) and preparing trispecific antibodies as described, e.g., in Tutt, A., et al. J. Immunol. 147 (1991) 60-69.
Engineered antibodies with three or more functional antigen binding sites, including "Octopus antibodies," are also included herein (see, e.g. US 2006/0025576A1).
The antibody or fragment herein also includes a "Dual Acting FAb" or "DAF" comprising an antigen binding site that binds to CD 19 as well as another, different antigen (see, US 2008/0069820, for example). The antibody or fragment herein also includes multispecific antibodies described in
WO 2009/080251, WO 2009/080252, WO 2009/080253, WO 2009/080254, WO 2010/112193, WO 2010/115589, WO 2010/136172, WO 2010/145792, and WO 2010/145793.
4. Antibody Variants a) Glycosylation variants
In certain embodiments, an antibody provided herein is altered to increase or decrease the extent to which the antibody is glycosylated. Addition or deletion of glycosylation sites to an antibody may be conveniently accomplished by altering the amino acid sequence such that one or more glycosylation sites is created or removed.
Where the antibody comprises an Fc region, the carbohydrate attached thereto may be altered. Native antibodies produced by mammalian cells typically comprise a branched, biantennary oligosaccharide that is generally attached by an N-linkage to Asn297 of the CH2 domain of the Fc region. See, e.g., Wright, et al, TIBTECH 15 (1997) 26-32. The oligosaccharide may include various carbohydrates, e.g., mannose, N-acetyl glucosamine (GlcNAc), galactose, and sialic acid, as well as a fucose attached to a GlcNAc in the "stem" of the biantennary oligosaccharide structure. In some embodiments, modifications of the oligosaccharide in an antibody of the invention may be made in order to create antibody variants with certain improved properties.
The amount of fucose is determined by calculating the average amount of fucose within the sugar chain at Asn297, relative to the sum of all glycostructures attached to Asn 297 (e.g. complex, hybrid and high mannose structures) as measured by
MALDI-TOF mass spectrometry, as described in WO 2008/077546, for example.
Asn297 refers to the asparagine residue located at about position 297 in the Fc region (Eu numbering of Fc region residues); however, Asn297 may also be located about ± 3 amino acids upstream or downstream of position 297, i.e., between positions 294 and 300, due to minor sequence variations in antibodies.
Such fucosylation variants may have improved ADCC function. See, e.g.,
US 2003/0157108 (Presta, L.); US 2004/0093621 (Kyowa Hakko Kogyo Co., Ltd).
Examples of publications related to "defucosylated" or "fucose-deficient" antibody variants include: US 2003/0157108; WO 2000/61739; WO 2001/29246;
US 2003/0115614; US 2002/0164328; US 2004/0093621; US 2004/0132140;
US 2004/0110704; US 2004/0110282; US 2004/0109865; WO 2003/085119;
WO 2003/084570; WO 2005/035586; WO 2005/035778; WO 2005/053742;
WO 2002/031140; Okazaki,. et al, J. Mol. Biol. 336 (2004) 1239-1249; Yamane- Ohnuki, N., et al, Biotech. Bioeng. 87 (2004) 614-622). Examples of cell lines capable of producing defucosylated antibodies include Led 3 CHO cells deficient in protein fucosylation (Ripka, J., et al, Arch. Biochem. Biophys. 249 (1986) 533-
545, US 2003/0157108 Al, Presta, L., and WO 2004/056312 Al, Adams, et al, especially at Example 11), and knockout cell lines, such as alpha- 1,6- fucosyltransferase gene, FUT8, knockout CHO cells (see, e.g., Yamane-Ohnuki,
N., et al. Biotech. Bioeng. 87 (2004) 614-622; Kanda, Y., et al, Biotechnol.
Bioeng. 94(4) (2006) 680-688; and WO 2003/085107).
Antibodies variants are further provided with bisected oligosaccharides, e.g., in which a biantennary oligosaccharide attached to the Fc region of the antibody is bisected by GlcNAc. Such antibody variants may have reduced fucosylation and/or improved ADCC function. Examples of such antibody variants are described, e.g., in WO 2003/011878, US 6,602,684, and US 2005/0123546. Antibody variants with at least one galactose residue in the oligosaccharide attached to the Fc region are also provided. Such antibody variants may have improved CDC function. Such antibody variants are described, e.g., in WO 1997/30087, WO 1998/58964, and
WO 1999/22764. b) Fc region variants
In certain embodiments, one or more amino acid modifications may be introduced into the Fc region of an antibody provided herein, thereby generating an Fc region variant. The Fc region variant may comprise a human Fc region sequence (e.g., a human IgGl , IgG2, IgG3 or IgG4 Fc region) comprising an amino acid modification (e.g. a substitution) at one or more amino acid positions.
In certain embodiments, the invention contemplates an antibody variant that possesses some but not all effector functions, which make it a desirable candidate for applications in which the half life of the antibody in vivo is important yet certain effector functions (such as complement and ADCC) are unnecessary or deleterious. In vitro and/or in vivo cytotoxicity assays can be conducted to confirm the reduction/depletion of CDC and/or ADCC activities. For example, Fc receptor (FcR) binding assays can be conducted to ensure that the antibody lacks FcyR binding (hence likely lacking ADCC activity), but retains FcRn binding ability. The primary cells for mediating ADCC, NK cells, express FcyRIII only, whereas monocytes express FcyRI, FcyRII and FcyRIII. FcR expression on hematopoietic cells is summarized in Table 3 on page 464 of Ravetch, J.V., and Kinet, J.P., Annu. Rev. Immunol. 9 (1991) 457-492. Non-limiting examples of in vitro assays to assess ADCC activity of a molecule of interest is described in US 5,500,362 (see, e.g. Hellstrom, I., et al. Proc. Natl. Acad. Sci. USA 83 (1986) 7059-7063) and Hellstrom, I., et al, Proc. Natl. Acad. Sci. USA 82 (1985) 1499-1502); US 5,821 ,337 (see Bruggemann, M. et al, J. Exp. Med. 166 (1987) 1351-1361). Alternatively, non-radioactive assays methods may be employed (see, for example, ACTI™ non-radioactive cytotoxicity assay for flow cytometry (CellTechnology, Inc. Mountain View, CA; and CytoTox 96® non-radioactive cytotoxicity assay
(Promega, Madison, WI). Useful effector cells for such assays include peripheral blood mononuclear cells (PBMC) and Natural Killer (NK) cells. Alternatively, or additionally, ADCC activity of the molecule of interest may be assessed in vivo, e.g., in an animal model such as that disclosed in Clynes, R., et al., Proc. Natl. Acad. Sci. USA 95 (1998) 652-656. Clq binding assays may also be carried out to confirm that the antibody is unable to bind C lq and hence lacks CDC activity. See, e.g., C lq and C3c binding ELISA in WO 2006/029879 and WO 2005/100402. To assess complement activation, a CDC assay may be performed (see, for example, Gazzano-Santoro, H., et al, J. Immunol. Methods 202 (1997) 163-171 ; Cragg, M.S., et al, Blood 101 (2003) 1045-1052; and Cragg, M.S., and M.J., Glennie,
Blood 103 (2004) 2738-2743). FcRn binding and in vivo clearance/half life determinations can also be performed using methods known in the art (see, e.g., Petkova, SB. et al, Int. Immunol. 18 (2006) 1759-1769).
Antibodies with reduced effector function include those with substitution of one or more of Fc region residues 238, 265, 269, 270, 297, 327 and 329 (US 6,737,056). Such Fc mutants include Fc mutants with substitutions at two or more of amino acid positions 265, 269, 270, 297 and 327, including the so-called "DANA" Fc mutant with substitution of residues 265 and 297 to alanine (US 7,332,581).
Certain antibody variants with improved or diminished binding to FcRs are described. (See, e.g., US 6,737,056; WO 2004/056312, and Shields, R.L., et al, J. Biol. Chem. 9 (2001) 6591-6604).
In certain embodiments, an antibody variant comprises an Fc region with one or more amino acid substitutions which improve ADCC, e.g., substitutions at positions 298, 333, and/or 334 of the Fc region (EU numbering of residues).
In some embodiments, alterations are made in the Fc region that result in altered (i.e., either improved or diminished) Clq binding and/or Complement Dependent
Cytotoxicity (CDC), e.g., as described in US 6,194,551, WO 99/51642, and Idusogie E.E., et al. J. Immunol. 164 (2000) 4178-4184.
Antibodies with increased half lives and improved binding to the neonatal Fc receptor (FcRn), which is responsible for the transfer of maternal IgGs to the fetus (Guyer, R.L., et al, J. Immunol. 117 (1976) 587-593) and Kim, J.K., et al, J.
European J of Immunol. 24 (1994) 2429-2434), are described in US 2005/0014934A1 (Hinton et al.). Those antibodies comprise an Fc region with one or more substitutions therein which improve binding of the Fc region to FcRn. Such Fc variants include those with substitutions at one or more of Fc region residues: 238, 256, 265, 272, 286, 303, 305, 307, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 413, 424 or 434, e.g., substitution of Fc region residue 434 (US 7,371,826).
See also Duncan, A.R., and Winter, G., Nature 332 (1988) 738-740; US 5,648,260; US 5,624,821; and WO 94/29351 concerning other examples of Fc region variants. c) Cysteine engineered antibody variants
In certain embodiments, it may be desirable to create cysteine engineered antibodies, e.g., "thioMAbs," in which one or more residues of an antibody are substituted with cysteine residues. In particular embodiments, the substituted residues occur at accessible sites of the antibody. By substituting those residues with cysteine, reactive thiol groups are thereby positioned at accessible sites of the antibody and may be used to conjugate the antibody to other moieties, such as drug moieties or linker-drug moieties, to create an immunoconjugate, as described further herein. In certain embodiments, any one or more of the following residues may be substituted with cysteine: V205 (Kabat numbering) of the light chain; Al 18 (EU numbering) of the heavy chain; and S400 (EU numbering) of the heavy chain Fc region. Cysteine engineered antibodies may be generated as described, e.g., in US 7,521,541. d) Antibody Derivatives
In certain embodiments, an antibody provided herein may be further modified to contain additional non-proteinaceous moieties that are known in the art and readily available. The moieties suitable for derivatization of the antibody include but are not limited to water soluble polymers. Non- limiting examples of water soluble polymers include, but are not limited to, polyethylene glycol (PEG), copolymers of ethylene glycol/propylene glycol, carboxymethylcellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone, poly-1, 3-dioxolane, poly-l,3,6-trioxane, ethylene/maleic anhydride copolymer, polyaminoacids (either homopolymers or random copolymers), and dextran or poly(n-vinyl pyrrolidone)poly ethylene glycol, propropylene glycol homopolymers, prolypropylene oxide/ethylene oxide co-polymers, polyoxyethylated polyols (e.g., glycerol), polyvinyl alcohol, and mixtures thereof. Polyethylene glycol propionaldehyde may have advantages in manufacturing due to its stability in water. The polymer may be of any molecular weight, and may be branched or unbranched. The number of polymers attached to the antibody may vary, and if more than one polymer is attached, they can be the same or different molecules. In general, the number and/or type of polymers used for derivatization can be determined based on considerations including, but not limited to, the particular properties or functions of the antibody to be improved, whether the antibody derivative will be used in a therapy under defined conditions, etc.
In another embodiment, conjugates of an antibody and non-proteinaceous moiety that may be selectively heated by exposure to radiation are provided. In one embodiment, the non-proteinaceous moiety is a carbon nanotube (Kam, N.W., et al, Proc. Natl. Acad. Sci. USA 102 (2005) 11600-11605). The radiation may be of any wavelength, and includes, but is not limited to, wavelengths that do not harm ordinary cells, but which heat the non-proteinaceous moiety to a temperature at which cells proximal to the antibody-non-proteinaceous moiety are killed.
B. Recombinant Methods and Compositions Antibodies may be produced using recombinant methods and compositions, e.g., as described in US 4,816,567. In one embodiment, isolated nucleic acid encoding an anti-CD 19 antibody described herein is provided. Such nucleic acid may encode an amino acid sequence comprising the VL and/or an amino acid sequence comprising the VH of the antibody (e.g., the light and/or heavy chains of the antibody). In a further embodiment, one or more vectors (e.g., expression vectors) comprising such nucleic acid are provided. In a further embodiment, a host cell comprising such nucleic acid is provided. In one such embodiment, a host cell comprises (e.g., has been transformed with): (1) a vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and an amino acid sequence comprising the VH of the antibody, or (2) a first vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and a second vector comprising a nucleic acid that encodes an amino acid sequence comprising the VH of the antibody. In one embodiment, the host cell is eukaryotic, e.g. a Chinese Hamster Ovary (CHO) cell or lymphoid cell (e.g., Y0, NSO, Sp20 cell). In one embodiment, a method of making an anti-CD 19 antibody is provided, wherein the method comprises culturing a host cell comprising a nucleic acid encoding the antibody, as provided above, under conditions suitable for expression of the antibody, and optionally recovering the antibody from the host cell (or host cell culture medium). For recombinant production of an anti-CD 19 antibody, nucleic acid encoding an antibody, e.g., as described above, is isolated and inserted into one or more vectors for further cloning and/or expression in a host cell. Such nucleic acid may be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the antibody).
Suitable host cells for cloning or expression of antibody-encoding vectors include prokaryotic or eukaryotic cells described herein. For example, antibodies may be produced in bacteria, in particular when glycosylation and Fc effector function are not needed. For expression of antibody fragments and polypeptides in bacteria, see, e.g., US 5,648,237, US 5,789,199, and US 5,840,523. (See also Charlton, K.A., Methods in Molecular Biology, Vol. 248 (B.K.C. Lo, ed., Humana Press, Totowa, NJ, 2004), pp. 245-254, describing expression of antibody fragments in E. coli.) After expression, the antibody may be isolated from the bacterial cell paste in a soluble fraction and can be further purified.
In addition to prokaryotes, eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for antibody-encoding vectors, including fungi and yeast strains whose glycosylation pathways have been "humanized" resulting in the production of an antibody with a partially or fully human glycosylation pattern. See Gerngross, Nat. Biotech. 22 (2004) 1409-1414, and Li et al, Nat. Biotech. 24 (2006) 210-215.
Suitable host cells for the expression of glycosylated antibody are also derived from multicellular organisms (invertebrates and vertebrates). Examples of invertebrate cells include plant and insect cells. Numerous baculoviral strains have been identified which may be used in conjunction with insect cells, particularly for transfection of Spodoptera frugiperda cells.
Plant cell cultures can also be utilized as hosts. See, e.g., US 5,959,177, US 6,040,498, US 6,420,548, US 7,125,978, and US 6,417,429 (describing PLANTIBODIES™ technology for producing antibodies in transgenic plants). Vertebrate cells may also be used as hosts. For example, mammalian cell lines that are adapted to grow in suspension may be useful. Other examples of useful mammalian host cell lines are monkey kidney CVl line transformed by SV40 (COS-7); human embryonic kidney line (293 or 293 cells as described, e.g., in Graham. F.L., et al, J. Gen Virol. 36 (1977) 59-74; baby hamster kidney cells (BHK); mouse Sertoli cells (TM4 cells as described, e.g., in Mather, J.P., Biol.
Reprod. 23 (1980) 243-252); monkey kidney cells (CVl); African green monkey kidney cells (VERO-76); human cervical carcinoma cells (HELA); canine kidney cells (MDCK; buffalo rat liver cells (BRL 3A); human lung cells (W138); human liver cells (Hep G2); mouse mammary tumor (MMT 060562); TRI cells, as described, e.g., in Mather, J.P., et al, Annals N.Y. Acad. Sci. 383 (1982) 44-68;
MRC 5 cells; and FS4 cells. Other useful mammalian host cell lines include Chinese hamster ovary (CHO) cells, including DHFR" CHO cells (Urlaub, G., et al, Proc. Natl. Acad. Sci. USA 77 (1980) 4216-4220); and myeloma cell lines such as Y0, NS0 and Sp2/0. For a review of certain mammalian host cell lines suitable for antibody production, see, e.g., Yazaki, P.J., and Wu, A.M. Methods in Molecular Biology, Vol. 248 (2004) (B.K.C. Lo, ed., Humana Press, Totowa, NJ), pp. 255-268.
C. Methods and Compositions for Diagnostics and Detection In certain embodiments, any of the anti-CD 19 antibodies provided herein is useful for detecting the presence of CD 19 in a biological sample. The term "detecting" as used herein encompasses quantitative or qualitative detection. In certain embodiments, a biological sample comprises a cell or tissue, such as tumor tissue.
In one embodiment, an anti-CD 19 antibody for use in a method of diagnosis or detection is provided. In a further aspect, a method of detecting the presence of CD 19 in a biological sample is provided. In certain embodiments, the method comprises contacting the biological sample with an anti-CD 19 antibody as described herein under conditions permissive for binding of the anti-CD 19 antibody to CD 19, and detecting whether a complex is formed between the anti- CD 19 antibody and CD 19. Such method may be an in vitro or in vivo method. In one embodiment, an anti-CD 19 antibody is used to select subjects eligible for therapy with an anti-CD 19 antibody, e.g. where CD 19 is a biomarker for selection of patients.
Exemplary disorders that may be diagnosed using an antibody of the invention are inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases.
In certain embodiments, labeled anti-CD 19 antibodies are provided. Labels include, but are not limited to, labels or moieties that are detected directly (such as fluorescent, chromophoric, electron-dense, chemiluminescent, and radioactive labels), as well as moieties, such as enzymes or ligands, that are detected indirectly, e.g., through an enzymatic reaction or molecular interaction. Exemplary labels
32 14 125 3 131 include, but are not limited to, the radioisotopes P, C, I, H, and I, fluorophores such as rare earth chelates or fluorescein and its derivatives, rhodamine and its derivatives, dansyl, umbelliferone, luceriferases, e.g., firefly luciferase and bacterial luciferase (US 4,737,456), luciferin, 2,3- dihydrophthalazinediones, horseradish peroxidase (HRP), alkaline phosphatase, β- galactosidase, glucoamylase, lysozyme, saccharide oxidases, e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase, heterocyclic oxidases such as uricase and xanthine oxidase, coupled with an enzyme that employs hydrogen peroxide to oxidize a dye precursor such as HRP, lactoperoxidase, or microperoxidase, biotin/avidin, spin labels, bacteriophage labels, stable free radicals, and the like.
D. Pharmaceutical Formulations
Pharmaceutical formulations of an anti-CD 19 antibody as described herein are prepared by mixing such antibody having the desired degree of purity with one or more optional pharmaceutically acceptable carriers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions. Pharmaceutically acceptable carriers are generally nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyl dimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinyl pyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g. Zn-protein complexes); and/or non-ionic surfactants such as polyethylene glycol (PEG). Exemplary pharmaceutically acceptable carriers herein further include interstitial drug dispersion agents such as soluble neutral-active hyaluronidase glycoproteins (sHASEGP), for example, human soluble PH-20 hyaluronidase glycoproteins, such as rhuPH20 (HYLENEX®, Baxter International, Inc.). Certain exemplary sHASEGPs and methods of use, including rhuPH20, are described in US 2005/0260186 and US 2006/0104968. In one aspect, a sHASEGP is combined with one or more additional glycosaminoglycanases such as chondroitinases. Exemplary lyophilized antibody formulations are described in US 6,267,958.
Aqueous antibody formulations include those described in US 6, 171 ,586 and WO 2006/044908, the latter formulations including a histidine-acetate buffer.
The formulation herein may also contain more than one active ingredients as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other. Such active ingredients are suitably present in combination in amounts that are effective for the purpose intended.
Active ingredients may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methyl methacrylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical Sciences, 16th edition, Osol, A. (ed.), (1980).
Sustained-release preparations may be prepared. Suitable examples of sustained- release preparations include semi permeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g. films, or microcapsules. The formulations to be used for in vivo administration are generally sterile.
Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes.
E. Therapeutic Methods and Compositions
Any of the anti-CD 19 antibodies provided herein may be used in therapeutic methods.
In one aspect, an anti-CD 19 antibody as reported herein for use as a medicament is provided. In further aspects, an anti-CD 19 antibody as reported herein for use in treating inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases is provided. In certain embodiments, an anti-CD 19 antibody as reported herein for use in a method of treatment is provided. In certain embodiments, the invention provides an anti-CD 19 antibody for use in a method of treating an individual having inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone disease comprising administering to the individual an effective amount of the anti-CD 19 antibody as reported herein. In one such embodiment, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., as described below. In further embodiments, the invention provides an anti-CD 19 antibody for use in blocking B- cell activation without causing B-cell depletion. In certain embodiments, the invention provides an anti-CD 19 antibody for use in a method of blocking B-cell activation without causing B-cell depletion in an individual comprising administering to the individual an effective of the anti-CD 19 antibody as reported herein. An "individual" according to any of the above embodiments is in one embodiment a human.
In a further aspect, the invention provides for the use of an anti-CD 19 antibody as reported herein in the manufacture or preparation of a medicament. In one embodiment, the medicament is for treatment of inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone disease. In a further embodiment, the medicament is for use in a method of treating inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone disease comprising administering to an individual having inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone disease an effective amount of the medicament. In one such embodiment, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., as described below. In a further embodiment, the medicament is for blocking B-cell activation without causing B-cell depletion. In a further embodiment, the medicament is for use in a method blocking B-cell activation without causing B-cell depletion in an individual comprising administering to the individual an amount effective of the medicament. An "individual" according to any of the above embodiments may be a human.
In a further aspect, the invention provides a method for treating an inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone disease. In one embodiment, the method comprises administering to an individual having such inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone disease an effective amount of an anti-CD 19 antibody as reported herein. In one such embodiment, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, as described below. An "individual" according to any of the above embodiments may be a human. In a further aspect, the invention provides a method for blocking B-cell activation without causing B-cell depletion in an individual. In one embodiment, the method comprises administering to the individual an effective amount of an anti-CD 19 antibody as reported herein. In one embodiment, an "individual" is a human. In a further aspect, the invention provides pharmaceutical formulations comprising any of the anti-CD19 antibodies provided herein, e.g., for use in any of the above therapeutic methods. In one embodiment, a pharmaceutical formulation comprises any of the anti-CD 19 antibodies provided herein and a pharmaceutically acceptable carrier. In another embodiment, a pharmaceutical formulation comprises any of the anti-CD 19 antibodies provided herein and at least one additional therapeutic agent, e.g., as described below.
Antibodies of the invention can be used either alone or in combination with other agents in a therapy. For instance, an antibody of the invention may be co- administered with at least one additional therapeutic agent.
Such combination therapies noted above encompass combined administration (where two or more therapeutic agents are included in the same or separate formulations), and separate administration, in which case, administration of the antibody of the invention can occur prior to, simultaneously, and/or following, administration of the additional therapeutic agent and/or adjuvant. Antibodies of the invention can also be used in combination with radiation therapy.
An antibody of the invention (and any additional therapeutic agent) can be administered by any suitable means, including parenteral, intrapulmonary, and intranasal, and, if desired for local treatment, intralesional administration. Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. Dosing can be by any suitable route, e.g. by injections, such as intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic. Various dosing schedules including but not limited to single or multiple administrations over various time- points, bolus administration, and pulse infusion are contemplated herein.
Antibodies of the invention would be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners. The antibody need not be, but is optionally formulated with one or more agents currently used to prevent or treat the disorder in question. The effective amount of such other agents depends on the amount of antibody present in the formulation, the type of disorder or treatment, and other factors discussed above. These are generally used in the same dosages and with administration routes as described herein, or about from 1 to 99 % of the dosages described herein, or in any dosage and by any route that is empirically/clinically determined to be appropriate. For the prevention or treatment of disease, the appropriate dosage of an antibody of the invention (when used alone or in combination with one or more other additional therapeutic agents) will depend on the type of disease to be treated, the type of antibody, the severity and course of the disease, whether the antibody is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody, and the discretion of the attending physician. The antibody is suitably administered to the patient at one time or over a series of treatments. Depending on the type and severity of the disease, about 1 μg/kg to 15 mg/kg (e.g. 0.1 mg/kg - 10 mg/kg) of antibody can be an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion. One typical daily dosage might range from about 1 μg/kg to 100 mg/kg or more, depending on the factors mentioned above. For repeated administrations over several days or longer, depending on the condition, the treatment would generally be sustained until a desired suppression of disease symptoms occurs. One exemplary dosage of the antibody would be in the range from about 0.05 mg/kg to about 10 mg/kg. Thus, one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 4.0 mg/kg or 10 mg/kg (or any combination thereof) may be administered to the patient. Such doses may be administered intermittently, e.g. every week or every three weeks (e.g. such that the patient receives from about two to about twenty, or e.g. about six doses of the antibody). An initial higher loading dose, followed by one or more lower doses may be administered. An exemplary dosing regimen comprises administering about 4 to 10 mg/kg. However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
It is understood that any of the above formulations or therapeutic methods may be carried out using an immunoconjugate of the invention in place of or in addition to an anti-CD 19 antibody.
F. Articles of Manufacture
In another aspect of the invention, an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of the disorders described above is provided. The article of manufacture comprises a container and a label or package insert on or associated with the container. Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc. The containers may be formed from a variety of materials such as glass or plastic. The container holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). At least one active agent in the composition is an antibody of the invention. The label or package insert indicates that the composition is used for treating the condition of choice.
Moreover, the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises an antibody of the invention; and (b) a second container with a composition contained therein, wherein the composition comprises a further therapeutic agent. The article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the compositions can be used to treat a particular condition. Alternatively, or additionally, the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate -buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
It is understood that any of the above articles of manufacture may include an immunoconjugate of the invention in place of or in addition to an anti-CD 19 antibody.
III. Description of the Sequence Listing
SEQ ID NO : 1 heavy chain variable region (VH) of Mab381
SEQ ID NO : 2 heavy chain CDRH 1 Mab381
SEQ ID NO : 3 heavy chain CDRH2 Mab381
SEQ ID NO: 4 heavy chain CDRH3 Mab381
SEQ ID NO : 5 light chain variable region (VL) of Mab381
SEQ ID NO : 6 light chain CDRL2 Mab381
SEQ ID NO : 7 light chain CDRL3 Mab381
SEQ ID NO: 8 heavy chain variable region (VH) of Mab396 (CD HA)
SEQ ID NO: 9 heavy chain CDRH1 Mab396 SEQ ID NO 10 heavy chain CDRH2 Mab396
SEQ ID NO 11 heavy chain CDRH3 Mab396
SEQ ID NO 12 light chain variable region (VL) of Mab396 (CD LB)
SEQ ID NO 13 light chain CDRL1 Mab396
SEQ ID NO 14 light chain CDRL2 Mab396
SEQ ID NO 15 light chain CDRL3 Mab396
SEQ ID NO 16 heavy chain CDRH2 of heavy chain variable region (VH) of CD
HC
SEQ ID NO: 17 heavy chain CDRH2 of heavy chain variable region (VH) of CD
HD
SEQ ID NO: 18 light chain CDRL2 of light chain variable region (VH) of CD LA SEQ ID NO: 19 light chain CDRL1 of light chain variable region (VH) of CD LC SEQ ID NO: 20 light chain CDRL1 of light chain variable region (VH) of CD LD SEQ ID NO: 21 heavy chain variable region (VH) of S J25
SEQ ID NO: 22 heavy chain CDRH1 of SJ25
SEQ ID NO: 23 heavy chain CDRH2 of SJ25
SEQ ID NO: 24 heavy chain CDRH3 of SJ25
SEQ ID NO: 25 Human kappa light chain
SEQ ID NO: 26 Human lambda light chain
SEQ ID NO: 27 Human IgGl (Caucasian Allotype)
SEQ ID NO: 28 Human IgGl (Afroamerican Allotype)
SEQ ID NO: 29 Human IgGl LALA-Mutant (Caucasian Allotype)
SEQ ID NO: 30 Human IgG4
SEQ ID NO: 31 Human IgG4 SPLE-Mutant
IV. Antibody Nomenclature
Figure imgf000043_0001
Sequence Number
CD HD T28A 9 17 11
CD HE T28A 9 10 11
Vl lL
K12V
CD HF T28A 9 10 11
Vl lL
K12V
Y91F
T108S
CD LA 12 13 18 15
CD LB 13 14 15
CD LC 19 14 15
CD LD 20 14 15
CD LE2) P80S 20 14 15
F83L
' CD HB is a heavy chain variable region comprising T28A mutation in SEQ ID NO: 8; T28A means that amino acid T of position 28 of SEQ ID NO: 8 is replaced by amino acid A; etc. CD HB comprises CDRHl of SEQ ID NO: 9, CDRH2 of SEQ ID NO: 10, and CDRH3 of SEQ ID NO: 11.
2) CD LE is alight chain variable region comprising P80S and F83L mutation in SEQ ID NO: 12. P80S means that amino acid P of position 80 of SEQ ID NO: 12 is replaced by amino acid S; etc. CD LE comprises CDRL1 of SEQ ID NO: 20, CDRL2 of SEQ ID NO: 14, and CDRL3 of SEQ ID NO: 15.
Mab396 and all CD HX and CD LX regions are humanized versions of Mab381 with analog nomenclature as CD HB and CD LE.
V. EXAMPLES
The following are examples of methods and compositions of the invention. It is understood that various other embodiments may be practiced, given the general description provided above.
Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, the descriptions and examples should not be construed as limiting the scope of the invention. The disclosures of all patent and scientific literature cited herein are expressly incorporated in their entirety by reference. Example 1
Immunization and generation of mouse anti-hCD19 antibodies (hybridomas)
Balb/c mice were immunized six times and boosted with CD19-transfected HEK293 cells (mean receptor density 35,000 per cell). The immune response was monitored by testing serum samples with a CD 19 - cell-ELISA on CD 19- transfected NIH-3T3 cells. Spleen cells from mice with sufficient titers of anti- CD^ immunoglobulin were used for immortalization by fusion with mouse myeloma cell line P3X63 Ag8.653. Three fusions were carried out and hybridoma supernatants screened by cell-ELISA on CD19-transfected NIH-3T3 cells and FACS binding assay using Daudi (CD19+) and CD 19" cells for anti-CD 19 specific antibodies.
The positive hybridoma 8C3, producing Mab381, was selected for the further evaluation based on the combination of the assays and properties. Mab381 was selected as preferred antibody due to its biochemical and functional properties described in the following. The functional properties of the antibody in biochemical and cellular assays are described in examples 2-8. A property of the hybridoma 8C3 and antibody Mab381, derived from this hybridoma, as well as for the humanized version Mab396 and the other humanized light and heavy chain versions, for the selection was their inhibitory activity on activated primary B-cells. Purified antibody from in-vitro production of the 8C3 hybridoma was used for characterization in different biochemical and cellular assays (see below). The hybridoma 8C3 was cloned by single cell FACS sorting.
Example 2
Hybridoma screening and cell biological functional evaluation of anti-CD19 antibody
Cell-ELISA for screening antibodies against hCD19
A cell ELISA was applied for screening of hybridomas, and to identify those hybridomas that secrete antibodies against human-CD 19. NIH3T3 cells transfected with human-CD 19 were used as positive cells; untransfected NIH3T3 cells were used as negative control cells. For the assessment of the positive hybridomas the OD ratio between transfected and untransfected NIH3T3 cells was quantified. - Culture Medium: DMEM high glucose (4.5 mg/ml), 10 % FCS, Na-Pyruvate, NEAA, Glutamin
- Antibodies positive control: anti CD 19 monoclonal antibody (IgGl) Pharmingen Cat# 555409 c = 1 mg/ml
- Detection antibody: Goat anti-Mouse IgG (H+L) HRP Conjugate Bio-Rad Cat#
170-06516
- Dilution 1 : 2000 in lx ELISA Blocking Reagent
- Other reagents: Fibronectin Roche Cat# 838039 c = 1 mg/ml
- Glutardialdehyde: 25 % stock solution // Grade Agar Scientific #R102 final concentration: 0.05 % in PBS
- ELISA Blocking Reagent: 1 Ox stock solution // Roche Cat# 1112589
- TMB substrate: Roche Cat# 11432559
- Stop Solution: 1 M H2S04
- BioRad Cat# 170-6516 Dilution 1 : 2000 in lx ELISA Blocking Reagent
Day 1 :
- Fibronectin coating: 5 μg/cm2 in PBS; 96well plate = 32 cm2; 160 μg/plate in 6 ml
- PBS, 50 μΐ/well
- incubate 45 min at RT, aspirate coating solution
- Seed 1.25 x 104 cells/well in 50 μΐ culture medium in a 96well plate
- incubate 40 hours at 37 °C
- add to upper half of the plate: NIH3T3 cells expressing CD 19
- add to lower half of the plate: non-transfected NIH3T3 cells
Day 3:
- Addition of positive control antibody or samples (supernatant or mouse serum) in 50 μΐ culture medium
- incubate for 2 h at 4 °C
- Remove medium, fix cells with 100 μΐ Glutardialdehyde (0.05 % in PBS)
- Wash two times with 200 μΐ PBS
- Addition of detection antibody 1 :2000, 50 μΐ/well
- incubate 2 h at RT
- wash three times with 200 μΐ PBS
- add 50 μΐ TMB, incubate for 30 min. at RT,
- stop by addition of 25 μΐ 1 M H2S04; read extinction at 450nm/620nm
- Calculation of results: ratio OD NIH3T3 CD19 : OD NIH3T3 untransfected Results:
The antibody Mab381 demonstrates specific binding to CD 19 transfected NIH3T3 cells as compared to untransfected NIH3T3 cells (ratio OD NIH3T3 CD 19 to OD NIH3T3 untransfected = 11.4).
Example 3
FACS Binding of anti-CD 19 antibodies on DAUDI cells
To evaluate the binding potency of and antibody according to the invention, e.g. Mab381, to human CD 19 expressed on the cell surface the human B-cell line
Daudi was used. Daudi cells were incubated with different concentrations of the purified Mab381 and in a second step with a goat-anti-mouse labeled antibody. Quantification of the binding was performed by FACS analysis; the EC50 value for the binding potency was calculated.
Material & Method
- Cell line: Daudi (Human Burkitt Lymphoma) ATCC-Nr. CCL-213
- Culture Medium: RPMI1640 supplemented with 10 % FCS, 2 mM L- Glutamine, 100 μΜ NEAA, 1 mM Na-Pyruvate
- Positive Control: anti-CD 19 antibody SJ25-C1 c = 0.31 mg/ml
- Isotype Control: mlgGl kappa Sigma Cat# M5284 Lot#023K4855 c = 0.2 mg/ml
- Detection antibody: goat anti mouse RPE F(ab')2 IgG (H+L) Caltag Cat# M355-004-3 Lot#26020107 c = 0.2 mg/ml
- Sample: purified antibody Mab381.
- 0-JX 105 cells per sample in 12 x 75 mm FACS-tube were stained with 100 μΐ antibody solution
- incubate in PBS/10 % FCS 20 min. on ice
o Antibody concentrations: Samples and positive control: from 30 to 0.04 μg/ml in 1 :3 dilutions
o Isotype control: 1 μg/100 μΐ
- Wash one with 3 ml PBS/10 % FCS
- Addition of 100 μΐ detection antibody (1 μg/100 μΐ); incubation 15 min. on ice
- Wash once with 3 ml PBS/10 % FCS
- Resuspend pellet in 500 μΐ PBS/10 % FCS and analyze in FACS-Calibur. Antibody Mab381 shows binding to Daudi cells as determined by FACS analysis (EC50 mean = 1.45 nM). Results are shown in table 1.
Table 1: Comparison of Mab381 and humanized Mab396
Figure imgf000048_0001
Example 4
CD 19 phosphorylation inhibiting assay / Western blot analysis of phospho- CD19 (Tyr531) with human RAMOS cells
CD 19 is a part of the B-cell co-receptor. On the cellular level stimulation of the B- cell coreceptor is possible with an anti-human-IgM antibody. The activated CD 19 can be detected with an anti-phospho-CD19 (Tyr531) antibody. The Ramos cell line (human B-cell lymphoma) was used and the quantification of the phosphorylated CD 19 was done in the cell extracts by Western-Blot- Analysis with a Lumi-Imager™.
Material & Method: - Cell line: RAMOS (ATCC CRL 1596, human, Caucasian, lymphoma, Burkitt)
- Pre-Incubation with anti-CD 19 antibody
- Stimulation: anti-human IgM (Pharmingen, Cat-Nr.: 555780) 12 μg/ml for 2 minutes
- Extracts: 2 x 106 cells in modified RIPA-buffer (+ 2 mM Na-Orthovanadate) - Gel: 7.5 % Tris-HCl Ready Gel, Biorad
- Nitrocellulose membrane: Trans-BlotR Transfer Medium (7 x 8.4 cm), Biorad
- Blotting: 30 min. in SEMI-Dry Blotting Apparatus (Biorad), 20 V const.
- Primary antibody: Anti-Phospho-CD19 (Tyr531), polyclonal rabbit, Cell Signaling #3571
- Secondary antibody: Anti-Rabbit-POD (Roche, Chemiluminescence Western
Blotting Kit) 1 : 1000
- Detection: Lumi-LightPLUS Western Blotting Substrate (Roche) Results:
Antibody Mab381 (1 μ /ηι1 and 10 μ /ηι1) induces inhibition of anti-IgM stimulated phospho Tyr531-CD19 levels. Level of inhibition reaches non- stimulated control. Example 5
Internalization / Down-modulation of CD19 in RAMOS cells
Goal was to test whether Mab381 causes internalization/down-modulation of the CD 19 receptor from the cell surface after binding. Ramos cells were incubated for 3 hours with antibodies, and a PE labeled goat-anti-mouse-antibody was used as detection antibody. The cell surface staining was analyzed by FACScan flow cytometer. The internalization was calculated applying a specific formula (see below) under consideration of the non-modulated probe (cells with antibody incubated by 0°C).
Material & Method Modulation of CD 19 induced by anti-CD 19 antibodies was evaluated by flow cytometry.
Cells: RAMOS (ATCC CRL 1596, human, Caucasian, lymphoma, Burkitt) cell line.
After centrifugation at 200 g 5 x 105 cells in RPMI1640 medium supplemented with 10 % FCS were mixed at 0 °C with 2.5 μ^πιΐ anti-CD 19 antibodies for 45 min. (100 μ1/5χ105 cells).
The suspensions were then incubated at 37 °C for 15 min. up to 18 hours in the continuous presence of antibody, so that the dissociation rate was negligible, and that the antigen was always saturated. Other samples treated with antibody were kept at 0 °C to evaluate the level of antigen expression on non-modulated cells.
After two washings with cold medium cells were stained with Goat F(ab')2 anti- mouse IgG (H+L) PE (see example 3) for 30 min. at 0 °C, washed twice again and kept in 1 ml cold medium on ice to measure the amount of surface-bound antibody on the cell. Ten thousand events were analyzed on a FACScan™ flow cytometer (Becton Dickinson). The extent of modulation at 37 °C was calculated for each sample as the percentage of MFI in relation to non-modulated cells according to the equation (calculation formula): Modulation (%) = 100 - 100 x (MFI [sample] - MFI [neg. control]/MFI [non- modulated cells]- MFI [neg. control]).
Results:
Down modulation was calculated after 3 hours incubation of Ramos cells with the antibodies (table 2).
Table 2:
Figure imgf000050_0001
Mab381 shows an internalization rate of 57 % after 3 hours.
Example 6
Proliferation assay by the B-cell lines SU-DHL4 and RCHACV
CD 19 as a member of the B-cell-co-receptor may play a role for proliferation of B-cells. The inhibitory activity of Mab381 is determined for the proliferation of human B-cell lines with different expression levels of CD 19 and CD20 (see below). Cells were incubated with antibodies for 3 days. Viable cells after this incubation period were analyzed and calculated by FACS (FACSArray™ BD
Biosciences).
Material & Method
- Cell lines: human B-cell lymphoma SU-DHL4 (CD19+ / CD20+) (DSMZ # ACC 495),
- human B-cell precursor leukemia RCHACV (CD19+ / CD20") (DSMZ #ACC
548
- human Plasmacytoma (B lymphocyte) RPMI 8226 (CD 19" / CD20") (ATCC, CCL 155)
- Medium: RPMI 1640 with 10% FBS and 2 mM L-Glutamin
- Plates: 96-well plates (Costar # 3799) Antibodies
Anti CD20 recombinant human monoclonal antibody c = 10 mg/ml
Mouse Anti-human CD 19 Antibody IgGl (PharMingen) c = 1.05 mg/ml
Clone HD37 (Chemicon) CD19 HD37; c = 0.56 mg/ml
Mouse Anti-human CD19 Antibody IgGl (Cymbus) CD19 SJ25-C1 c = 0.31 mg/ml
Mouse Anti-human CD 19 Antibody IgGl Mab381 c = 1.22 mg/ml Day 1 :
Seed cells with 2xl04 cells/well in 100 μΐ culture medium in 96-well round bottom plate. Add antibodies diluted in culture medium 2-fold of the desired concentrations in 100 μΐ/well. Incubate for 72 h at 37 °C, 5 % C02, 95 % rH.
Day 3:
Analyze viable cell number by FACS (FACSArray™ BD Biosciences) and calculate % inhibition of proliferation. Maximal inhibition of proliferation achieved with the antibody 381 was 25-39 % at a concentration of 2.5 - 10 μg/ml (values at an antibody concentration of 10 μg/ml: SU-DHL4: 37 %; RCHACV 25 %; RPMI 8226: 0 %). The antibody shows no inhibition of proliferation of a CD 19 negative cell line.
Example 7
Proliferation assay on B-cells isolated from Leucapheresis
Proliferation-inhibitory activity of Mab381 was tested using primary human B- cells after stimulation with a combination of an anti-IgM-antibody and h-IL4. The primary B-cells were isolated from Leucapheresis material from different healthy donors applying a negative selection process (depletion of non-B-cells); purity of the B-cells used in these experiments was >90 %. The cells were incubated with antibodies for 7 days; proliferation rate was measured using a BrdU assay. Material & Method
B-cells isolated from Leucapheresis Material of healthy donors isolated by depletion of non-B-cells (negative selection) with Magnetobeads (Dynal® B-cell Negative Isolation Kit [CD2,14,16,36,43,235a]). B-cells; purity 92 % - 96 %; thawed, viability 90-96 %. 2 x wash and seeded ~2- 3 x 105 cells/ well in 100 μΐ RPMI/2 % FBS/NEAA/pen-strep in 96 well plate. Anti-hCD19 antibodies were added in concentration from 0.08 μ§/ι 1 to 10 μ§/ι 1 dilution 1 :5 (positive control - SJ25-C1 and MabThera). One hour preincubation with anti-CD 19 antibodies in incubator at 37 °C/5 % C02. Cells were stimulated with 10 μ§/ι 1 anti-IgM/20 ng/ml IL-4; end Volume = 200 μΐ/well; incubation for 7 days at 37 °C/5 % C02 ; read out BrdU Assay.
Materials:
1. Medium: RPMI +1 % L-Glutamin, PAN Biotech, Cat. No P04-17500; Lot No 030905; Exp: 09/2007 with 10 % FBS, ultra low IgG, Gibco, Lot No 3102090A;
NEAA, PAN Biotech, Cat. No 08-32100; Lot No 420506; Pen/Strep, Roche, 50,000 U/ml/50 mg/ml)
2. Read out: BrdU, Roche, Cat No: 1 647 229;
3. Plate: NUNC 96 Micro Well™ Plate, Flatbottom, Cat No 167008,
4. for Stimulation: Anti IgM (AffiniPure F(ab')2 Fragment Goat Anti-human IgA +
IgG + IgM (H+L), Dianova, code 109-006-064, Lot. 67679) 1.3 mg/ml
Human, recombinant IL-4
Results:
The antibody Mab381 shows a significant inhibition of proliferation of primary B- cells from three donors stimulated with anti-IgM and IL-4 (table 4).
Table 4:
% Inhibition of proliferation; B-cells from
Antibody [^g/ml]
Leukapheresis
Donor 1 Donor 2 Donor 3
2 (Mab381) 34 % 1 1 % 42 %
10 (Mab381) 45 % 45 % 40 %
2 ((Mab396 =
83 % n.d. 38 %
381LBHA)
10 (Mab396 =
91 % 90 % 49 %
381LBHA) Example 8
Ca(2+) Influx Assay with the Ramos cell line
B-cells show changes of intracellular Ca2+ levels upon activation. Goal here was to show the inhibitory activity of Mab381 on activation of B-cells with Ca2+ Influx as read out. The Ramos cell line was used for these studies. Modulation of the Ca2+ Influx (changes in intracellular calcium) was measured using fluorescent Ca2+ indicators. After loading cells with the calcium sensitive fluorescent dye, cells were incubated with antibodies for 2.5-3.0 hours. Quantification of fluorescence was done by using the Flex Station workstation. Material & Method
Modulation of Ca2+ Influx induced by anti-CD 19 antibodies in Ramos cells was evaluated by FLIPR Calcium Kit/Flex Station™ (Molecular Devices)- Fluorescence-based assay for changes detecting in intracellular calcium. After centrifugation at 200 g about 2 x 105 cells in HBSS medium (50 μΐ/well) were seeded in 96 well plate (black with clear bottom). The antibodies were added - 25 μΐ/ well in end concentrations - from 10 μg/ml to 0.001 μg/ml (positive control- SJ25-C1). The cells were then incubated in the incubator at 37 °C/5 % C02 for 1 hour in the continuous presence of antibody. The Loading puffer added (loading the cells with calcium sensitive fluorescent dye) - 100 μΐ/well. The antibodies were added - 25 μΐ/ well. The cells were then incubated in the incubator at 37 °C/5 %
C02 for 1 hour in the continuous presence of antibody. And 30 to 60 min at RT for a total time of incubation with anti-CD 19 antibodies 2.5-3 hours. Fluorescence monitored in Flex Station™ after addition 3 μg/ml of anti-IgM antibodies to the cells. Flex Station™ is benchtop scanning fluorometer with integrated fluid transfer workstation; the machine monitors the wells over a time course, during which compounds can be added robotically. Responses were measured as peak fluorescence intensity minus basal fluorescent intensity.
Result:
In Ramos cells after activation, the antibody Mab381 inhibits Ca2+ Influx by an EC50 of 0.37 nM (56 ng/ml, with a maximal inhibition of 50 %) and the antibody
Mab396 inhibits Ca2+ Influx by an EC50 of 0.20 nM (30 ng/ml, with a maximal inhibition of 50 %). Example 9
Sequencing of anti-human CD19 mouse hybridoma monoclonal antibody Mab381
The murine monoclonal antibody Mab381 expressed by a hybridoma cell line was cloned to determine the protein sequence of the antibody. The isotype of the murine antibody was immunologically identified as IgGl and kappa.
Example 10
Humanization of anti-CD19 antibody Mab381
The CD 19 binding specificity of the murine antibody Mab381 was transferred onto a human acceptor framework to eliminate potential immunogenicity issues arising from sequence stretches that the human body will recognize as foreign. This was done by engrafting the entire complementary determining regions (CDR) of the murine (donor) antibody onto a human (acceptor) antibody framework, and is called CDR-grafting or antibody humanization. The murine amino acid sequence was aligned with a collection of human germ- line antibody V genes, and sorted according to sequence identity and homology. Before selecting one particular acceptor sequence, the so-called canonical loop structures of the donor antibody have to be determined (Morea, V., et al, Methods, Vol 20, Issue 3 (2000) 267-279). These canonical loop structures are determined by the type of residues present at the so-called canonical positions. These positions lie (partially) outside of the CDR regions, and have to be kept functionally equivalent in the final construct in order to retain the CDR conformation of the parental (donor) antibody. The human germ- line sequence IGHVl-69*06 (IMGT Ac No L22583; old nomenclature: DP88) was chosen as the acceptor for the heavy chain and sequence IGKV1-9 (IMGT Acc No. Z00013) was chosen for the light chain.
Table 5: Determination of the canonical loop structures of Mab381
Determination of canonical classes of CDR loops CDR residues CDR length Canonical Structure
CDR Residues CDR length Canonical
Structure
CDR-L1 L2=I; L25=A; 11 aa Class 2A
L29=V; L30=G;
L33=V; L71=F.
CDR-L2 L34=A 7 aa Class 1 Determination of canonical classes of CDR loops CDR residues CDR length
Canonical Structure
(L48=I; L64=G)
CDR-L3 L90=Q; L94=Y; 9 aa Class 1
L95=P; L97=T
CDR-H1 H24=A; H26=G; 10 aa Class 1
(H27=Y); H29=F;
H34=M; H94=R
CDR-H2 H52a=P; [H54=D]; 4 aa Class 2
H55=G; H71=A;
The canonical structure class is: 1-2-2-1-1. (H1-H2-L1-L2-L3)
The potential acceptor sequence was selected based on high overall homology and the presence of the right canonical residues already in the acceptor sequence. The humanized constructs are labeled CD LA, CD LB, CD LC, CD LD, and CD LE for the light chain, and CD HA, CD HB, CD HC, CD HD, CD HE, and CD HF for the heavy chain.
Example 11
Binding of humanized antibodies compared to murine antibody Daudi target cells were incubated with the antibodies and binding was quantified using a fluorescently labeled secondary antibody and subsequent analysis via flow cytometry (FACS). The EC50 values of the parental murine antibody and the humanized variants are essentially unchanged. The different signal amplitude of the murine antibody compared to the humanized ones is due to a different secondary antibody used for detection of human or murine Fc.
Comparison of binding affinity of different humanized and chimeric Mab381 variants, as determined with flow cytometry is shown in Figure 1 and Figure 2.
Example 12
Binding to Clq (competence for CDC) The binding property of the antibody to the Clq protein of the human complement system is an indicator for its CDC potency. Different variants of the humanized Mab381 (such as Mab396) produced in HEK293 or CHO cells were tested by Clq- ELISA. Result: The humanized antibody Mab396 did not show any binding activity to the human Clq protein; therefore the expectation is that it will not trigger CDC.
Example 13
Glycoengineering of Mab396 (Mab396-g2 (an antibody with low fucosylation, see WO 99/54342))
The expression system comprises the MPSV promoter system for transient expression and is described in tables 6 and 7.
Table 6: pETR 4078 / pETR3388 (antibody expression vectors)
Figure imgf000056_0001
Table 7: pETR1519 / pCLF9 (glycosylation vectors)
Figure imgf000056_0002
All four plasmids were transfected in HEK293 EBNA host cells using a Ca2+- phosphate dependent co-transfection procedure. For antibody expression transfected cells were incubated in DMEM/10 % FCS at 37 °C in humidified incubators with a 5 % C02 atmosphere for seven days. Mab396-g2 was isolated from the supernant and purified by chromatographic methods. Example 14
ADCC assay
The ADCC potency of the humanized antibody 396 was investigated. The ADCC was performed using the Raji cell line and a high affinity Fcgamma-Receptor expressing cell line as effector cells. The read out for killing of Raji cells was LDH release. It was found, that the antibody does not show any ADCC. Mab396 (wt Fc region) and Mab396 with mutations L234A/L235A (LALA-Fc region) did not mediate significant amount of ADCC. Only by using very high concentrations of up to 300 μg/ml a moderate ADCC effect (15 %) has been observed. In contrast, Mab396 with low fucosylation (Mab396-g2, see WO 99/54342 and example 13) shows already ADCC mediated cell killing at lower concentrations. Results are shown in Figure 3.
Example 15
Whole blood (B-cells) depletion assays Goal was to evaluate the depletion potency of the antibody 381 and its humanized variants in human B-cells in- vitro. Whole Blood samples (after erythrocyte removal) from different healthy donors were incubated for 24 hours with the antibodies; B-cell depletion was calculated by the ratio: B-cells (CD 19 positive cells): T-cells (CD3 positive cells) with FACS analysis. A human/mouse chimeric variant of Mab381 antibody, the construct with chimeric heavy chain and light chain LB, as well as light chain LE combined with heavy chain HF do not mediate significant B-cell depletion. The other humanized variants do induce B-cell depletion.
Figures 4 and 5 show the results of a B-cell depletion experiment, where human whole blood samples were incubated for 24 hours with the indicated antibody samples (ch: chimeric; LBHch: LB and heavy chain chimeric 8C3 Mab381)). B- cell depletion was measured by counting the CD 19 positive cells stained with a labeled anti-CD20 antibody in a flow cytometer (after erythrocyte removal by cell lysis) relative to the CD3 positive cells as a reference. Constructs that are comprised of the heavy chain variants HA, HB, or HD show a B-cell depleting behavior, whereas constructs having the chimeric heavy chain (either together with the chimeric light chain, or the humanized light chain LE), or the heavy chain variants HE, and HF show a strongly reduced activity. In both experiments, variants HE and HF show B-cell depletion activity at the same low level as the parental antibody, or even less. The humanized variant antibody 396 shows a depletion potency of human B-cells of 28 %. This depletion potency is achieved at a concentration of 5 ng/ml and remains with higher concentrations.
Example 16
Cross-reactivity of anti-CD19 antibodies
Cross-reactivity was measured by FACS using B-cells of the respective species. It was found that Mab381 and its humanized variants show cross-reactivity with human and chimpanzee CD 19 only, but not with cynomolgus, dog, rabbit, hamster, mouse and other species investigated.

Claims

Patent Claims
An isolated antibody that binds to CD 19, characterized in binding to the same CD 19 epitope to which monoclonal antibody 381 binds, wherein antibody 381 is characterized by comprising as heavy chain variable region SEQ ID NO: l and as light chain variable region SEQ ID NO:5.
The antibody of claim 1, characterized in being a human, humanized, or chimeric antibody.
The antibody according to any one of claims 1 to 2, characterized in that the heavy chain variable domain comprises as CDRs a) a CDRH1 region of SEQ ID NO: 2, a CDRH2 region of SEQ ID NO: 3 and a CDRH3 region of SEQ ID NO: 4,
b) a CDRH1 region of SEQ ID NO: 9, a CDRH2 region of SEQ ID NO:
10 and a CDRH3 region of SEQ ID NO: 11,
c) a CDRH1 region of SEQ ID NO: 9, a CDRH2 region of SEQ ID NO:
16 and a CDRH3 region of SEQ ID NO: 11, or
d) a CDRH1 region of SEQ ID NO: 9, a CDRH2 region of SEQ ID NO:
17 and a CDRH3 region of SEQ ID NO: 11.
The antibody according to any one of claims 1 to 3, characterized in that the light chain variable domain comprises as CDRs a) a CDRL 1 region of SEQ ID NO : 20, a CDRL2 region of SEQ ID NO : 6 and a CDRL3 region of SEQ ID NO:7,
b) a CDRLl region of SEQ ID NO: 13, a CDRL2 region of SEQ ID NO : 14 a CDRL3 region of SEQ ID NO: 15,
c) a CDRLl region of SEQ ID NO: 13, a CDRL2 region of SEQ ID NO : 18 and a CDRL3 region of SEQ ID NO : 15 ,
d) a CDRLl region of SEQ ID NO: 19, a CDRL2 region of SEQ ID NO : 14 and a CDRL3 region of SEQ ID NO: 15, or
e) a CDRLl region of SEQ ID NO: 20, a CDRL2 region of SEQ ID NO:
14 and a CDRL3 region of SEQ ID NO: 15.
The antibody according to any one of claims 1 to 4, characterized in that the heavy chain variable domain and the light chain variable domain comprises as CDRs a) a CDRH1 region of SEQ ID NO: 2, a CDRH2 region of SEQ ID NO: 3 and a CDRH3 region of SEQ ID NO: 4, and a CDRL1 region of SEQ ID NO: 20, a CDRL2 region of SEQ ID NO: 6 and a CDRL3 region of SEQ ID NO:7, or
b) a CDRH1 region of SEQ ID NO: 9, a CDRH2 region of SEQ ID NO:
10 and a CDRH3 region of SEQ ID NO: 11, and a CDRLl region of SEQ ID NO: 13, a CDRL2 region of SEQ ID NO: 14 a CDRL3 region of SEQ ID NO: 15.
The antibody according to any one of claims 1 to 5, characterized in that the antibody comprises as heavy chain variable domain a) the heavy chain variable domain of SEQ ID NO: 1 ,
b) the heavy chain variable domain of SEQ ID NO: 8,
c) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A,
d) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A and as CDRH2 SEQ ID NO: 16
e) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A and as CDRH2 SEQ ID NO: 17,
f) the heavy chain variable domain of SEQ ID NO: 8 including mutations T28A, Vl lL and K12V, or
g) the heavy chain variable domain of SEQ ID NO: 8 including mutations T28A, VI 1L, K12V, Y91F, and T108S.
The antibody according to any one of claims 1 to 6, characterized in that the antibody comprises as light chain variable domain a) the light chain variable domain of SEQ ID NO:5,
b) the light chain variable domain of SEQ ID NO: 12,
c) the light chain variable domain of SEQ ID NO: 12 including as CDRL2 SEQ ID NO : 18 instead of SEQ ID NO : 14
d) the light chain variable domain of SEQ ID NO: 12 including as CDRLl SEQ ID NO : 19 instead of SEQ ID NO : 13 ,
e) the light chain variable domain of SEQ ID NO: 12 including as CDRLl SEQ ID NO:20 instead of SEQ ID NO: 13, or f) the light chain variable domain of SEQ ID NO: 12 including mutations P80S and F83L and as CDRL1 SEQ ID NO: 20 instead of SEQ ID NO: 13.
The antibody according to any one of claims 1 to 7, characterized in that the antibody comprises as heavy chain variable domain and as light chain variable domain a) the heavy chain variable domain of SEQ ID NO:l and the light chain variable domain of SEQ ID NO:5,
b) the heavy chain variable domain of SEQ ID NO: 8 and the light chain variable domain of SEQ ID NO: 12,
c) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A and the light chain variable domain of SEQ ID NO: 12, d) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A and as CDRH2 SEQ ID NO: 16 or 17, and the light chain variable domain of SEQ ID NO: 12,
e) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A and as CDRH2 SEQ ID NO: 16 or 17, and the light chain variable domain of SEQ ID NO: 12, and as CDRL1 SEQ ID NO: 19 or 20, f) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A and as CDRH2 SEQ ID NO: 16 or 17, and the light chain variable domain of SEQ ID NO: 12, and as CDRL2 SEQ ID NO: 18, or g) the heavy chain variable domain of SEQ ID NO: 8 including mutation T28A and as CDRH2 SEQ ID NO: 16 or 17, and the light chain variable domain of SEQ ID NO: 12 including mutations P80S and F83L, and as CDRLl SEQ ID NO: 20.
The antibody according to any one of claims 1 to 8, characterized in that the heavy chain variable domain comprises in addition to mutation T28A, a) mutations Vl lL and K12V, or
b) mutations VI 1L, K12V, Y91F and T108S.
The antibody according to any one of claims 1 to 9, characterized in being of human IgGl isotype comprising mutations L234A, which is alanine instead of leucine at amino acid position 234, and L235A, which is alanine instead of leucine at amino acid position 235.
11. The antibody according to any one of claims 1 to 9, characterized in being of human IgG4 isotype with or without mutation S228P.
12. Isolated nucleic acid encoding the antibody according to any one of claims 1 to 11.
13. A host cell comprising the nucleic acid of claim 12.
14. Method for the production of a recombinant antibody binding to CD19, characterized by expressing a nucleic acid according to claim 12 in a prokaryotic or eukaryotic host cell and recovering said antibody from said cell or the cell culture supernatant.
15. A pharmaceutical formulation comprising the antibody according to any one of claims 1 to 11 and a pharmaceutically acceptable carrier.
16. The antibody of according to any one of claims 1 to 11 for use as a medicament.
17. The antibody of according to any one of claims 1 to 11 for use in treating inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases.
18. Use of the antibody according to any one of claims 1 to 11 in the manufacture of a medicament.
19. The use of claim 18, wherein the medicament is for treating inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone diseases.
20. The use of claim 18, wherein the medicament is for blocking B-cell activation.
21. A method of treating an individual having inflammatory, autoimmune, rheumatoid arthritis, lupus, psoriasis, or bone disease comprising administering to the individual an effective amount of the antibody of any one of claims 1 to 11.
22. The antibody according to anyone of claims 1 to 9, characterized in being partially- fucosylated or non-fucosylated.
23. A pharmaceutical formulation comprising the antibody according to claim 22 and a pharmaceutically acceptable carrier.
24. The antibody of according to claim 22 for use as a medicament.
25. The antibody of according to claim 22 for use in treating cancer diseases.
26. Use of the antibody according to claim 22 in the manufacture of a medicament.
27. The use of claim 26, wherein the medicament is for treating cancer diseases.
28. A method of treating an individual having a cancer disease comprising administering to the individual an effective amount of the antibody of claim 22.
PCT/EP2011/058482 2010-05-26 2011-05-24 Antibodies against cd19 and uses thereof WO2011147834A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP10163921.9 2010-05-26
EP10163921 2010-05-26

Publications (1)

Publication Number Publication Date
WO2011147834A1 true WO2011147834A1 (en) 2011-12-01

Family

ID=43447023

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2011/058482 WO2011147834A1 (en) 2010-05-26 2011-05-24 Antibodies against cd19 and uses thereof

Country Status (1)

Country Link
WO (1) WO2011147834A1 (en)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013050335A1 (en) 2011-10-05 2013-04-11 F. Hoffmann-La Roche Ag Process for antibody g1 glycoform production
WO2016189014A1 (en) 2015-05-26 2016-12-01 Morphosys Ag Combination of an anti-cd19 antibody and a bruton's tyrosine kinase inhibitor and uses thereof
WO2017032679A1 (en) 2015-08-21 2017-03-02 Morphosys Ag Combinations and uses thereof
WO2017055314A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Bispecific anti-cd19xcd3 t cell activating antigen binding molecules
WO2017055328A1 (en) * 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Anti-human cd19 antibodies with high affinity
EP3243836A1 (en) 2016-05-11 2017-11-15 F. Hoffmann-La Roche AG C-terminally fused tnf family ligand trimer-containing antigen binding molecules
WO2017207574A1 (en) 2016-05-30 2017-12-07 Morphosys Ag Methods for predicting therapeutic benefit of anti-cd19 therapy in patients
WO2018002031A1 (en) 2016-06-27 2018-01-04 Morphosys Ag Anti-cd19 antibody formulations
WO2018078123A1 (en) 2016-10-28 2018-05-03 Morphosys Ag Combination of anti cd19 antibody with a bcl-2 inhibitor and uses thereof
EP3489256A1 (en) 2014-11-14 2019-05-29 F. Hoffmann-La Roche AG Antigen binding molecules comprising a tnf family ligand trimer
WO2020141145A1 (en) 2018-12-30 2020-07-09 F. Hoffmann-La Roche Ag Anti-rabbit cd19 antibodies and methods of use
WO2020225196A1 (en) 2019-05-03 2020-11-12 Morphosys Ag Anti-cd19 therapy in patients having a limited number of nk cells
EP3747472A1 (en) 2015-09-15 2020-12-09 Acerta Pharma B.V. Therapeutic combinations of a cd19 inhibitor and a btk inhibitor
WO2021067598A1 (en) 2019-10-04 2021-04-08 Ultragenyx Pharmaceutical Inc. Methods for improved therapeutic use of recombinant aav
WO2021084062A1 (en) 2019-10-31 2021-05-06 Morphosys Ag Anti-cd19 therapy in combination with lenalidomide for the treatment of leukemia or lymphoma
WO2021084063A1 (en) 2019-10-31 2021-05-06 Morphosys Ag Anti-tumor combination therapy comprising anti-cd19 antibody and gamma delta t-cells
WO2021259902A1 (en) 2020-06-22 2021-12-30 Morphosys Ag Anti-tumor combination therapy comprising anti-cd19 antibody and polypeptides blocking the sirpα-cd47 innate immune checkpoint
US11286300B2 (en) 2015-10-01 2022-03-29 Hoffmann-La Roche Inc. Humanized anti-human CD19 antibodies and methods of use
WO2022076573A1 (en) 2020-10-06 2022-04-14 Xencor, Inc. Biomarkers, methods, and compositions for treating autoimmune disease including systemic lupus erythematous (sle)
WO2022117799A2 (en) 2020-12-04 2022-06-09 Morphosys Ag Anti-cd19 combination therapy
WO2023073645A1 (en) 2021-10-29 2023-05-04 Takeda Pharmaceutical Company Limited Therapy comprising anti-cd19 antibody and sumo-activating enzyme inhibitor
WO2024038115A1 (en) 2022-08-17 2024-02-22 Morphosys Ag Therapy comprising anti-cd19 antibody and ezh2 modulators

Citations (81)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US4737456A (en) 1985-05-09 1988-04-12 Syntex (U.S.A.) Inc. Reducing interference in ligand-receptor binding assays
EP0307434A1 (en) 1987-03-18 1989-03-22 Medical Res Council Altered antibodies.
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
EP0404097A2 (en) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Bispecific and oligospecific, mono- and oligovalent receptors, production and applications thereof
WO1993001161A1 (en) 1991-07-11 1993-01-21 Pfizer Limited Process for preparing sertraline intermediates
WO1993008829A1 (en) 1991-11-04 1993-05-13 The Regents Of The University Of California Compositions that mediate killing of hiv-infected cells
WO1993016185A2 (en) 1992-02-06 1993-08-19 Creative Biomolecules, Inc. Biosynthetic binding protein for cancer marker
WO1994029351A2 (en) 1993-06-16 1994-12-22 Celltech Limited Antibodies
US5500362A (en) 1987-01-08 1996-03-19 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
US5571894A (en) 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
US5648237A (en) 1991-09-19 1997-07-15 Genentech, Inc. Expression of functional antibody fragments
WO1997030087A1 (en) 1996-02-16 1997-08-21 Glaxo Group Limited Preparation of glycosylated antibodies
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US5789199A (en) 1994-11-03 1998-08-04 Genentech, Inc. Process for bacterial production of polypeptides
US5821337A (en) 1991-06-14 1998-10-13 Genentech, Inc. Immunoglobulin variants
US5840523A (en) 1995-03-01 1998-11-24 Genetech, Inc. Methods and compositions for secretion of heterologous polypeptides
WO1998058964A1 (en) 1997-06-24 1998-12-30 Genentech, Inc. Methods and compositions for galactosylated glycoproteins
US5869046A (en) 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
WO1999022764A1 (en) 1997-10-31 1999-05-14 Genentech, Inc. Methods and compositions comprising glycoprotein glycoforms
US5959177A (en) 1989-10-27 1999-09-28 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
WO1999051642A1 (en) 1998-04-02 1999-10-14 Genentech, Inc. Antibody variants and fragments thereof
WO1999054342A1 (en) 1998-04-20 1999-10-28 Pablo Umana Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
US6040498A (en) 1998-08-11 2000-03-21 North Caroline State University Genetically engineered duckweed
WO2000061739A1 (en) 1999-04-09 2000-10-19 Kyowa Hakko Kogyo Co., Ltd. Method for controlling the activity of immunologically functional molecule
US6171586B1 (en) 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
WO2001029246A1 (en) 1999-10-19 2001-04-26 Kyowa Hakko Kogyo Co., Ltd. Process for producing polypeptide
US6248516B1 (en) 1988-11-11 2001-06-19 Medical Research Council Single domain ligands, receptors comprising said ligands methods for their production, and use of said ligands and receptors
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
WO2002031140A1 (en) 2000-10-06 2002-04-18 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions
US6420548B1 (en) 1999-10-04 2002-07-16 Medicago Inc. Method for regulating transcription of foreign genes
US20020164328A1 (en) 2000-10-06 2002-11-07 Toyohide Shinkawa Process for purifying antibody
WO2003011878A2 (en) 2001-08-03 2003-02-13 Glycart Biotechnology Ag Antibody glycosylation variants having increased antibody-dependent cellular cytotoxicity
WO2003048209A1 (en) 2001-11-14 2003-06-12 Affimed Therapeutics Ag Bispecific anti-cd19 x anti-cd16 antibodies and uses thereof
US20030115614A1 (en) 2000-10-06 2003-06-19 Yutaka Kanda Antibody composition-producing cell
US20030157108A1 (en) 2001-10-25 2003-08-21 Genentech, Inc. Glycoprotein compositions
WO2003085107A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. Cells with modified genome
WO2003085119A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. METHOD OF ENHANCING ACTIVITY OF ANTIBODY COMPOSITION OF BINDING TO FcϜ RECEPTOR IIIa
WO2003084570A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. DRUG CONTAINING ANTIBODY COMPOSITION APPROPRIATE FOR PATIENT SUFFERING FROM FcϜRIIIa POLYMORPHISM
US20040093621A1 (en) 2001-12-25 2004-05-13 Kyowa Hakko Kogyo Co., Ltd Antibody composition which specifically binds to CD20
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US20040110282A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Cells in which activity of the protein involved in transportation of GDP-fucose is reduced or lost
US20040109865A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Antibody composition-containing medicament
WO2004056312A2 (en) 2002-12-16 2004-07-08 Genentech, Inc. Immunoglobulin variants and uses thereof
US20040132140A1 (en) 2002-04-09 2004-07-08 Kyowa Hakko Kogyo Co., Ltd. Production process for antibody composition
WO2004106381A1 (en) 2003-05-31 2004-12-09 Micromet Ag Pharmaceutical compositions comprising bispecific anti-cd3, anti-cd19 antibody constructs for the treatment of b-cell related disorders
US20050014934A1 (en) 2002-10-15 2005-01-20 Hinton Paul R. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
WO2005012493A2 (en) 2003-07-31 2005-02-10 Immunomedics, Inc. Anti-cd19 antibodies
WO2005035586A1 (en) 2003-10-08 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. Fused protein composition
WO2005035778A1 (en) 2003-10-09 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. PROCESS FOR PRODUCING ANTIBODY COMPOSITION BY USING RNA INHIBITING THE FUNCTION OF α1,6-FUCOSYLTRANSFERASE
US20050123546A1 (en) 2003-11-05 2005-06-09 Glycart Biotechnology Ag Antigen binding molecules with increased Fc receptor binding affinity and effector function
WO2005053742A1 (en) 2003-12-04 2005-06-16 Kyowa Hakko Kogyo Co., Ltd. Medicine containing antibody composition
WO2005100402A1 (en) 2004-04-13 2005-10-27 F.Hoffmann-La Roche Ag Anti-p-selectin antibodies
US20050260186A1 (en) 2003-03-05 2005-11-24 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
US20060025576A1 (en) 2000-04-11 2006-02-02 Genentech, Inc. Multivalent antibodies and uses therefor
WO2006029879A2 (en) 2004-09-17 2006-03-23 F.Hoffmann-La Roche Ag Anti-ox40l antibodies
WO2006044908A2 (en) 2004-10-20 2006-04-27 Genentech, Inc. Antibody formulation in histidine-acetate buffer
US20060104968A1 (en) 2003-03-05 2006-05-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminogly ycanases
WO2006089133A2 (en) 2005-02-15 2006-08-24 Duke University Anti-cd19 antibodies and uses in oncology
US7125978B1 (en) 1999-10-04 2006-10-24 Medicago Inc. Promoter for regulating expression of foreign genes
WO2006121852A2 (en) 2005-05-05 2006-11-16 Duke University Anti-cd19 antibody therapy for autoimmune disease
WO2006133450A2 (en) 2005-06-08 2006-12-14 Duke University Anti-cd19 antibody therapy for the transplantation
WO2007002223A2 (en) 2005-06-20 2007-01-04 Medarex, Inc. Cd19 antibodies and their uses
WO2008022152A2 (en) 2006-08-14 2008-02-21 Xencor, Inc. Optimized antibodies that target cd19
US20080069820A1 (en) 2006-08-30 2008-03-20 Genentech, Inc. Multispecific antibodies
US7371826B2 (en) 1999-01-15 2008-05-13 Genentech, Inc. Polypeptide variants with altered effector function
US20080138336A1 (en) 2006-09-08 2008-06-12 Medlmmune, Inc. Humanized Anti-CD19 Antibodies And Their Use In Treatment Of Oncology, Transplantation And Autoimmune Disease
WO2008077546A1 (en) 2006-12-22 2008-07-03 F. Hoffmann-La Roche Ag Antibodies against insulin-like growth factor i receptor and uses thereof
US7521541B2 (en) 2004-09-23 2009-04-21 Genetech Inc. Cysteine engineered antibodies and conjugates
WO2009080254A1 (en) 2007-12-21 2009-07-02 F. Hoffmann-La Roche Ag Bivalent, bispecific antibodies
WO2009080253A1 (en) 2007-12-21 2009-07-02 F. Hoffmann-La Roche Ag Bivalent, bispecific antibodies
WO2009080252A1 (en) 2007-12-21 2009-07-02 F. Hoffmann-La Roche Ag Bivalent, bispecific antibodies
WO2009080251A1 (en) 2007-12-21 2009-07-02 F. Hoffmann-La Roche Ag Bivalent, bispecific antibodies
WO2009089004A1 (en) 2008-01-07 2009-07-16 Amgen Inc. Method for making antibody fc-heterodimeric molecules using electrostatic steering effects
WO2010112193A1 (en) 2009-04-02 2010-10-07 Roche Glycart Ag Multispecific antibodies comprising full length antibodies and single chain fab fragments
WO2010115589A1 (en) 2009-04-07 2010-10-14 Roche Glycart Ag Trivalent, bispecific antibodies
WO2010136172A1 (en) 2009-05-27 2010-12-02 F. Hoffmann-La Roche Ag Tri- or tetraspecific antibodies
WO2010145793A1 (en) 2009-06-18 2010-12-23 F. Hoffmann-La Roche Ag Bispecific, tetravalent antigen binding proteins
WO2010145792A1 (en) 2009-06-16 2010-12-23 F. Hoffmann-La Roche Ag Bispecific antigen binding proteins

Patent Citations (95)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4737456A (en) 1985-05-09 1988-04-12 Syntex (U.S.A.) Inc. Reducing interference in ligand-receptor binding assays
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US5500362A (en) 1987-01-08 1996-03-19 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
EP0307434A1 (en) 1987-03-18 1989-03-22 Medical Res Council Altered antibodies.
US5648260A (en) 1987-03-18 1997-07-15 Scotgen Biopharmaceuticals Incorporated DNA encoding antibodies with altered effector functions
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US6248516B1 (en) 1988-11-11 2001-06-19 Medical Research Council Single domain ligands, receptors comprising said ligands methods for their production, and use of said ligands and receptors
EP0404097A2 (en) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Bispecific and oligospecific, mono- and oligovalent receptors, production and applications thereof
US6417429B1 (en) 1989-10-27 2002-07-09 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
US5959177A (en) 1989-10-27 1999-09-28 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
US5571894A (en) 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
US5821337A (en) 1991-06-14 1998-10-13 Genentech, Inc. Immunoglobulin variants
WO1993001161A1 (en) 1991-07-11 1993-01-21 Pfizer Limited Process for preparing sertraline intermediates
US5648237A (en) 1991-09-19 1997-07-15 Genentech, Inc. Expression of functional antibody fragments
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
WO1993008829A1 (en) 1991-11-04 1993-05-13 The Regents Of The University Of California Compositions that mediate killing of hiv-infected cells
WO1993016185A2 (en) 1992-02-06 1993-08-19 Creative Biomolecules, Inc. Biosynthetic binding protein for cancer marker
WO1994029351A2 (en) 1993-06-16 1994-12-22 Celltech Limited Antibodies
US5789199A (en) 1994-11-03 1998-08-04 Genentech, Inc. Process for bacterial production of polypeptides
US5840523A (en) 1995-03-01 1998-11-24 Genetech, Inc. Methods and compositions for secretion of heterologous polypeptides
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US5869046A (en) 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
WO1997030087A1 (en) 1996-02-16 1997-08-21 Glaxo Group Limited Preparation of glycosylated antibodies
US6171586B1 (en) 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
WO1998058964A1 (en) 1997-06-24 1998-12-30 Genentech, Inc. Methods and compositions for galactosylated glycoproteins
WO1999022764A1 (en) 1997-10-31 1999-05-14 Genentech, Inc. Methods and compositions comprising glycoprotein glycoforms
WO1999051642A1 (en) 1998-04-02 1999-10-14 Genentech, Inc. Antibody variants and fragments thereof
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
WO1999054342A1 (en) 1998-04-20 1999-10-28 Pablo Umana Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
US6602684B1 (en) 1998-04-20 2003-08-05 Glycart Biotechnology Ag Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
US6040498A (en) 1998-08-11 2000-03-21 North Caroline State University Genetically engineered duckweed
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US7371826B2 (en) 1999-01-15 2008-05-13 Genentech, Inc. Polypeptide variants with altered effector function
US7332581B2 (en) 1999-01-15 2008-02-19 Genentech, Inc. Polypeptide variants with altered effector function
WO2000061739A1 (en) 1999-04-09 2000-10-19 Kyowa Hakko Kogyo Co., Ltd. Method for controlling the activity of immunologically functional molecule
US6420548B1 (en) 1999-10-04 2002-07-16 Medicago Inc. Method for regulating transcription of foreign genes
US7125978B1 (en) 1999-10-04 2006-10-24 Medicago Inc. Promoter for regulating expression of foreign genes
WO2001029246A1 (en) 1999-10-19 2001-04-26 Kyowa Hakko Kogyo Co., Ltd. Process for producing polypeptide
US20060025576A1 (en) 2000-04-11 2006-02-02 Genentech, Inc. Multivalent antibodies and uses therefor
WO2002031140A1 (en) 2000-10-06 2002-04-18 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions
US20020164328A1 (en) 2000-10-06 2002-11-07 Toyohide Shinkawa Process for purifying antibody
US20030115614A1 (en) 2000-10-06 2003-06-19 Yutaka Kanda Antibody composition-producing cell
US6946292B2 (en) 2000-10-06 2005-09-20 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions with increased antibody dependent cytotoxic activity
WO2003011878A2 (en) 2001-08-03 2003-02-13 Glycart Biotechnology Ag Antibody glycosylation variants having increased antibody-dependent cellular cytotoxicity
US20030157108A1 (en) 2001-10-25 2003-08-21 Genentech, Inc. Glycoprotein compositions
WO2003048209A1 (en) 2001-11-14 2003-06-12 Affimed Therapeutics Ag Bispecific anti-cd19 x anti-cd16 antibodies and uses thereof
US20040093621A1 (en) 2001-12-25 2004-05-13 Kyowa Hakko Kogyo Co., Ltd Antibody composition which specifically binds to CD20
WO2003085107A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. Cells with modified genome
US20040110282A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Cells in which activity of the protein involved in transportation of GDP-fucose is reduced or lost
US20040109865A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Antibody composition-containing medicament
US20040110704A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Cells of which genome is modified
WO2003085119A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. METHOD OF ENHANCING ACTIVITY OF ANTIBODY COMPOSITION OF BINDING TO FcϜ RECEPTOR IIIa
WO2003084570A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. DRUG CONTAINING ANTIBODY COMPOSITION APPROPRIATE FOR PATIENT SUFFERING FROM FcϜRIIIa POLYMORPHISM
US20040132140A1 (en) 2002-04-09 2004-07-08 Kyowa Hakko Kogyo Co., Ltd. Production process for antibody composition
US20050014934A1 (en) 2002-10-15 2005-01-20 Hinton Paul R. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
WO2004056312A2 (en) 2002-12-16 2004-07-08 Genentech, Inc. Immunoglobulin variants and uses thereof
US20050260186A1 (en) 2003-03-05 2005-11-24 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
US20060104968A1 (en) 2003-03-05 2006-05-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminogly ycanases
EP1629012A1 (en) 2003-05-31 2006-03-01 Micromet AG Pharmaceutical compositions comprising bispecific anti-cd3, anti-cd19 antibody constructs for the treatment of b-cell related disorders
WO2004106381A1 (en) 2003-05-31 2004-12-09 Micromet Ag Pharmaceutical compositions comprising bispecific anti-cd3, anti-cd19 antibody constructs for the treatment of b-cell related disorders
EP1648512A2 (en) 2003-07-31 2006-04-26 Immunomedics, Inc. Anti-cd19 antibodies
WO2005012493A2 (en) 2003-07-31 2005-02-10 Immunomedics, Inc. Anti-cd19 antibodies
US20060257398A1 (en) 2003-07-31 2006-11-16 Immunomedics, Inc. Anti-CD19 antibodies
US7109304B2 (en) 2003-07-31 2006-09-19 Immunomedics, Inc. Humanized anti-CD19 antibodies
WO2005035586A1 (en) 2003-10-08 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. Fused protein composition
WO2005035778A1 (en) 2003-10-09 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. PROCESS FOR PRODUCING ANTIBODY COMPOSITION BY USING RNA INHIBITING THE FUNCTION OF α1,6-FUCOSYLTRANSFERASE
US20050123546A1 (en) 2003-11-05 2005-06-09 Glycart Biotechnology Ag Antigen binding molecules with increased Fc receptor binding affinity and effector function
WO2005053742A1 (en) 2003-12-04 2005-06-16 Kyowa Hakko Kogyo Co., Ltd. Medicine containing antibody composition
WO2005100402A1 (en) 2004-04-13 2005-10-27 F.Hoffmann-La Roche Ag Anti-p-selectin antibodies
WO2006029879A2 (en) 2004-09-17 2006-03-23 F.Hoffmann-La Roche Ag Anti-ox40l antibodies
US7521541B2 (en) 2004-09-23 2009-04-21 Genetech Inc. Cysteine engineered antibodies and conjugates
WO2006044908A2 (en) 2004-10-20 2006-04-27 Genentech, Inc. Antibody formulation in histidine-acetate buffer
WO2006089133A2 (en) 2005-02-15 2006-08-24 Duke University Anti-cd19 antibodies and uses in oncology
US20060233791A1 (en) 2005-02-15 2006-10-19 Duke University Anti-CD19 antibodies and uses in oncology
WO2006121852A2 (en) 2005-05-05 2006-11-16 Duke University Anti-cd19 antibody therapy for autoimmune disease
US20060263357A1 (en) 2005-05-05 2006-11-23 Tedder Thomas F Anti-CD19 antibody therapy for autoimmune disease
WO2006133450A2 (en) 2005-06-08 2006-12-14 Duke University Anti-cd19 antibody therapy for the transplantation
US20060280738A1 (en) 2005-06-08 2006-12-14 Tedder Thomas F Anti-CD19 antibody therapy for transplantation
WO2007002223A2 (en) 2005-06-20 2007-01-04 Medarex, Inc. Cd19 antibodies and their uses
WO2008022152A2 (en) 2006-08-14 2008-02-21 Xencor, Inc. Optimized antibodies that target cd19
US20080069820A1 (en) 2006-08-30 2008-03-20 Genentech, Inc. Multispecific antibodies
US20080138336A1 (en) 2006-09-08 2008-06-12 Medlmmune, Inc. Humanized Anti-CD19 Antibodies And Their Use In Treatment Of Oncology, Transplantation And Autoimmune Disease
WO2008077546A1 (en) 2006-12-22 2008-07-03 F. Hoffmann-La Roche Ag Antibodies against insulin-like growth factor i receptor and uses thereof
WO2009080254A1 (en) 2007-12-21 2009-07-02 F. Hoffmann-La Roche Ag Bivalent, bispecific antibodies
WO2009080253A1 (en) 2007-12-21 2009-07-02 F. Hoffmann-La Roche Ag Bivalent, bispecific antibodies
WO2009080252A1 (en) 2007-12-21 2009-07-02 F. Hoffmann-La Roche Ag Bivalent, bispecific antibodies
WO2009080251A1 (en) 2007-12-21 2009-07-02 F. Hoffmann-La Roche Ag Bivalent, bispecific antibodies
WO2009089004A1 (en) 2008-01-07 2009-07-16 Amgen Inc. Method for making antibody fc-heterodimeric molecules using electrostatic steering effects
WO2010112193A1 (en) 2009-04-02 2010-10-07 Roche Glycart Ag Multispecific antibodies comprising full length antibodies and single chain fab fragments
WO2010115589A1 (en) 2009-04-07 2010-10-14 Roche Glycart Ag Trivalent, bispecific antibodies
WO2010136172A1 (en) 2009-05-27 2010-12-02 F. Hoffmann-La Roche Ag Tri- or tetraspecific antibodies
WO2010145792A1 (en) 2009-06-16 2010-12-23 F. Hoffmann-La Roche Ag Bispecific antigen binding proteins
WO2010145793A1 (en) 2009-06-18 2010-12-23 F. Hoffmann-La Roche Ag Bispecific, tetravalent antigen binding proteins

Non-Patent Citations (98)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 1980
ANGAL, S. ET AL., MOL. IMMUNOL., vol. 30, 1993, pages 105 - 108
ARMOUR, K.L. ET AL., EUR. J. IMMUNOL., vol. 29, 1999, pages 2613 - 2624
BEJCEK, B.E. ET AL., CANCER RES., vol. 55, 1995, pages 2346 - 2351
BRENNAN, M. ET AL., SCIENCE, vol. 229, 1985, pages 81 - 83
BRUENKE, J. ET AL., BR. J. HEMATOL., vol. 130, 2005, pages 218 - 228
BRUGGEMANN, M. ET AL., J. EXP. MED., vol. 166, 1987, pages 1351 - 1361
BRUNHOUSE, R., CEBRA, J.J., MOL. IMMUNOL., vol. 16, 1979, pages 907 - 917
BURTON, D.R. ET AL., NATURE, vol. 288, 1980, pages 338 - 344
BURTON, D.R., MOL. IMMUNOL., vol. 22, 1985, pages 161 - 206
CAPEL, P.J. ET AL., IMMUNOMETHODS, vol. 4, 1994, pages 25 - 34
CARTER, R.H. ET AL., IMMUNOL. RES., vol. 26, 2002, pages 45 - 54
CHARLTON, K.A: "Methods in Molecular Biology", vol. 248, 2004, HUMANA PRESS, pages: 245 - 254
CHEN, Y. ET AL., J. MOL. BIOL., vol. 293, 1999, pages 865 - 881
CLACKSON, T. ET AL., NATURE, vol. 352, 1991, pages 624 - 628
CLYNES, R. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 95, 1998, pages 652 - 656
CONRY, R.M. ET AL., J. IMMUNOTHER. EMPHASIS TUMOR IMMUNOL., vol. 18, 1995, pages 231 - 241
CRAGG, M.S. ET AL., BLOOD, vol. 101, 2003, pages 1045 - 1052
CRAGG, M.S., M.J., GLENNIE, BLOOD, vol. 103, 2004, pages 2738 - 2743
DAVIES, J. ET AL., BIOTECHNOL. BIOENG., vol. 74, 2001, pages 288 - 294
DE HAAS, M. ET AL., J. LAB. CLIN. MED., vol. 126, 1995, pages 330 - 341
DUNCAN, A.R., WINTER, G., NATURE, vol. 332, 1988, pages 738 - 740
EBERL, G. ET AL., CLIN. EXP. IMMUNOL., vol. 114, 1998, pages 173 - 178
FLATMAN, S. ET AL., J. CHROMATOGR., vol. B 848, 2007, pages 79 - 87
GAZZANO-SANTORO, H. ET AL., J. IMMUNOL. METHODS, vol. 202, 1997, pages 163 - 171
GERNGROSS, NAT. BIOTECH., vol. 22, 2004, pages 1409 - 1414
GESSNER, J.E. ET AL., ANN. HEMATOL., vol. 76, 1998, pages 231 - 248
GHETIE, M.A. ET AL., BLOOD, vol. 104, 2004, pages 178 - 183
GRAHAM. F.L. ET AL., J. GEN VIROL., vol. 36, 1977, pages 59 - 74
GRUBER, M. ET AL., J. IMMUNOL., vol. 152, 1994, pages 5368 - 5374
GUYER, R.L. ET AL., J. IMMUNOL., vol. 117, 1976, pages 587 - 593
HAGEN, I.A. ET AL., BLOOD, vol. 85, 1995, pages 3208 - 3212
HEKMAN, A. ET AL., CANCER IMMUNOL. IMMUNOTHER., vol. 32, no. 191, pages 364 - 372
HELLSTROM, I. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 82, 1985, pages 1499 - 1502
HELLSTROM, I. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 83, 1986, pages 7059 - 7063
HEZAREH, M. ET AL., J. VIROL., vol. 75, 2001, pages 12161 - 12168
HOLLIGER, P. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 6444 - 6448
HOLLINGER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 6444 - 6448
HUDSON ET AL., NAT. MED., vol. 9, 2003, pages 129 - 134
IDUSOGIE E.E. ET AL., J. IMMUNOL., vol. 164, 2000, pages 4178 - 4184
IDUSOGIE, E.E. ET AL., J. IMMUNOL., vol. 164, 2000, pages 4178 - 4184
JEFFERIS, R. ET AL., IMMUNOL. REV., vol. 163, 1998, pages 59 - 76
KABAT, E.A. ET AL.: "Sequences of Proteins of Immunological Interest", 1991, NATIONAL INSTITUTES OF HEALTH
KABAT, E.A. ET AL.: "Sequences of Proteins of Immunological Interest", vol. 1-3, 1991, NIH PUBLICATION 91-3242
KAM, N.W. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 102, 2005, pages 11600 - 11605
KANDA, Y. ET AL., BIOTECHNOL. BIOENG., vol. 94, no. 4, 2006, pages 680 - 688
KIM, J.K. ET AL., J. EUROPEAN J OF IMMUNOL., vol. 24, 1994, pages 2429 - 2434
KINDT ET AL.: "Kuby Immunology", 2007, W.H. FREEMAN AND CO., pages: 91
KOSTELNY, S.A. ET AL., J. IMMUNOL., vol. 148, no. 5, 1992, pages 1547 - 1553
LANG, P. ET AL., BLOOD, vol. 103, 2004, pages 3982 - 3985
LE GALL, F. ET AL., FEBS LETT., vol. 453, 1999, pages 164 - 168
LI ET AL., NAT. BIOTECH., vol. 24, 2006, pages 210 - 215
LI, Q. ET AL., CANCER IMMUNOL. IMMUNOTHER., vol. 47, 1998, pages 121 - 130
LIFELY, M.R. ET AL., GLYCOBIOLOGY, vol. 5, 1995, pages 813 - 822
LOEFFLER, A. ET AL., BLOOD, vol. 95, 2000, pages 2098 - 2103
LUKAS, T.J. ET AL., J. IMMUNOL., vol. 127, 1981, pages 2555 - 2560
LUND, J. ET AL., FASEB J., vol. 9, 1995, pages 115 - 119
MANZKE, 0. ET AL., INT. J. CANCER, vol. 91, 2001, pages 516 - 522
MATHER, J.P. ET AL., ANNALS N.Y. ACAD. SCI., vol. 383, 1982, pages 44 - 68
MATHER, J.P., BIOL. REPROD., vol. 23, 1980, pages 243 - 252
MILSTEIN, C., CUELLO, NATURE, vol. 305, 1983, pages 537 - 540
MIMURA, Y. ET AL., J. BIOL. CHEM., vol. 276, 2001, pages 45539 - 45547
MOREA, V. ET AL., METHODS, vol. 20, no. 3, 2000, pages 267 - 279
MORGAN, A. ET AL., IMMUNOLOGY, vol. 86, 1995, pages 319 - 324
MYERS, D.E. ET AL., LEUK LYMPHOMA, vol. 18, 1995, pages 93 - 102
OKAZAKI ET AL., J. MOL. BIOL., vol. 336, 2004, pages 1239 - 1249
PETKOVA, S.B. ET AL., INT. IMMUNOL., vol. 18, 2006, pages 1759 - 1769
PEZZUTTO, A. ET AL., J IMMUNOL., vol. 138, 1987, pages 2793 - 2799
PIETERSZ, G.A. ET AL., CANCER IMMUNOL. IMMUNOTHER., vol. 41, 1995, pages 53 - 60
PLUCKTHUN: "The Pharmacology of Monoclonal Antibodies", vol. 113, 1994, SPRINGER-VERLAG, pages: 269 - 315
POTOLANO, S. ET AL., J. IMMUNOL., vol. 150, 1993, pages 880 - 887
RADAEV, S. ET AL., J. BIOL. CHEM., vol. 276, 2001, pages 16478 - 16483
RAJU, T.S., BIOPROCESS INT., vol. 1, 2003, pages 44 - 53
RAVETCH, J.V., BOLLAND, S., ANNU. REV. IMMUNOL., vol. 19, 2001, pages 275 - 290
RAVETCH, J.V., KINET, J.P., ANNU. REV. IMMUNOL., vol. 9, 1991, pages 457 - 492
RHODES, E.G. ET AL., BONE MARROW TRANSPLANT., vol. 10, 1992, pages 485 - 489
RIPKA, J. ET AL., ARCH. BIOCHEM. BIOPHYS., vol. 249, 1986, pages 533 - 545
ROUTIER, F.H., GLYCOCONJUGATE J., vol. 14, 1997, pages 201 - 207
SHIELDS, R.L. ET AL., J. BIOL. CHEM., vol. 276, 2001, pages 6591 - 6604
SHIELDS, R.L. ET AL., J. BIOL. CHEM., vol. 277, 2002, pages 26733 - 26740
SHIELDS, R.L. ET AL., J. BIOL. CHEM., vol. 9, 2001, pages 6591 - 6604
SIMMONS, L.C. ET AL., J. IMMUNOL. METHODS, vol. 263, 2002, pages 133 - 147
THOMMESEN, J.E. ET AL., MOL. IMMUNOL., vol. 37, 2000, pages 995 - 1004
TRAUNECKER, A ET AL., EMBO J., vol. 10, 1991, pages 3655 - 3659
TUTT, A. ET AL., J. IMMUNOL., vol. 147, 1991, pages 60 - 69
UCKUN, F.M. ET AL., BLOOD, vol. 71, 1988, pages 13 - 29
UMANA, P. ET AL., NATURE BIOTECHNOL., vol. 17, 1999, pages 176 - 180
URLAUB, G. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 77, 1980, pages 4216 - 4220
VALLERA, D.A. ET AL., CANCER BIOTHER. RADIOPHARM., vol. 19, 2004, pages 11 - 23
VAN DE WINKEL, J.G., ANDERSON, C.L., J. LEUKOC. BIOL., vol. 49, 1991, pages 511 - 524
VLASFELD, L.T. ET AL., CANCER IMMUNOL. IMMUNOTHER., vol. 40, 1995, pages 37 - 47
WARD, E.S., GHETIE, V., THER. IMMUNOL., vol. 2, 1995, pages 77 - 94
WATANABE, M. ET AL., CANCER RES., vol. 50, 1990, pages 3245 - 3248
WRIGHT ET AL., TIBTECH, vol. 15, 1997, pages 26 - 32
WRIGHT, A., MORRISON, S.L., TRENDS BIOTECHNOL., vol. 15, 1997, pages 26 - 32
YAMANE-OHNUKI, N. ET AL., BIOTECH. BIOENG., vol. 87, 2004, pages 614 - 622
YAZAKI, P.J., WU, A.M.: "Methods in Molecular Biology", vol. 248, 2004, HUMANA PRESS, pages: 255 - 268
ZOLA, H. ET AL., IMMUNOL. CELL BIOL., vol. 69, 1991, pages 411 - 422

Cited By (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013050335A1 (en) 2011-10-05 2013-04-11 F. Hoffmann-La Roche Ag Process for antibody g1 glycoform production
EP3489256A1 (en) 2014-11-14 2019-05-29 F. Hoffmann-La Roche AG Antigen binding molecules comprising a tnf family ligand trimer
WO2016189014A1 (en) 2015-05-26 2016-12-01 Morphosys Ag Combination of an anti-cd19 antibody and a bruton's tyrosine kinase inhibitor and uses thereof
US10617691B2 (en) 2015-05-26 2020-04-14 Morphosys Ag Combination of an anti-CD19 antibody and a Bruton's tyrosine kinase inhibitor and uses thereof
EP3603672A1 (en) 2015-05-26 2020-02-05 MorphoSys AG Combination of an anti-cd19 antibody and a bruton's tyrosine kinase inhibitor and uses thereof
WO2017032679A1 (en) 2015-08-21 2017-03-02 Morphosys Ag Combinations and uses thereof
US11224654B2 (en) 2015-08-21 2022-01-18 Morphosys Ag Combinations and uses thereof
EP3954388A1 (en) 2015-08-21 2022-02-16 MorphoSys AG Combinations and uses thereof
EP3747472A1 (en) 2015-09-15 2020-12-09 Acerta Pharma B.V. Therapeutic combinations of a cd19 inhibitor and a btk inhibitor
US11286300B2 (en) 2015-10-01 2022-03-29 Hoffmann-La Roche Inc. Humanized anti-human CD19 antibodies and methods of use
CN108137698B (en) * 2015-10-02 2022-04-19 豪夫迈·罗氏有限公司 Anti-human CD19 antibody
CN108137698A (en) * 2015-10-02 2018-06-08 豪夫迈·罗氏有限公司 Anti human CD 19 antibody with high-affinity
IL258271A (en) * 2015-10-02 2018-05-31 Hoffmann La Roche Anti-human cd19 antibodies with high affinity
AU2016330807B2 (en) * 2015-10-02 2023-08-10 F. Hoffmann-La Roche Ag Anti-human CD19 antibodies with high affinity
EP3913000A1 (en) 2015-10-02 2021-11-24 F. Hoffmann-La Roche AG Bispecific anti-cd19xcd3 t cell activating antigen binding molecules
WO2017055328A1 (en) * 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Anti-human cd19 antibodies with high affinity
WO2017055314A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Bispecific anti-cd19xcd3 t cell activating antigen binding molecules
WO2017194438A1 (en) 2016-05-11 2017-11-16 F. Hoffmann-La Roche Ag C-terminally fused tnf family ligand trimer-containing antigen binding molecules
EP3243836A1 (en) 2016-05-11 2017-11-15 F. Hoffmann-La Roche AG C-terminally fused tnf family ligand trimer-containing antigen binding molecules
WO2017207574A1 (en) 2016-05-30 2017-12-07 Morphosys Ag Methods for predicting therapeutic benefit of anti-cd19 therapy in patients
EP3916392A1 (en) 2016-05-30 2021-12-01 MorphoSys AG Methods for predicting therapeutic benefit of anti-cd19 therapy in patients
WO2018002031A1 (en) 2016-06-27 2018-01-04 Morphosys Ag Anti-cd19 antibody formulations
EP3909985A1 (en) 2016-06-27 2021-11-17 MorphoSys AG Anti-cd19 antibody formulations
EP3903821A1 (en) 2016-10-28 2021-11-03 MorphoSys AG Combination of anti cd19 antibody with a bcl-2 inhibitor and uses thereof
WO2018078123A1 (en) 2016-10-28 2018-05-03 Morphosys Ag Combination of anti cd19 antibody with a bcl-2 inhibitor and uses thereof
WO2020141145A1 (en) 2018-12-30 2020-07-09 F. Hoffmann-La Roche Ag Anti-rabbit cd19 antibodies and methods of use
WO2020225196A1 (en) 2019-05-03 2020-11-12 Morphosys Ag Anti-cd19 therapy in patients having a limited number of nk cells
WO2021067598A1 (en) 2019-10-04 2021-04-08 Ultragenyx Pharmaceutical Inc. Methods for improved therapeutic use of recombinant aav
WO2021084063A1 (en) 2019-10-31 2021-05-06 Morphosys Ag Anti-tumor combination therapy comprising anti-cd19 antibody and gamma delta t-cells
WO2021084062A1 (en) 2019-10-31 2021-05-06 Morphosys Ag Anti-cd19 therapy in combination with lenalidomide for the treatment of leukemia or lymphoma
WO2021259902A1 (en) 2020-06-22 2021-12-30 Morphosys Ag Anti-tumor combination therapy comprising anti-cd19 antibody and polypeptides blocking the sirpα-cd47 innate immune checkpoint
WO2022076573A1 (en) 2020-10-06 2022-04-14 Xencor, Inc. Biomarkers, methods, and compositions for treating autoimmune disease including systemic lupus erythematous (sle)
WO2022117799A2 (en) 2020-12-04 2022-06-09 Morphosys Ag Anti-cd19 combination therapy
WO2023073645A1 (en) 2021-10-29 2023-05-04 Takeda Pharmaceutical Company Limited Therapy comprising anti-cd19 antibody and sumo-activating enzyme inhibitor
WO2024038115A1 (en) 2022-08-17 2024-02-22 Morphosys Ag Therapy comprising anti-cd19 antibody and ezh2 modulators

Similar Documents

Publication Publication Date Title
WO2011147834A1 (en) Antibodies against cd19 and uses thereof
US20200354473A1 (en) Multispecific antigen-binding molecules having blood coagulation factor viii (fviii) cofactor function-substituting activity and pharmaceutical formulations containing such a molecule as an active ingredient
US8883975B2 (en) Antibodies against IL-18R1 and uses thereof
TWI780680B (en) Bispecific anti-human a-beta/human transferrin receptor antibodies and methods of use
AU2021202360A1 (en) Anti-TIGIT antibodies
ES2791989T3 (en) Humanized anti-human CD19 antibodies and procedures for use
TW202204415A (en) HUMANIZED AND AFFINITY MATURED ANTIBODIES TO FcRH5 AND METHODS OF USE
EA037554B1 (en) Anti-lag-3 binding proteins
EA036531B1 (en) Type ii anti-cd20 humanized antibody (variants), pharmaceutical composition comprising these antibody variants, and use thereof
NO338313B1 (en) Humanized antibody that binds human CD20, isolated nucleic acid or expression vector encoding the antibody, host cells, method for its preparation, its use in the treatment of an autoimmune disease, a CD20 positive cancer or to deplete B cells in vivo, and for preparation of a drug.
KR20110122859A (en) Humanized antibodies that bind to cd19 and their uses
US20240026005A1 (en) Anti-human pd-l2 antibodies
MX2013015061A (en) Anti-cxcr4 antibody with effector functions and its use for the treatment of cancer.
JP2017536101A (en) Antibodies that bind to CCR6 and uses thereof
CN114341181A (en) anti-MS 4A4A antibodies and methods of use thereof
WO2012010582A1 (en) Anti-cxcr5 antibodies and methods of use
US9187563B2 (en) Anti-human EPO receptor antibodies and methods of use
EP4301472A1 (en) Anti-vista constructs and uses thereof
US20220242944A1 (en) Antigen binding molecules that bind pdgf-b and pdgf-d and uses thereof
TW201326212A (en) Anti-MCSP antibodies
US11958902B2 (en) Anti-TIGIT antibodies
WO2023086989A1 (en) Transthyretin (ttr) monomer binding antibodies
US20240084005A1 (en) Anti-siglec-6 antibodies and methods of use thereof
WO2024054929A1 (en) Anti-vista constructs and uses thereof
TW201245227A (en) Antibody Fc variants

Legal Events

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

Ref document number: 11722789

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11722789

Country of ref document: EP

Kind code of ref document: A1