WO2003040170A9 - Antibodies to cd40 - Google Patents

Antibodies to cd40

Info

Publication number
WO2003040170A9
WO2003040170A9 PCT/US2002/036107 US0236107W WO03040170A9 WO 2003040170 A9 WO2003040170 A9 WO 2003040170A9 US 0236107 W US0236107 W US 0236107W WO 03040170 A9 WO03040170 A9 WO 03040170A9
Authority
WO
WIPO (PCT)
Prior art keywords
amino acid
acid sequence
antibody
light chain
heavy chain
Prior art date
Application number
PCT/US2002/036107
Other languages
French (fr)
Other versions
WO2003040170A3 (en
WO2003040170A2 (en
Inventor
Vahe Bedian
Ronald P Gladue
Jose Corvalan
Xiao-Chi Jia
Xiao Feng
Original Assignee
Pfizer Prod Inc
Abgenix Inc
Vahe Bedian
Ronald P Gladue
Jose Corvalan
Xiao-Chi Jia
Xiao Feng
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=23370387&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2003040170(A9) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to MEP-2008-814A priority Critical patent/ME00503B/en
Priority to ES02802898T priority patent/ES2432968T3/en
Priority to EA200400654A priority patent/EA011449B1/en
Priority to AU2002356926A priority patent/AU2002356926B2/en
Priority to JP2003542215A priority patent/JP4616555B2/en
Priority to CA2466128A priority patent/CA2466128C/en
Priority to BRPI0214137A priority patent/BRPI0214137B8/en
Priority to DK02802898.3T priority patent/DK1476185T3/en
Priority to SI200231036T priority patent/SI1476185T1/en
Priority to EP02802898.3A priority patent/EP1476185B1/en
Priority to HU0402247A priority patent/HU229829B1/en
Priority to CN028245709A priority patent/CN1582165B/en
Priority to NZ533180A priority patent/NZ533180A/en
Priority to APAP/P/2004/003034A priority patent/AP1918A/en
Priority to IL16182302A priority patent/IL161823A0/en
Priority to MXPA04004467A priority patent/MXPA04004467A/en
Priority to YU39204A priority patent/RS52484B/en
Application filed by Pfizer Prod Inc, Abgenix Inc, Vahe Bedian, Ronald P Gladue, Jose Corvalan, Xiao-Chi Jia, Xiao Feng filed Critical Pfizer Prod Inc
Publication of WO2003040170A2 publication Critical patent/WO2003040170A2/en
Publication of WO2003040170A3 publication Critical patent/WO2003040170A3/en
Priority to TNP2004000078A priority patent/TNSN04078A1/en
Priority to IS7253A priority patent/IS2940B/en
Priority to IL161823A priority patent/IL161823A/en
Priority to ZA2004/04207A priority patent/ZA200404207B/en
Priority to HRP20040525AA priority patent/HRP20040525B1/en
Priority to NO20042388A priority patent/NO334339B1/en
Publication of WO2003040170A9 publication Critical patent/WO2003040170A9/en
Priority to HK05102440.7A priority patent/HK1069122A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • 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
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/06Radiation therapy using light
    • A61N5/0613Apparatus adapted for a specific treatment
    • A61N5/062Photodynamic therapy, i.e. excitation of an agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/10X-ray therapy; Gamma-ray therapy; Particle-irradiation therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/117Nucleic acids having immunomodulatory properties, e.g. containing CpG-motifs
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/10Immunoglobulins specific features characterized by their source of isolation or production
    • C07K2317/14Specific host cells or culture conditions, e.g. components, pH or temperature
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • 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
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/75Agonist effect on antigen
    • 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 CD40 antigen is a 50 kDa cell surface glycoprotein which belongs to the Tumor Necrosis Factor Receptor (TNF-R) family.
  • TNF-R Tumor Necrosis Factor Receptor
  • CD40 is expressed in many normal and tumor cell types, including B lymphocytes, dendritic cells, monocytes, macrophages, thymic epithelium, endothelial cells, fibroblasts, and smooth muscle cells.
  • CD40 is expressed in all B-lymphomas and in 70% of all solid tumors.
  • CD40 is up-regulated in antigen presenting cells by maturation signals, such as LPS, IL-lj8, -FN- ⁇ and GM-CSF.
  • maturation signals such as LPS, IL-lj8, -FN- ⁇ and GM-CSF.
  • CD40 activation plays a critical role in regulating humoral and cellular immune responses. Antigen presentation without CD40 activation can lead to tolerance, while CD40 signaling can reverse such tolerance, enhance antigen presentation by all antigen presenting cells (APCs), lead to secretion of helper cytokines and chemokines, increase co-stimulatory molecule expression and signaling, and stimulate cytolytic activity of immune cells.
  • APCs antigen presenting cells
  • CD40 plays a critical role in B cell proliferation, maturation and class switching.
  • CD40 signaling pathway Disruption of the CD40 signaling pathway leads to abnormal serum immunoglobulin isotype distribution, lack of CD4+ T cell priming, and defects in secondary humoral responses.
  • the X-linked hyper-IgM syndrome is a disease associated with a mutation in the human CD40L gene, and it is characterized by the inability of affected individuals to produce antibodies other than those of the IgM isotype, indicating that the productive interaction between CD40 and CD40L is required for an effective immune response.
  • CD40 engagement by CD40L leads to the association of the CD40 cytoplasmic domain with TRAFs (TNF-R associated factors). (Lee H.H. et al., Proc. N ⁇ tl.
  • adhesion molecules e.g., ICAM
  • ICAM adhesion molecules
  • Lee H.H. et al. Proc. Natl. Acad. Sci. USA. 96:1421-6 (1999); Grousson L et al., Archives of Dermatol. Res. 290:325-30 (1998); Katada Y. et al., Europ. J. of Immunol. 26:192-200 (1996); Mayumi M. et al., J. of Allergy & Clin. Immunol. 96:1136-44 (1995); Flores-Romo L. et al., Immunol.
  • ICAM adhesion molecules
  • CD40 activation include: cell recruitment and differentiation by chemokines and cytokines; activation of monocytes; increased cytolytic activity of cytolytic T lymphocyte (CTL) and natural killer (NK) cells; induction of apoptosis in CD40 positive tumors; enhancement of immunogenicity of CD40 positive tumors; and tumor-specific antibody production.
  • CTL cytolytic T lymphocyte
  • NK natural killer
  • CD40 activation converts otherwise tolerogenic, antigen bearing B cells into competent APCs.
  • CD40 activation induces maturation and differentiation of cord blood progenitors into dendritic cells.
  • CD40 activation also induces differentiation of monocytes into functional dendritic cells. (Brossart P. et al., Blood 92:4238-47 (1998).) Further, CD40 activation enhances cytolytic activity of NK cells through APC-CD40 induced cytokines. (Carbone E. et al., J. of Exp. Med. 185:2053-60 (1997); Martin-Fontecha A. et al., J. of Immunol.
  • CD40 plays an essential role in the initiation and enhancement of immune responses by inducing maturation of APCs, secretion of helper cytokines, upregulation of costimulatory molecules, and enhancement of effector functions.
  • the critical role of CD40 signaling in the initiation and maturation of humoral and cytotoxic immune responses makes this system an ideal target for immune enhancement.
  • Such enhancement can be particularly important for mounting effective immune responses to tumor antigens, which are generally presented to the immune system through cross-priming of activated APCs.
  • CD40 is expressed in lymphomas, leukemias, multiple myeloma, a majority of carcinomas of nasopharynx, bladder, ovary, and liver, and some breast and colorectal cancers, activation of CD40 can have a broad range of clinical applications.
  • Anti-CD40 activating monoclonal antibodies can contribute to tumor eradication via several important mechanisms. Foremost among these is activation of host dendritic cells for enhanced tumor antigen processing and presentation, as well as enhanced antigen presentation or immunogenicity of CD40 positive tumor cells themselves, leading to activation of tumor specific CD4 + and CD8 + lymphocytes. Additional antitumor activity can be mediated by other immune- enhancing effects of CD40 signaling (production of chemokines and cytokines, recruitment and activation monocytes, and enhanced CTL and NK cytolytic activity), as well as direct killing of CD40 + tumors by induction of apoptosis or by stimulating a humoral response leading to ADCC. Apoptotic and dying tumor cells can also become an important source of tumor-specific antigens that are processed and presented by CD40 activated APCs.
  • Figures 1A-1H are sequence alignments of predicted amino acid sequences of isolated anti-CD40 monoclonal antibody light and heavy chain variable domains with the germline amino acid sequences of the corresponding light and heavy chain genes.). Differences between the clones and the germline sequence are indicated by shading. The germline CDR1, CDR2, and CDR3 sequences are underlined. In alignments of heavy chain sequences, apparent insertions to the CDR3 region are indicated by a dash (-) in the germline sequence and apparent deletions in the CDR3 region are indicated by a dash (-) in the clone sequence.
  • Figure IC the predicted kappa light chain variable region amino acid sequences from mAbs 10.8.3 and 21.4.1 and the germline amino acid sequence
  • Figure 2A-2H are sequence alignments of predicted amino acid sequences of isolated anti-CD40 monoclonal antibody light and heavy chain variable domains with the germline amino acid sequences of the corresponding light and heavy chain genes.). Differences between the clones and the germline sequence are indicated in bold. The germline CDR1, CDR2, and CDR3 sequences are underlined.
  • Figure 2B the predicted kappa light chain amino acid sequence from mAb 21.2.1 and the germline amino acid sequence
  • Figure 2D the predicted heavy chain amino acid sequence from mAb
  • Figure 3 is a dose-response curve that illustrates the ability of an anti- CD40 antibody of the invention (21.4.1) to enhance IL-12p40 production by human dendritic cells.
  • Figure 4 is a dose-response curve that illustrates the ability of an anti- CD40 antibody of the invention (21.4.1) to enhance IL-12p70 production by human dendritic cells.
  • Figure 5 is a graph that illustrates the ability of an anti-CD40 antibody of the invention (21.4.1) to increase immunogenicity of Jy stimulator cells and enhance CTL activity against Jy target cells.
  • Figure 6 is a tumor growth inhibition curve that illustrates the reduced growth of CD40 positive Daudi tumors in SCID-beige mice treated with an anti- CD40 antibody of the invention (21.4.1).
  • Figure 7 is a tumor growth inhibition curve that illustrates the reduced growth of CD40 negative K562 tumors in SCID-beige mice treated with an anti- CD40 antibody of the invention (21.4.1) and human dendritic cells and T cells.
  • Figure 8 shows inhibition in the growth of CD40 negative K562 tumors in SCID mice by different concentrations of anti-CD40 agonist mAb 23.29.1.
  • Figure 9 shows inhibition in the growth of CD40 negative K562 tumors in SCID mice by different concentrations of anti-CD40 agonist mAb 3.1.1.
  • Figure 10 shows inhibition in the growth of CD40 positive Raji tumors in the presence and absence of T cells and dendritic cells in SCID mice by an anti- CD40 agonist mAb.
  • Figure 11 shows inhibition in the growth of CD40 positive Raji tumors in SCID mice by anti-CD40 agonist antibodies.
  • Figure 12 shows inhibition in the growth of BT 474 breast cancer cells in SCID-beige mice by anti-CD40 agonist antibodies.
  • Figure 13 shows inhibition in the growth of PC-3 prostate tumors in SCID-beige mice by anti-CD40 agonist antibodies.
  • Figure 14 is a survival curve for SCID-beige mice injected (iv) with Daudi tumor cells and treated with anti-CD40 agonist antibodies.
  • Figure 15 is a Western blot analysis of anti-CD40 agonist antibodies to reduced (R) and non-reduced (NR) human CD40.
  • Figure 16 is an alignment of the D1-D4 domains of mouse and human CD40.
  • Figure 17 is an alignment of the mouse and human CD40 amino acid sequences showing the location of the fusion sites of the chimeras.
  • Figure 18 is a group of schematic diagrams of the chimeric CD40 constructs.
  • the present invention provides an isolated antibody or antigen-binding portion thereof that binds CD40 and acts as a CD40 agonist.
  • the invention provides a composition comprising the anti-CD40 antibody, or antigen binding portion thereof, and a pharmaceutically acceptable carrier.
  • the composition may further comprise another component, such as an anti-tumor agent or an imaging agent. Diagnostic and therapeutic methods are also provided by the invention.
  • the invention provides an isolated cell line, such as a hybridoma, that produces an anti-CD40 antibody or antigen binding portion thereof. [0049]
  • the invention also provides nucleic acid molecules encoding the heavy and/or light chain, or antigen-binding portions thereof, of an anti-CD40 antibody.
  • the invention provides vectors and host cells comprising the nucleic acid molecules, as well as methods of recombinantly producing the polypeptides encoded by nucleic acid molecules.
  • Non-human transgenic animals that express the heavy and/or light chain, or antigen-binding portions thereof, of an anti-CD40 antibody are also provided.
  • the invention also provides a method for treating a subject in need thereof with an effective amount of a nucleic acid molecule encoding the heavy and/or light chain, or antigen-binding portions thereof, of an anti-CD40 antibody.
  • polypeptide encompasses native or artificial proteins, protein fragments and polypeptide analogs of a protein sequence.
  • a polypeptide may be monomeric or polymeric.
  • isolated protein is a protein, polypeptide or antibody that by virtue of its origin or source of derivation (1) is not associated with naturally associated components that accompany it in its native state, (2) is free of other proteins from the same species, (3) is expressed by a cell from a different species, or (4) does not occur in nature.
  • a polypeptide that is chemically synthesized or synthesized in a cellular system different from the cell from which it naturally originates will be “isolated” from its naturally associated components.
  • a protein may also be rendered substantially free of naturally associated components by isolation, using protein purification techniques well known in the art.
  • Examples of isolated antibodies include an anti-CD40 antibody that has been affinity purified using CD40, an anti-CD40 antibody that has been synthesized by a hybridoma or other cell line in vitro, and a human anti-CD40 antibody derived from a transgenic mouse.
  • a protein or polypeptide is "substantially pure,” “substantially homogeneous,” or “substantially purified” when at least about 60 to 75% of a sample exhibits a single species of polypeptide.
  • the polypeptide or protein may be monomeric or multimeric.
  • a substantially pure polypeptide or protein will typically comprise about 50%, 60%, 70%, 80% or 90% W/W of a protein sample, more usually about 95%, and preferably will be over 99% pure.
  • Protein purity or homogeneity may be indicated by a number of means well known in the art, such as polyacrylamide gel electrophoresis of a protein sample, followed by visualizing a single polypeptide band upon staining the gel with a stain well known in the art. For certain purposes, higher resolution may be provided by using HPLC or other means well known in the art for purification.
  • polypeptide fragment refers to a polypeptide that has an amino-terminal and/or carboxy-terminal deletion, but where the remaining amino acid sequence is identical to the corresponding positions in the naturally-occurring sequence.
  • fragments are at least 5, 6, 8 or 10 amino acids long.
  • the fragments are at least 14 , at least 20, at least 50, or at least 70, 80, 90, 100, 150 or 200 amino acids long.
  • polypeptide analog refers to a polypeptide that comprises a segment that has substantial identity to a portion of an amino acid sequence and that has at least one of the following properties: (1) specific binding to CD40 under suitable binding conditions, (2) ability to activate CD40, (3) the ability to upregulate the expression of cell surface molecules such as ICAM, MHC- ⁇ , B7-1, B7-2, CD71, CD23 and CD83, or (4) the ability to enhance the secretion of cytokines such as IFN-/31, IL-2, IL-8, IL-12, IL-15, IL-18 and IL-23.
  • polypeptide analogs comprise a conservative amino acid substitution (or insertion or deletion) with respect to the naturally-occurring sequence.
  • Analogs typically are at least 20 or 25 amino acids long, preferably at least 50, 60, 70, 80, 90, 100, 150 or 200 amino acids long or longer, and can often be as long as a full-length naturally-occurring polypeptide.
  • Preferred amino acid substitutions are those which: (1) reduce susceptibility to proteolysis, (2) reduce susceptibility to oxidation, (3) alter binding affinity for forming protein complexes, and (4) confer or modify other physicochemical or functional properties of such analogs.
  • Analogs can include various muteins of a sequence other than the naturally-occurring peptide sequence. For example, single or multiple amino acid substitutions (preferably conservative amino acid substitutions) may be made in the naturally-occurring sequence (preferably in the portion of the polypeptide outside the domain(s) forming intermolecular contacts).
  • a conservative amino acid substitution should not substantially change the structural characteristics of the parent sequence (e.g., a replacement amino acid should not tend to break a helix that occurs in the parent sequence, or disrupt other types of secondary stracture that characterizes the parent sequence). Examples of art-recognized polypeptide secondary and tertiary structures are described in Proteins, Structures and Molecular Principles
  • ⁇ on-peptide analogs are commonly used in the pharmaceutical industry as drags with properties analogous to those of the template peptide. These types of non-peptide compound are termed "peptide mimetics” or “peptidomimetics.” Fauchere, J. Adv. Drug Res. 15:29 (1986); Neber and Freidinger, TINSp.392 (1985); and Evans et al, J. Med. Chem. 30:1229 (1987), which are incorporated herein by reference. Such compounds are often developed with the aid of computerized molecular modeling. Peptide mimetics that are structurally similar to therapeutically useful peptides may be used to produce an equivalent therapeutic or prophylactic effect.
  • a paradigm polypeptide i.e., a polypeptide that has a desired biochemical property or pharmacological activity
  • Systematic substitution of one or more amino acids of a consensus sequence with a D-amino acid of the same type may also be used to generate more stable peptides.
  • constrained peptides comprising a consensus sequence or a substantially identical consensus sequence variation may be generated by methods known in the art (Rizo and Gierasch, Ann. Rev. Biochem. 61:387 (1992), incorporated herein by reference); for example, by adding internal cysteine residues capable of forming intramolecular disulfide bridges which cyclize the peptide.
  • an "antibody” refers to a complete antibody or to an antigen-binding portion thereof, that competes with the intact antibody for specific binding. See generally, Fundamental Immunology. Ch. 7 (Paul, W., ed., 2nd ed. Raven Press, N.Y. (1989)) (incorporated by reference in its entirety for all purposes). Antigen- binding portions may be produced by recombinant DNA techniques or by enzymatic or chemical cleavage of intact antibodies.
  • Antigen-binding portions include, z.zter alia, Fab, Fab', F(ab') 2 , Fd, Fv, dAb, and complementarity determining region (CDR) fragments, single-chain antibodies (scFv), chimeric antibodies, diabodies and polypeptides that contain at least a portion of an antibody that is sufficient to confer specific antigen binding to the polypeptide.
  • CDR complementarity determining region
  • an antibody that is referred to by number is a monoclonal antibody that is obtained from the hybridoma of the same number.
  • monoclonal antibody 3.1.1 is obtained from hybridoma 3.1.1.
  • a Fd fragment means an antibody fragment that consists of the V H and C H 1 domains; an Fv fragment consists of the N and V H domains of a single arm of an antibody; and a dAb fragment (Ward et al., Nature 341:544-546 (1989)) consists of a V H domain.
  • the antibody is a single-chain antibody (scFv) in which a V L and V H domains are paired to form a monovalent molecules via a synthetic linker that enables them to be made as a single protein chain.
  • scFv single-chain antibody
  • the antibodies are diabodies, i.e., are bivalent antibodies in which V H and V L domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites.
  • diabodies i.e., are bivalent antibodies in which V H and V L domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites.
  • one or more CDRs from an antibody of the invention may be incorporated into a molecule either covalently or noncovalently to make it an immunoadhesin that specifically binds to CD40.
  • the CDR(s) may be incorporated as part of a larger polypeptide chain, may be covalently linked to another polypeptide chain, or may be incorporated noncovalently.
  • the binding sites may be identical to one another or may be different.
  • human antibody means any antibody in which all of the variable and constant domain sequences are human sequences. These antibodies may be prepared in a variety of ways, as described below.
  • chimeric antibody as used herein means an antibody that comprises regions from two or more different antibodies.
  • one or more of the CDRs are derived from a human anti-CD40 antibody.
  • all of the CDRs are derived from a human anti-CD40 antibody.
  • the CDRs from more than one human anti-CD40 antibodies are combined in a chimeric antibody.
  • a chimeric antibody may comprise a CDR1 from the light chain of a first human anti-CD40 antibody, a CDR2 from the light chain of a second human anti-CD40 antibody and a CDR3 and CDR3 from the light chain of a third human anti-CD40 antibody, and the CDRs from the heavy chain may be derived from one or more other anti-CD40 antibodies.
  • the framework regions may be derived from one of the same anti-CD40 antibodies or from one or more different human.
  • An "activating antibody” also referred to herein as an "agonist antibody” as used herein means an antibody that increases one or more CD40 activities by at least about 20% when added to a cell, tissue or organism expressing CD40.
  • the antibody activates CD40 activity by at least 40%, 50%, 60%, 70%), 80%), 85%.
  • the activating antibody is added in the presence of CD40L.
  • the activity of the activating antibody is measured using a whole blood surface molecule upregulation assay. See Example VII.
  • the activity of the activating antibody is measured using a dendritic cell assay to measure IL-12 release. See Example VIII.
  • the activity of the activating antibody is measured using an in vivo tumor model. See Example X. *
  • fragments or analogs of antibodies or immunoglobulin molecules can be readily prepared by those of ordinary skill in the art following the teachings of this specification.
  • Preferred amino- and carboxy-termini of fragments or analogs occur near boundaries of functional domains.
  • Structural and functional domains can be identified by comparison of the nucleotide and/or amino acid sequence data to public or proprietary sequence databases.
  • computerized comparison methods are used to identify sequence motifs or predicted protein conformation domains that occur in other proteins of known stracture and/or function. Methods to identify protein sequences that fold into a known three-dimensional stracture are known. See Bowie et al., Science 253:164 (1991).
  • the term "surface plasmon resonance”, as used herein, refers to an optical phenomenon that allows for the analysis of real-time biospecific interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BIAcore system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, N.J.).
  • BIAcore Phharmacia Biosensor AB, Uppsala, Sweden and Piscataway, N.J.
  • K D refers to the equilibrium dissociation constant of a particular antibody-antigen interaction.
  • epitopic determinants includes any protein determinant capable of specific binding to an immunoglobulin or T-cell receptor.
  • Epitopic determinants usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics.
  • An antibody is said to specifically bind an antigen when the equilibrium dissociation constant is -.1 ⁇ M, preferably --100 nM and most preferably --10 nM.
  • the twenty conventional amino acids and their abbreviations follow conventional usage. See Immunology - A Synthesis (2 nd
  • polynucleotide as referred to herein means a polymeric form of nucleotides of at least 10 bases in length, either ribonucleotides or deoxynucleotides or a modified form of either type of nucleotide.
  • the term includes single and double stranded forms.
  • isolated polynucleotide as used herein means a polynucleotide of genomic, cDNA, or synthetic origin or some combination thereof, which by virtue of its origin the "isolated polynucleotide” (1) is not associated with all or a portion of a polynucleotides with which the "isolated polynucleotide” is found in nature, (2) is operably linked to a polynucleotide to which it is not linked in nature, or (3) does not occur in nature as part of a larger sequence.
  • oligonucleotide includes naturally occurring, and modified nucleotides linked together by naturally occurring and non-naturally occurring oligonucleotide linkages.
  • Oligonucleotides are a polynucleotide subset generally comprising a length of 200 bases or fewer.
  • Preferably oligonucleotides are 10 to 60 bases in length and most preferably 12, 13, 14, 15, 16, 17, 18, 19, or 20 to 40 bases in length.
  • Oligonucleotides are usually single stranded, e.g. for primers and probes; although oligonucleotides may be double stranded, e.g. for use in the construction of a gene mutant.
  • Oligonucleotides of the invention can be either sense or antisense oligonucleotides.
  • nucleotides as used herein includes deoxyribonucleotides and ribonucleotides.
  • modified nucleotides as used herein includes nucleotides with modified or substituted sugar groups and the like.
  • oligonucleotide linkages referred to herein includes oligonucleotides linkages such as phosphorothioate, phosphorodithioate, phosphoroselenoate, phosphorodiselenoate, phosphoroanilothioate, phoshoraniladate, phosphoroamidate, and the like. See e.g., LaPlanche et al., Nucl. Acids Res.
  • oligonucleotide can include a label for detection, if desired.
  • operably linked sequences include both expression control sequences that are contiguous with the gene of interest and expression control sequences that act in trans or at a distance to control the gene of interest.
  • expression control sequence means polynucleotide sequences that are necessary to effect the expression and processing of coding sequences to which they are ligated.
  • Expression control sequences include appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation signals; sequences that stabilize cytoplasmic mRNA; sequences that enhance translation efficiency (i.e., Kozak consensus sequence); sequences that enhance protein stability; and when desired, sequences that enhance protein secretion.
  • control sequences differs depending upon the host organism; in prokaryotes, such control sequences generally include promoter, ribosomal binding site, and transcription termination sequence; in eukaryotes, generally, such control sequences include promoters and transcription termination sequence.
  • control sequences is intended to include, at a minimum, all components whose presence is essential for expression and processing, and can also include additional components whose presence is advantageous, for example, leader sequences and fusion partner sequences.
  • the term "vector”, as used herein, means a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • the vector is a plasmid, i.e., a circular double stranded DNA loop into which additional DNA segments may be ligated.
  • the vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome.
  • the vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors).
  • the vectors e.g., non-episomal mammalian vectors
  • the vectors can be integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome.
  • certain vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as "recombinant expression vectors" (or simply, “expression vectors").
  • recombinant host cell means a cell into which a recombinant expression vector has been introduced. It should be understood that "recombinant host cell” and “host cell” mean not only the particular subject cell but also the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term “host cell” as used herein. [0085] The term “selectively hybridize” referred to herein means to detectably and specifically bind.
  • Polynucleotides, oligonucleotides and fragments thereof in accordance with the invention selectively hybridize to nucleic acid strands under hybridization and wash conditions that minimize appreciable amounts of detectable binding to nonspecific nucleic acids.
  • “High stringency” or “highly stringent” conditions can be used to achieve selective hybridization conditions as known in the art and discussed herein.
  • high stringency or “highly stringent” conditions is the incubation of a polynucleotide with another polynucleotide, wherein one polynucleotide may be affixed to a solid surface such as a membrane, in a hybridization buffer of 6X SSPE or SSC, 50% formamide, 5X Denhardt's reagent, 0.5% SDS, 100 j-ig/ml denatured, fragmented salmon sperm DNA at a hybridization temperature of 42°C for 12-16 hours, followed by twice washing at 55°C using a wash buffer of IX SSC, 0.5% SDS. See also Sambrook et al., supra, pp. 9.50-9.55.
  • sequence identity in the context of nucleic acid sequences means the residues in two sequences that are the same when aligned for maximum correspondence.
  • the length of sequence identity comparison may be over a stretch of at least about nine nucleotides, usually at least about 18 nucleotides, more usually at least about 24 nucleotides, typically at least about 28 nucleotides, more typically at least about 32 nucleotides, and preferably at least about 36, 48 or more nucleotides.
  • polynucleotide sequences can be compared using FASTA, ' Gap or Bestfit, which are programs in Wisconsin Package Version 10.0, Genetics Computer Group (GCG), Madison, Wisconsin.
  • FASTA which includes, e.g., the programs
  • FASTA2 and FASTA3 provides alignments and percent sequence identity of the regions of the best overlap between the query and search sequences (Pearson, Methods Enzymol. 183:63-98 (1990); Pearson, Methods Mol. Biol. 132:185-219 (2000); Pearson, Methods Enzymol. 266:227-258 (1996); Pearson, J. Mol. Biol. 276:71-84 (1998); herein incorporated by reference). Unless otherwise specified, default parameters for a particular program or algorithm are used.
  • percent sequence identity between nucleic acid sequences can be deteraiined using FASTA with its default parameters (a word size of 6 and the NOP AM factor for the scoring matrix) or using Gap with its default parameters as provided in GCG Version 6.1, herein incorporated by reference.
  • FASTA FASTA with its default parameters
  • Gap Gap with its default parameters as provided in GCG Version 6.1, herein incorporated by reference.
  • a reference to a nucleotide sequence encompasses its complement unless otherwise specified.
  • a reference to a nucleic acid having a particular sequence should be understood to encompass its complementary strand, with its complementary sequence.
  • nucleic acid or fragment thereof when referring to a nucleic acid or fragment thereof, means that when optimally aligned with appropriate nucleotide insertions or deletions with another nucleic acid (or its complementary strand), there is nucleotide sequence identity in at least about 85%, preferably at least about 90%, and more preferably at least about 95%, 96%, 97%, 98% or 99% of the nucleotide bases, as measured by any well-known algorithm of sequence identity, such as FASTA, BLAST or Gap, as discussed above.
  • the term "substantial identity” means that two peptide sequences, when optimally aligned, such as by the programs GAP or BESTFIT using default gap weights, share at least 70, 75 or 80 percent sequence identity, preferably at least 90 or 95 percent sequence identity, and more preferably at least 97, 98 or 99 percent sequence identity.
  • residue positions that are not identical differ by conservative amino acid substitutions.
  • a “conservative amino acid substitution” is one in which an amino acid residue is substituted by another amino acid residue having a side chain R group) with similar chemical properties (e.g., charge or hydrophobicity). In general, a conservative amino acid substitution will not substantially change the functional properties of a protein.
  • the percent sequence identity or degree of similarity may be adjusted upwards to correct for the conservative nature of the substitution. Means for making this adjustment are well-known to those of skill in the art. See, e.g., Pearson, Methods Mol. Biol. 243:307-31 (1994).
  • Examples of groups of amino acids that have side chains with similar chemical properties include 1) aliphatic side chains: glycine, alanine, valine, leucine, and isoleucine; 2) aliphatic-hydroxyl side chains: serine and threonine; 3) amide-containing side chains: asparagine and glutamine; 4) aromatic side chains: phenylalanine, tyrosine, and tryptophan; 5) basic side chains: lysine, arginine, and histidine; 6) acidic side chains: aspartic acid and glutamic acid; and 7) sulfur-containing side chains: cysteine and methionine.
  • Preferred conservative amino acids substitution groups are: valine-leucine-isoleucine, phenylalanine-tyrosine, lysine-arginine, alanine-valine, glutamate-aspartate, and asparagine-glutamine.
  • a conservative replacement is any change having a positive value in the PAM250 log-likelihood matrix disclosed in Gonnet et al., Science 256:1443-45 (1992), herein incorporated by reference.
  • a “moderately conservative" replacement is any change having a nonnegative value in the
  • Sequence similarity for polypeptides is typically measured using sequence analysis software. Protein analysis software matches similar sequences using measures of similarity assigned to various substitutions, deletions and other modifications, including conservative amino acid substitutions.
  • GCG contains programs such as "Gap” and "Bestfit” which can be used with default parameters to determine sequence homology or sequence identity between closely related polypeptides, such as homologous polypeptides from different species of organisms or between a wild type protein and a mutein thereof. See, e.g., GCG Version 6.1. Polypeptide sequences also can be compared using FASTA using default or recommended parameters, a program in GCG Version 6.1.
  • FASTA e.g., FASTA2 and FASTA3
  • FASTA2 and FASTA3 provides alignments and percent sequence identity of the regions of the best overlap between the query and search sequences (Pearson, Methods Enzymol. 183:63-98 (1990); Pearson, Methods Mol. Biol. 132:185-219 (2000)).
  • Another preferred algorithm when comparing a sequence of the invention to a database containing a large number of sequences from different organisms is the computer program BLAST, especially blastp or tblastn, using default parameters. See, e.g., Altschul et al., J. Mol. Biol. 215:403-410 (1990); Altschul et al., Nucleic Acids Res. 25:3389-402 (1997); herein incorporated by reference.
  • the length of polypeptide sequences compared for homology will generally be at least about 16 amino acid residues, usually at least about 20 residues, more usually at least about 24 residues, typically at least about 28 residues, and preferably more than about 35 residues. When searching a database containing sequences from a large number of different organisms, it is preferable to compare amino acid sequences.
  • label or “labeled” refers to incorporation of another molecule in the antibody.
  • the label is a detectable marker, e.g., incorporation of a radiolabeled amino acid or attachment to a polypeptide of biotinyl moieties that can be detected by marked avidin (e.g., streptavidin containing a fluorescent marker or enzymatic activity that can be detected by optical or colorimetric methods).
  • the label or marker can be therapeutic, e.g., a drag conjugate or toxin.
  • Various methods of labeling polypeptides and glycoproteins are known in the art and may be used.
  • labels for polypeptides include, but are not limited to, the following: radioisotopes or radionuclides (e.g., 3 H, 14 C, 15 N, 35 S, 90 Y, 99 Tc, n l In, 125 I, I31 I), fluorescent labels (e.g., FITC, rhodamine, lanthanide phosphors), enzymatic labels
  • chemiluminescent markers e.g., horseradish peroxidase, /5-galactosidase, luciferase, alkaline phosphatase
  • chemiluminescent markers biotinyl groups, predetermined polypeptide epitopes recognized by a secondary reporter (e.g., leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags), magnetic agents, such as gadolinium chelates, toxins such as pertussis toxin, taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorabicin, daunorabicin, dihydroxy anthracin dione, mitoxantrone, mithrarnycin, actinomycin D, 1-dehydrotestosterone, glucocortic
  • Human antibodies avoid certain of the problems associated with antibodies that possess non-human (e.g., rodent) variable and/or constant regions. Such problems include the rapid clearance of the antibodies or immune response against the antibody. Therefore, in one embodiment, the invention provides humanized anti-CD40 antibodies. In another embodiment, the invention provides human anti-CD40 antibodies. In some embodiments, human anti-CD40 antibodies are produced by immunizing a rodent whose genome comprises human immunoglobulin genes so that the rodent produces human antibodies. Human anti- CD40 antibodies are expected to minimize the immunogenic and allergic responses intrinsic to non-human or non-human-derivatized monoclonal antibodies (Mabs) and thus to increase the efficacy and safety of the administered antibodies. The use of fully human antibodies can be expected to provide a substantial advantage in the treatment of chronic and recurring human diseases, such as inflammation and cancer, which may require repeated antibody administrations.
  • Mabs monoclonal antibodies
  • the invention provides eleven activating human anti-CD40 monoclonal antibodies (mAbs) and the hybridoma cell lines that produce them.
  • mAbs human anti-CD40 monoclonal antibodies
  • able A lists the sequence identifiers (SEQ ID NOS:) of the nucleic acids encoding the full- length heavy and light chains (including leader sequence), the corresponding full- length deduced amino acid sequences, and the nucleotide and deduced amino acid sequence of the heavy and light chain variable regions. Table A
  • the invention further provides human anti-CD40 mAb 23.25.1 and the hybridoma cell line that produces it.
  • the invention further provides heavy and/or light chain variants of certain of the above-listed human anti-CD40 mAbs, comprising one or more amino acid substitutions.
  • the invention provides two variant heavy chains of mAb 3.1.1. In one, the alanine at residue 78 is changed to threonine. In the second, the alanine at residue 78 is changed to threonine, and the valines at residues 88 and 97 are changed to alanines.
  • the invention also provides a variant light chain of mAb
  • the leucine at residue 4 and the leucine at residue 83 are changed to methionine and valine, respectively.
  • Combination with a variant heavy or light chain with a wild type light or heavy chain, respectively is designated by the mutant chain.
  • an antibody containing a wild type light chain and a heavy chain comprising the alanine to threonine mutation at residue 78 is designated as 3.1.1H-A78T.
  • antibodies containing any combination of a variant heavy chain and the variant light chain of 3.1.1 are included.
  • the invention provides a variant of the heavy chain of mAb 22.1.1 in which the cysteine at residue 109 is changed to an alanine.
  • a monoclonal antibody comprising the variant heavy chain and the 22.1.1 light chain chain is designated mAb 22.1.1 H-C 109 A.
  • the invention further provides two variant heavy chains and a variant light chain of mAb 23.28.1.
  • one heavy chain variant the aspartic acid at residue 16 is changed to glutamic acid.
  • a mAb comprising the variant heavy chain variant and the 23.28.1 light chain is designated 23.28.1 H- D16E.
  • the invention also includes a 23.28.1 light chain variant in which the cysteine at residue 92 is changed to an alanine.
  • a mAb comprising the 23.28.1 heavy chain and the variant light chain is designated 23.28.1 L C92A.
  • the invention also provides mAbs comprising either of the 23.28.1 heavy chain variants with the 23.28.1 light chain variant.
  • the light chain produced by hybridoma 23.29.1 contains a mutation in the constant region at residue 174.
  • the light chain produced by the hybridoma has arginine at this position instead of the canonical lysine.
  • the invention also provides a 23.29.1 light chain with the canonical lysine at residue 174 and a mAb, designated 23.29.1L-R174K, comprising the 23.29.1 heavy chain and the variant light chain.
  • the anti-CD40 antibody is 3.1.1, 3.1.1H- A78T, 3.1.1H-A78T-V88A-V97A, 3.1.1L-L4M-L83V, 7.1.2, 10.8.3, 15.1.1,
  • the anti-CD40 antibody comprises a light chain comprising an amino acid sequence selected from SEQ ID NO: 8, 16, 24, 32, 40, 48, 56, 64, 72, 80, 88, 94, 100 or 102 or the variable region therefrom, or encoded by a nucleic acid sequence selected from SEQ ID NO: 7, 15, 23, 31, 39, 47, 55, 63, 71, 79, 87, 93, 99 or 101.
  • the anti-CD40 antibody comprises a light chain comprising at least the CDR2 from one of listed antibodies, one of the above-identified amino acid sequences (as shown in Figs. 1A-1C and 2A-2C) or encoded by one of the above- identified nucleic acid sequences.
  • the light chain further comprises a CDR1 and CDR3 independently selected from a light chain variable region that comprises no more than ten amino acids from the amino acid sequence encoded by a germline VK A3/A19, L5 or A27 gene, or comprises a CDRl and CDR3 independently selected from one of a CDRl and CDR3 of (1) an antibody selected from 3.1.1, 3.1.1L-L4M-L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K or 24.2.1 ; (2) the amino acid sequence of SEQ ED NO: 4, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 94, 100 or 102 or (3) encoded by the nucleic acid sequence of SEQ ID NO: 3, 11, 19, 27, 35, 43, 51, 59, 67, 75, 83, 93, 99 or
  • the anti-CD40 antibody comprises a heavy chain comprising an amino acid sequence selected from SEQ ID NOS: 6, 14, 22, 30, 38, 46, 54, 62, 70, 78 or 86 or the variable region therefrom or encoded by a nucleic acid sequence selected from SEQ ID NOS: 5, 13, 21, 29, 37, 45, 53, 61, 69, 77 or 85.
  • the anti-CD40 antibody comprises a heavy chain comprising at least the CDR3 from one of listed antibodies, one of the above-identified amino acid sequences (as shown in Figs. 1A-1C and 2A-2C) or encoded by one of the above-identified nucleic acid sequences.
  • the heavy chain further comprises a CDRl and CDR2 independently selected from a heavy chain variable region that comprises no more than eighteen amino acids from the amino acid sequence encoded by a germline V H 3-30+, 4-59, 1-02, 4.35 or 3-30.3 gene, or comprises a CDRl and CDR2 independently selected from one of a CDRl and CDR2 of ( 1 ) an antibody selected from 3.1.1, 3.1.1 H-
  • the anti-CD40 antibody comprises a heavy chain and a light chain as defined above.
  • antibody 3.1.1H-A78T is identical to that of 3.1.1 except that residue 78 of the heavy chain is threonine instead of alanine.
  • residue 78 of the heavy chain is threonine instead of alanine.
  • residue 78 is changed to A, and residues 88 and 97 are changed from valine to alanine in the heavy chain.
  • L4M-L83V is identical to that of 3.1.1 except that residue 4 is methionine instead of leucine and residue 83 is valine instead of leucine in the light chain.
  • Antibody 22.1.1H-C109A is identical to that of 22.1.1 except that residue 109 of the heavy chain is changed from a cysteine to an alanine.
  • Antibodies 23.28.1H-D16E and 23.28.1L-C92A are identical to that of 23.28.1 except that residue 16 of the heavy chain is changed from aspartate to glutamate, and residue 92 of the light chain is changed from cysteine to alanine, respectively.
  • Antibody 23.29.1L-R174K is identical to that of 23.29.1 except that residue 174 of the light chain is changed from arginine to lysine.
  • the class and subclass of anti-CD40 antibodies may be determined by any method known in the art.
  • the class and subclass of an antibody may be determined using antibodies that are specific for a particular class and subclass of antibody. Such antibodies are available commercially.
  • the class and subclass can be determined by ELISA, or Western Blot as well as other techniques.
  • the class and subclass may be determined by sequencing all or a portion of the constant domains of the heavy and/or light chains of the antibodies, comparing their amino acid sequences to the known amino acid sequences of various class and subclasses of immunoglobulins, and determining the class and subclass of the antibodies.
  • the anti-CD40 antibody is a monoclonal antibody.
  • the anti-CD40 antibody can be an IgG, an IgM, an IgE, an IgA or an IgD molecule.
  • the anti-CD40 antibody is an IgG and is an IgGl, IgG2, IgG3 or IgG4 subclass.
  • the anti- CD40 antibodies are subclass IgG2.
  • the anti-CD40 antibodies demonstrate both species and molecule selectivity.
  • the anti-CD40 antibody binds to primate and human CD40.
  • the anti-CD40 antibody binds to human, cynomolgus or rhesus CD40.
  • the anti-CD40 antibody does not bind to mouse, rat, dog or rabbit CD40.
  • the species selectivity for the anti-CD40 antibody using methods well known in the art. For instance, one can determine species selectivity using Western blot, FACS, ELISA or RIA. (See, e.g., Example IV.)
  • the anti-CD40 antibody has a selectivity for CD40 that is more than 100 times greater than its selectivity for RANK (receptor activator of nuclear factor-kappa B), 4- IBB (CD 137), TNFR- 1 (Tumor Necrosis Factor Receptor- 1) and TNFR-2 (Tumor Necrosis Factor Receptor-2).
  • the anti-CD40 antibody does not exhibit any appreciable specific binding to any other protein other than CD40.
  • the invention provides a human anti-CD40 monoclonal antibody that binds CD40 and cross-competes with and/or binds the same epitope and or binds to CD40 with the same K D as a human anti-CD40 antibody selected from an antibody 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-V88A-V97A, 3.1.1L-L4M-L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K or 24.2.1; or a human anti-CD40 antibody that comprises a heavy chain variable region having an amino acid sequence of SEQ ID NO: 2, 10, 18, 26, 34, 42, 50, 58, 66, 74, 82, 90, 92, 96 or 98 or a human anti-CD40 antibody selected
  • test antibody if the test antibody is not able to bind to the CD40 at the same time, then the test antibody binds to the same epitope, an overlapping epitope, or an epitope that is in close proximity to the epitope bound by the human anti-CD40 antibody.
  • This experiment can be performed using ELISA, RIA, FACS or surface plasmon resonance. (See, e.g., Example NI.) In a prefened embodiment, the experiment is performed using surface plasmon resonance. In a more preferred embodiment, BIAcore is used.
  • the anti-CD40 antibody binds to CD40 with high affinity. In some embodiments, the anti-CD40 antibody binds to CD40 with a K D of 2 x 10 " M or less. In another preferred embodiments, the antibody binds to CD40 with a K D of 2 x 10 "9 , 2 x 10 "10 , 4.0 x 10 "11 M or less. In an even more prefened embodiment, the antibody binds to CD40 with a K D of 2.5 x 10 "12 M or less.
  • the antibody binds to CD40 with substantially the same KD as an antibody selected from 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-N88A-N97A, 3.1.1L-L4M-L83N, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.28.1L-C92A, 23.29.1 , 23.29.1L-R174K or 24.2.1.
  • the antibody binds to CD40 with substantially the same KD as an antibody that comprises a CDR2 of a light chain, and/or a CDR3 of a heavy chain from an antibody selected from 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-N88A-N97A, 3.1.1L-L4M-L83N, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, . 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.28.1L-C92A, 23.29.1, 23.29.1L- Rl 74K and 24.2.1.
  • the antibody binds to CD40 with substantially the same KD as an antibody that comprises a heavy chain variable region having an amino acid sequence of SEQ ID NO: 2, 10, 18, 26, 34, 42, 50, 58, 66, 74, 82, 90, 92, 96 or 98 or that comprises a light chain having an amino acid sequence of SEQ ID NO: 4, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 94, 100 or 102.
  • the antibody binds to CD40 with substantially the same K D as an antibody that comprises a CDR2 of " a light chain variable region having an amino acid sequence of SEQ ID NO: 4, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 94, 100 or 102 or a CDR3 of a heavy chain variable region having an amino acid sequence of SEQ ID NO: 2, 10, 18, 26, 34, 42, 50, 58, 66, 74, 82, 90, 92, 96 or 98.
  • the anti-CD40 antibody has a low dissociation rate.
  • the anti-CD40 antibody has an K off of 2.0 x 10 "4 or lower.
  • the K off is 2.0 x 10 "7 or lower. In some embodiments, the K off is substantially the same as an antibody described herein, including an antibody selected from 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-N88A- N97A, 3.1.1L-L4M-L83N, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H- C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K and 24.2.1.
  • the antibody binds to CD40 with substantially the same K off as an antibody that comprises a CDR3 of a heavy chain or a CDR2 of a light chain from an antibody selected from 3.1.1, 3.1.1H- A78T, 3.1.1H-A78T-N88A-N97A, 3.1.1L-L4M-L83N, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K and 24.2.1.
  • the antibody binds to CD40 with substantially the same K off as an antibody that comprises a heavy chain variable region having an amino acid sequence of SEQ ID NO: 2, 10, 18, 26, 34, 42, 50, 58, 66, 74, 82, 90, 92, 96 or 98 or that comprises a light chain variable region having an amino acid sequence of SEQ ID NO: 4, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 94, 100 or 102.
  • the antibody binds to CD40 with substantially the same K off as an antibody that comprises a CDR2 of a light chain variable region having an amino acid sequence of SEQ ID NO: 4, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 94, 100 or 102 or a CDR3 of a heavy chain variable region having an amino acid sequence of SEQ ID NO: 6, 14, 22, 30, 38, 46, 54, 62, 70, 78, 86, 90, 92, 96 or 98.
  • the binding affinity and dissociation rate of an anti-CD40 antibody to CD40 can be determined by any method known in the art.
  • the binding affinity can be measured by competitive ELIS As, RIAs or surface plasmon resonance, such as BIAcore.
  • the dissociation rate also can be measured by surface plasmon resonance.
  • the binding affinity and dissociation rate is measured by surface plasmon resonance. More preferably, the binding affinity and dissociation rate are measured using a BIAcoreTM. See, e.g., Example XIV. Light and Heavy Chain Gene Usage
  • An anti-CD40 antibody of the invention can comprise a human kappa or a human lambda light chain or an amino acid sequence derived therefrom.
  • the light chain variable domain (VL) is encoded in part by a human A3/A19 (DPK- 15), L5 (DP5), or A27 (DPK-22) V/c gene.
  • the N L of the anti-CD40 antibody contains one or more amino acid substitutions relative to the germline amino acid sequence. In some embodiments, the N L of the anti-CD40 antibody comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid substitutions relative to the germline amino acid sequence. In some embodiments, one or more of those substitutions from germline is in the CDR regions of the light chain.
  • the amino acid substitutions relative to germline are at one or more of the same positions as the substitutions relative to germline in any one or more of the N L of antibodies 3.1.1, 3.1.1L-L4M-L83N, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K and 24.2.1.
  • the N L of the anti-CD40 antibody may contain one or more amino acid substitutions compared to germline found in antibody 21.4.1, and other amino acid substitutions compared to germline found in antibody 10.8.3 which utilizes the same N gene as antibody 21.4.1.
  • the amino acid changes are at one or more . of the same positions but involve a different mutation than in the reference antibody.
  • amino acid changes relative to germline occur at one or more of the same positions as in any of the N L of antibodies 3.1.1, 3.1.1 L- L4M-L83N, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K and 24.2.1, but the changes may represent conservative amino acid substitutions at such position(s) relative to the amino acid in the reference antibody. For example, if a particular position in one of these antibodies is changed relative to germline and is glutamate, one may conservatively substitute aspartate at that position. Similarly, if an amino acid substitution compared to germline is serine, one may conservatively substitute threonine for serine at that position. Conservative amino acid substitutions are discussed supra.
  • the light chain of the human anti-CD40 antibody comprises the amino acid sequence that is the same as the amino acid sequence of the V L of antibody 3.1.1 (SEQ. LD NO: 4), 3.1.1L-L4M-L83V (SEQ ID NO: 94), 7.1.2 (SEQ. ID NO: 12), 10.8.3 (SEQ. ID NO: 20), 15.1.1 (SEQ. ID NO: 28), 21.4.1(SEQ. ID NO:), 21.2.1 (SEQ. ID NO: 36), 21.4.1 (SEQ ID NO: 44), 22.1.1 (SEQ. ID NO: 52), 23.5.1 (SEQ. ID NO: 60), 23.28.1 (SEQ. ID NO: 68), 23.28.1L-C92A (SEQ.
  • the light chain of the anti-CD40 antibody comprises at least the light chain CDR2, and may also comprise the CDRl and CDR3 regions of a germline sequence, as described herein.
  • the light chain may comprise a CDRl and CDR2 of an antibody independently selected from 3.1.1, 3.1.1L-L4M-L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.29.1 and 24.2.1, or CDR regions each having less than 8, less than 6, less than 4 or less than 3 conservative amino acid substitutions and/or a total of three or fewer non- conservative amino acid substitutions.
  • the light chain of the anti-CD40 antibody comprises at least the light chain CDR2, and may also comprise the CDRl and CDR3 regions, each of which are independently selected from the CDRl and CDR3 regions of an antibody having a light chain variable region comprising the amino acid sequence selected from SEQ ID NOS: 4, 12, 20,
  • variable region of the heavy chain amino acid sequence is encoded in part by a human V H 3-30+, V H 4-59, V H 1-02, V H 4.35 or V H 3-30.3 gene.
  • the V H of the anti-CD40 antibody contains one or more amino acid substitutions, deletions or insertions (additions) relative to the germline amino acid sequence.
  • variable domain of the heavy chain comprises 1, 2, 3, 4, 5, 6, 1, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17 or 18 mutations from the germline amino acid sequence.
  • the mutation(s) are non-conservative substitutions compared to the germline amino acid sequence.
  • the mutations are in the CDR regions of the heavy chain.
  • the amino acid changes are made at one or more of the same positions as the mutations from germline in any one or more of the V H of antibodies 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-V88A-V97A, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.29.1 and 24.2.1.
  • the amino acid changes are at one or more of the same positions but involve a different mutation than in the reference antibody.
  • the heavy chain comprises an amino acid sequence of the variable domain (V H ) of antibody 3.1.1 (SEQ JD NO: 2), 3.1.1H-A78T (SEQ ID NO: 90), 3.1.1H-A78T-V88A-V97A (SEQ ID NO: 92), 7.1.2 (SEQ ID NO: 10), 10.8.3 (SEQ ID NO: 18), 15.1.1 (SEQ ID NO: 26), 21.2.1 (SEQ ID NO: 34), 21.4.1 (SEQ ID NO: 42), 22.1.1 (SEQ ID NO: 50), 22.1.1H-C109A (SEQ ID NO: 96), 23.5.1 (SEQ ID NO: 58), 23.28.1 (SEQ ID NO: 66), 23.28.1H-D16E (SEQ ID NO: 98), 23.29.1 (SEQ ID NO: 74) and 24.2.1 (SEQ ID NO: 82), or said amino acid sequence having up to 1, 2, 3, 4, 6, 8 or 10 conservative amino acid substitutions and/or a total
  • the heavy chain comprises the heavy chain CDRl, CDR2 and CDR3 regions of antibody 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-N88A- N97A, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.29.1 and 24.2.1 (as shown in Figs. lD-lH or 2D-2H), or said CDR regions each having less than 8, less than 6, less than 4, or less than 3 conservative amino acid substitutions and/or a total of three or fewer non-conservative amino acid substitutions.
  • the heavy chain comprises a CDR3, and may also comprise the CDRl and CDR2 regions of a germline sequence, as described above, or may comprise a CDRl and CDR2 of an antibody, each of which are independently selected from an antibody comprising a heavy chain of an antibody selected from 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-N88A-N97A, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H- D16E, 23.29.1 and 24.2.1.
  • the heavy chain comprises a CDR3, and may also comprise the CDRl and CDR2 regions, each of which are
  • J5 independently selected from a CDRl and CDR2 region of a heavy chain variable region comprising an amino acid sequence selected from SEQ ID ⁇ OS: 2, 10, 18, 26, 34, 42, 50, 58, 66, 74, 82, 90, 92, 96 or 98 (as shown in Figs. 1D-1H or Figs. 2D-2H)or encoded by a nucleic acid sequence selected from SEQ ID ⁇ OS: 1, 9, 17, 25, 33, 41, 49, 57, 65, 73, 81, 89, 91, 95 or 97.
  • the 0 antibody comprises a heavy chain as disclosed above and a light chain as disclosed above.
  • One type of amino acid substitution that may be made is to change one or more cysteines in the antibody, which may be chemically reactive, to another residue, such as, without limitation, alanine or serine.
  • the 5 cysteine substitution is made in a framework region of a variable domain or in the constant domain of an antibody.
  • the cysteine is in a non- canonical region of the antibody.
  • Another type of amino acid substitution that may be made is to change any potential proteolytic sites in the antibody, particularly those that are in a framework region of a variable domain, in the constant domain 0 of an antibody, or in a non-canonical region of the antibody Substitution of cysteine residues and removal of proteolytic sites may decrease the risk of any heterogeneity in the antibody product and thus increase its homogeneity.
  • Another type of amino acid substitution is to eliminate asparagine-glycine pairs, which form potential deamidation sites, by altering one or both of the residues. This is 5 preferably done in framework regions, the constant domain or non-canonical regions of the antibody.
  • an anti-CD40 antibody that is an activating antibody, i.e., a CD40 agonist.
  • An activating antibody 0 amplifies or substitutes for the effects of CD40L on CD40.
  • the activating antibody is essentially a mimic of CD40L, and competes with CD40L for binding to CD40.
  • the antibody does not compete with CD40L for binding to CD40, but amplifies the effect of CD40L binding to CD40.
  • the anti-CD40 antibody activates CD40 in the presence or absence of CD40L.
  • the invention provides an anti-CD40 antibody that inhibits the proliferation of tumor cells in vitro or tumor growth in vivo.
  • the antibody inhibits tumor growth by at least 50%, 55%, 60%, 65%, 70%, 75%. In some embodiments, the antibody inhibits tumor growth by 75%. In one embodiment, the inhibition of tumor growth is detectable 14 days after initial treatment with the antibody. In other embodiments, the inhibition of tumor growth is detectable 7 days after initial treatment with the antibody.
  • another antineoplastic agent is administered to the animal with the anti-CD40 antibody. In some embodiments, the antineoplastic agent further inhibits tumor growth. In some embodiments, the antineoplastic agent is adriamycin or taxol. In some embodiments, the co-admimstration of an antineoplastic agent and the anti-CD40 antibody inhibits tumor growth by at least 50%, after a period of 22-24 days from initiation of treatment compared to tumor growth on an untreated animal.
  • Another aspect of the invention provides an anti-CD40 antibody that induces cell death of CD40 positive cells.
  • the antibody causes apoptosis of CD40 positive cells either in vivo or in vitro.
  • the anti-CD40 antibody enhances the expression of B cell surface molecules, including but not limited to ICAM, MHC-II, B7-2, CD71, CD23 and CD83.
  • 1 ⁇ g/ml of the antibody enhances ICAM expression in a whole blood B-cell surface molecule up-regulation assay by at least 2 fold, or more preferably by at least 4 fold.
  • 1 ⁇ g/ml of the antibody enhances MHC-II expression in a whole blood B-cell surface molecule upregulation assay by at least 2 fold, or more preferably by at least 3 fold.
  • 1 ⁇ g ml of the antibody enhances CD23 expression in whole blood B-cell surface molecule up-regulation assay by at least 2 fold, or more preferably by at least 5 fold. See, e.g., Example VII, Table 25.
  • the anti-CD40 antibody enhances the expression of dendritic cell surface molecules including but not limited to MHC-II, ICAM, B7-2, CD83 and B7-1.
  • the range of upregulation is similar to the range of upregulation observed in B cells. See, e.g., Tables 25 and 26, infra.
  • the antibody preferentially upregulates the expression of dendritic cell surface molecules, such as B7-2 and MHC-II, compared to B cell expression of these molecules. See, e.g., Table 27.
  • the antibody enhances cellular secretion of cytokines including but not limited to IL-8, IL-12, IL-15, IL-18 and IL-23. [0132] In some embodiments the antibody enhances cytokine secretion by dendritic cells and adherent monocytes. In some embodiments cytokine production is further enhanced by co-stimulation with one or more of LPS, IFN- ⁇ or IL-ljS. In yet another aspect of the invention, the antibody with LPS co- stimulation enhances IL-12p70 production in a dendritic cell assay with an EC 50 of about 0.48 ⁇ g/ml. In some embodiments, the antibody enhances IL-12p40 production in dendritic cells with an EC 50 of about 0.21 ⁇ g/ml. (See, e.g., Example
  • the antibody enhances secretion of IFN-gamma by
  • the antibody enhances IFN-gamma secretion in an allogenic T cell dendritic cell assay with an EC 50 of about 0.3 ⁇ g/ml. In some embodiments, the antibody enhances IFN-gamma secretion in an allogenic T cel .dendritic cell assay with an EC50 of about 0.2 ⁇ g/ml. In one embodiment, the antibody enhances IFN-gamma secretion in an allogenic T cell/dendritic cell assay with an EC 50 of about 0.03 ⁇ g/ml.
  • human antibodies are produced by immunizing a non-human animal comprising in its genome some or all of human immunoglobulin heavy chain and light chain loci with a CD40 antigen.
  • the non-human animal is a XenoMouseTM animal.
  • XenoMouseTM mice are engineered mouse strains that comprise large fragments of human immunoglobulin heavy chain and light chain loci and are deficient in mouse antibody production. See, e.g., Green et al., Nature Genetics 7:13-21 (1994) and U.S. Patents 5,916,771, 5,939,598, 5,985,615, 5,998,209,
  • the invention provides a method for making anti-CD40 antibodies from non-human, non-mouse animals by immunizing non-human transgenic animals that comprise human immunoglobulin loci with a CD40 antigen.
  • the non-human animals are rats, sheep, pigs, goats, cattle or horses.
  • XenoMouseTM mice produce an adult-like human repertoire of fully human antibodies and generate antigen-specific human antibodies.
  • the XenoMouseTM mice contain approximately 80% of the human antibody V gene repertoire through introduction of megabase sized, germline configuration yeast artificial chromosome (YAC) fragments of the human heavy chain loci and kappa light chain loci.
  • YAC yeast artificial chromosome
  • the non-human animal comprising human immunoglobulin genes are animals that have a human immunoglobulin "minilocus".
  • minilocus an exogenous Ig locus is mimicked through the inclusion of individual genes from the Ig locus.
  • one or more N H genes, one or more D H genes, one or more J H genes, a mu constant domain, and a second constant domain (preferably a gamma constant domain) are formed into a construct for insertion into an animal.
  • This approach is described, inter alia, in U.S. Patent os. 5,545,807, 5,545,806, 5,569,825, 5,625,126, 5,633,425,
  • An advantage of the minilocus approach is the rapidity with which constructs including portions of the Ig locus can be generated and introduced into animals.
  • a potential disadvantage of the minilocus approach is that there may not be sufficient immunoglobulin diversity to support full B-cell development, such that there may be lower antibody production.
  • the invention provides a method for making humanized anti-CD40 antibodies.
  • non-human animals are immunized with a CD40 antigen as described below under conditions that permit antibody production.
  • Antibody-producing cells are isolated from the animals, fused with myelomas to produce hybridomas, and nucleic acids encoding the heavy and light chains of an anti-CD40 antibody of interest are isolated. These nucleic acids are subsequently engineered using techniques known to those of skill in the art and as described further below to reduce the amount of non-human sequence, i.e., to humanize the antibody to reduce the immune response in humans [0141]
  • the CD40 antigen is isolated and/or purified CD40.
  • the CD40 antigen is human CD40.
  • the CD40 antigen is a fragment of CD40.
  • the CD40 fragment is the extracellular domain of CD40.
  • the CD40 fragment comprises at least one epitope of CD40.
  • the CD40 antigen is a cell that expresses or overexpresses CD40 or an immunogenic fragment thereof on its surface.
  • the CD40 antigen is a CD40 fusion protein.
  • the CD40 antigen is administered with an adjuvant to stimulate the immune response.
  • adjuvants include complete or incomplete Freund's adjuvant, RIBI (muramyl dipeptides) or ISCOM (immunostimulating complexes).
  • RIBI muramyl dipeptides
  • ISCOM immunocomplementary metal-oxide-semiconductor
  • antibodies and/or antibody-producing cells can be obtained from the animal.
  • anti-CD40 antibody-containing serum is obtained from the animal by bleeding or sacrificing the animal.
  • the serum may be used as it is obtained from the animal, an immunoglobulin fraction may be obtained from the serum, or the anti-CD40 antibodies may be purified from the serum.
  • serum or immunoglobulins obtained in this manner will be polyclonal
  • the disadvantage is using polyclonal antibodies prepared from serum is that the amount of antibodies that can be obtained is limited and the polyclonal antibody has a heterogeneous anay of properties.
  • antibody-producing immortalized cell lines are prepared from cells isolated from the immunized animal. After immunization, the animal is sacrificed and lymph node and/or splenic B cells are immortalized.
  • Methods of immortalizing cells include, but are not limited to, transferring them with oncogenes, inflecting them with the oncogenic virus cultivating them under conditions that select for immortalized cells, subjecting them to carcinogenic or mutating compounds, fusing them with an immortalized cell, e.g., a myeloma cell, and inactivating a tumor suppressor gene. See, e.g., Harlow and Lane, supra.
  • the myeloma cells preferably do not secrete immunoglobulin polypeptides (a non-secretory cell line).
  • Immortalized cells are screened using CD40, a portion thereof, or a cell expressing CD40.
  • the initial screening is performed using an enzyme-linked immunoassay (ELISA) or a radioimmunoassay.
  • ELISA enzyme-linked immunoassay
  • radioimmunoassay An example of ELISA screening is provided in WO 00/37504, herein incorporated by reference.
  • Anti-CD40 antibody-producing cells e.g., hybridomas
  • Hybridomas can be expanded in vivo in syngeneic animals, in animals that lack an immune system, e.g., nude mice, or in cell culture in vitro.
  • the immunized animal is a non-human animal that expresses human immunoglobulin genes and the splenic B cells are fused to a myeloma cell line from the same species as the non-human animal.
  • the immunized animal is a XENOMOUSETM animal and the myeloma cell line is a non-secretory mouse myeloma.
  • the myeloma cell line is P3-X63-AG8.653. See, e.g., Example I.
  • the invention provides hybridomas that produce an human anti-CD40 antibody.
  • the hybridomas are mouse hybridomas, as described above.
  • the hybridomas are produced in a non-human, non-mouse species such as rats, sheep, pigs, goats, cattle or horses.
  • the hybridomas are human hybridomas.
  • the present invention also encompasses nucleic acid molecules encoding anti-CD40 antibodies.
  • different nucleic acid molecules encode a heavy chain and a light chain of an anti-CD40 immunoglobulin.
  • the same nucleic acid molecule encodes a heavy chain and a light chain of an anti-CD40 immunoglobulin.
  • the nucleic acid molecule encoding the variable domain of the light chain comprises a human A3/A19 (DPK-15), L5 (DP5) or A27 (DPK-22) V/c gene sequence or a sequence derived therefrom.
  • the nucleic acid molecule comprises a nucleotide sequence of a A3/A19 V/c gene and a J/cl, Jc2 or Jc3 gene or sequences derived therefrom. In some embodiments, the nucleic acid molecule comprises a nucleotide sequence of an L5 V c gene and a J/c4 gene. In some embodiments, the nucleic acid molecule comprises a nucleotide sequence of a A27 V/c gene and a J ⁇ 3 gene. [0151] In some embodiments, the nucleic acid molecule encoding the light chain, encodes an amino acid sequence comprising 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 mutations from the germline amino acid sequence.
  • the nucleic acid molecule comprises a nucleotide sequence that encodes a VL amino acid sequence comprising 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 non-conservative amino acid substitutions and/or 1, 2 or 3 non-conservative substitutions compared to the germline sequence. Substitutions may be in the CDR regions, the framework regions or in the constant domain.
  • the nucleic acid molecule encoding the variable domain of the light chain (V L ) encodes a V L amino acid sequence comprising one or more mutations compared to the germline sequence that are identical to the mutations found in the V L of one of the antibodies 3.1.1, 3.1.1L-L4M-L83V, 7.1.2,
  • the nucleic acid molecule encodes at least three amino acid mutations compared to the germline sequence found in the V of one of the antibodies 3.1.1, 3.1.1L-L4M-L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.29.1 and 24.2.1.
  • the nucleic acid molecule comprises a nucleotide sequence that encodes the V L amino acid sequence of monoclonal antibody 3.1.1 (SEQ ID NO: 4), 3.1.1L-L4M-L83V (SEQ ID NO: 94), 7.1.2 (SEQ'ID NO: 12), 10.8.3 (SEQ ID NO: 20), 15.1.1 (SEQ ID NO: 28), 21.2.1 (SEQ ID NO: 36), 2.1.4.1 (SEQ ID NO: 44), 22.1.1 (SEQ ID NO: 52), 23.5.1 (SEQ ID NO: 60),
  • said portion comprises at least the CDR3 region.
  • the nucleic acid encodes the amino acid sequence of the light chain CDRs of said antibody.
  • said portion is a contiguous portion comprising CDR1-CDR3.
  • the nucleic acid molecule comprises a nucleotide sequence that encodes the amino acid sequence of one of SEQ ID NOS: 4, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 94 or 100, or said sequence lacking the signal sequence.
  • the nucleic acid molecule comprises the nucleotide sequence of SEQ ID NOS: 3, 11, 19, 27, 35, 43, 51, 59, 67, 75, 83, 93 or 99, or a portion thereof, said sequences optionally lacking the signal sequence.
  • said portion encodes a V L region. In some embodiments, said portion encodes at least the CDR2 region. In some embodiments, the nucleic acid encodes the amino acid sequence of the light chain CDRs of said antibody. In some embodiments, said portion encodes a contiguous region from CDR1-CDR3.
  • the nucleic acid molecule encodes a V L amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98% or 99% identical to a V L amino acid sequence of any one of antibodies 3.1.1, 3.1.1L-L4M- L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1,
  • Nucleic acid molecules of the invention include nucleic acids that hybridize under highly stringent conditions, such as those described above, to a nucleic acid sequence encoding the amino acid sequence of SEQ ID NOS: 4, 12, 20, 28, 36, 44, 52, 60,
  • the nucleic acid encodes a full-length light chain of an antibody selected from 3.1.1, 3.1.1L-L4M-L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.29.1, 23.29.1L-
  • nucleic acid may comprise the nucleotide sequence of SEQ ID NOS: 7, 15, 23, 31, 39, 47, 55, 63, 71, 79 or 87, or a nucleic acid molecule encoding a light chain comprise a mutation, such as one disclosed herein.
  • the nucleic acid molecule encodes the variable domain of the heavy chain (V H ) that comprises a human 3-30+, 4-59, 1- 02, 4.35 or 3-30.3 V H gene sequence or a sequence derived therefrom.
  • V H variable domain of the heavy chain
  • the nucleic acid molecule comprises a human 3-30+ V H gene, a D4 (DIR3) gene and a human J H 6 gene; a human 3-30+ VH gene, a human Dl-26 (DIR5) gene and a human J H 6 gene; a human 4.35 V H gene, a human DIR3 gene and a human H 6 gene; a human 4-59 V H gene, a human D4-23 gene and a human J H 4 gene; a human 1-02 V H gene, a human DLR1 gene and a human J H 4 gene; a human 3-30+ V H gene, a human D6-19 (DIR3) gene and a human J H 4 gene; a human 3-30+ VH gene, a human Dl-1 gene and a human J H 6 gene; a human 3-30+ V H gene, a human D4-17 gene and a human J H 6 gene; a human 3-30.3 VH gene, a human D4-17 gene and a human JH6 gene;
  • the nucleic acid molecule encodes an amino acid sequence comprising 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17 or 18 mutations compared to the germline amino acid sequence of the human V, D or J genes.
  • said mutations are in the V H region.
  • said mutations are in the CDR regions.
  • the nucleic acid molecule encodes one or more amino acid mutations compared to the germline sequence that are identical to amino acid mutations found in the V H of monoclonal antibody 3.1.1, 3.1.1 H-A78T,
  • the nucleic acid encodes at least three amino acid mutations compared to the germline sequences that are identical to at least three amino acid mutations found in one of the above-listed monoclonal antibodies.
  • the nucleic acid molecule comprises a nucleotide sequence that encodes at least a portion of the V H amino acid sequence of antibody 3.1.1 (SEQ ID NO: 2), 3.1.1H-A78T (SEQ ID NO: 90), 3.1.1H-A78T-V88A- V97A (SEQ ID NO: 92), 7.1.2 (SEQ ID NO: 10), 10.8.3 (SEQ ID NO: 18), 15.1.1 (SEQ ID NO: 26), 21.2.1 (SEQ ID NO: 34), 21.4.1 (SEQ ID NO: 42), 22.1.1 (SEQ ID NO: 50), 22.1.1H-C109A (SEQ ID NO: 96), 23.5.1 (SEQ ID NO: 58), 23.28.1 (SEQ ID NO: 66), 23.28.1H-D16E (SEQ ID NO: 98), 23.29.1 (SEQ ID NO: 74) or 24.2.1 (SEQ ID NO: 82), or said sequence having conservative amino acid mutations and/or a
  • the sequence encodes one or more CDR regions, preferably a CDR3 region, all three CDR regions, a contiguous portion including CDR1-CDR3, or the entire V H region, with or without a signal sequence.
  • the nucleic acid molecule comprises a nucleotide sequence that encodes the amino acid sequence of one of SEQ ID NOS: 2, 10, 18, 26, 34, 42, 50, 58, 66, 74, 82, 90, 92, 96 or 98, or said sequence lacking the signal sequence.
  • the nucleic acid molecule comprises at least aportion of the nucleotide sequence of SEQ ID NO: 1, 9, 17, 25, 33, 41, 49, 57, 65, 73, 81, 89, 91, 95 or 97, or said sequence lacking the signal sequence.
  • said portion encodes the V H region (with or without a signal sequence), a CDR3 region, all three CDR regions, or a contiguous region including CDR1-CDR3.
  • the nucleic acid molecule encodes a V H amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98% or 99% identical to the V H amino acid sequences shown in FIGS.
  • Nucleic acid molecules of the invention include nucleic acids that hybridize under highly stringent conditions, such as those described above, to a nucleic acid sequence encoding the amino acid sequence of SEQ ID NOS: 2, 10, 18, 26, 34, 42, 50, 58, 66, 74, 82, 90, 92, 96 or 98, or that has the nucleic acid sequence of SEQ ID NOS: 1, 9, 17, 25, 33, 41, 49, 57, 65, 73, 81, 89, 91, 95 or 97.
  • Nucleic acid molecule of the invention include nucleic acid molecule that hybridize under highly stringent conditions, such as those described above, to a nucleic acid sequence encoding a V H described immediately above.
  • the nucleic acid encodes a full-length heavy chain of an antibody selected from 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-V88A- V97A, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.29.1 and 24.2.1, or a heavy chain having the amino acid sequence of SEQ ID NOS: 6, 14, 22, 30, 38, 46, 54, 62, 70, 78 or 86, or a heavy chain comprising a mutation, such as one of the mutations discussed herein Further, the nucleic acid may comprise the nucleotide sequence of SEQ ID NOS: 5, 13, 21, 29, 37, 45
  • a nucleic acid molecule encoding the heavy or entire light chain of an anti-CD40 antibody or portions thereof can be isolated from any source that produces such antibody.
  • the nucleic acid molecules are isolated from a B cell isolated from an animal immunized with CD40 or from an immortalized cell derived from such a B cell that expresses an anti-CD40 antibody.
  • Methods of isolating mRNA encoding an antibody are well-known in the art. See, e.g., Sambrook et al. The mRNA may be used to produce cDNA for use in the polymerase chain reaction (PCR) or cDNA cloning of antibody genes.
  • the nucleic acid molecule is isolated from a hybridoma that has as one of its fusion partners a human immunoglobulin-producing cell from a non-human transgenic animal.
  • the human immunoglobulin producing cell is isolated from a XenoMouse animal.
  • the human immunoglobulin-producing cell is from a non- human, non-mouse transgenic animal, as described above.
  • the nucleic acid is isolated from a non-human, non-transgenic animal
  • the nucleic acid molecules isolated from a non-human, non-transgenic animal may be used, e.g., for humanized antibodies.
  • a nucleic acid encoding a heavy chain of an anti- CD40 antibody of the invention can comprise a nucleotide sequence encoding a V H domain of the invention joined in-frame to a nucleotide sequence encoding a heavy chain constant domain from any source.
  • a nucleic acid molecule encoding a light chain of an anti-CD40 antibody of the invention can comprise a nucleotide sequence encoding a V L domain of the invention joined in-frame to a nucleotide sequence encoding a light chain constant domain from any source.
  • nucleic acid molecules encoding the variable domain of the heavy (V H ) and light (V L ) chains are "converted" to full- length antibody genes.
  • nucleic acid molecules encoding the VH or V L domains are converted to full-length antibody genes by insertion into an expression vector already encoding heavy chain constant or light chain constant domains, respectively, such that the V H segment is operatively linked to the CH segment(s) within the vector, and the VL segment is operatively linked to the CL segment within the vector.
  • nucleic acid molecules encoding the VH and/or V L domains are converted into full-length antibody genes by linking, e.g., ligating, a nucleic acid molecule encoding a VH and/or VL domains to a nucleic acid molecule encoding a CH and/or CL domain using standard molecular biological techniques.
  • Nucleic acid sequences of human heavy and light chain immunoglobulin constant domain genes are known in the art. See, e.g.,
  • nucleic acid molecules encoding the full-length heavy and/or light chains may then be expressed from a cell into which they have been introduced and the anti-CD40 antibody isolated.
  • the nucleic acid molecules may be used to recombinantly express large quantities of anti-CD40 antibodies.
  • the nucleic acid molecules also may be used to produce chimeric antibodies, bispecific antibodies, single chain antibodies, immunoadhesins, diabodies, mutated antibodies and antibody derivatives, as described further below. If the nucleic acid molecules are derived from a non- human, non-transgenic animal, the nucleic acid molecules may be used for antibody humanization, also as described below.
  • a nucleic acid molecule of the invention is used as a probe or PCR primer for a specific antibody sequence.
  • the nucleic acid can be used as a probe in diagnostic methods or as a PCR primer to amplify regions of DNA that could be used, inter alia, to isolate additional nucleic acid molecules encoding variable domains of anti-CD40 antibodies.
  • the nucleic acid molecules are oligonucleotides.
  • the oligonucleotides are from highly variable regions of the heavy and light chains of the antibody of interest, hi some embodiments, the oligonucleotides encode all or a part of one or more of the CDRs of antibody 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-V88A-V97A, 3.1.1L-L4M-L83N, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H- D16E, 23.28.1L-C92A, 23.29.1 or 24.2.1.
  • the invention provides vectors comprising nucleic acid molecules that encode the heavy chain of an anti-CD40 antibody of the invention or an antigen- binding portion thereof.
  • the invention also provides vectors comprising nucleic acid molecules that encode the light chain of such antibodies or antigen-binding portion thereof.
  • the invention further provides vectors comprising nucleic acid molecules encoding fusion proteins, modified antibodies, antibody fragments, and probes thereof.
  • the anti-CD40 antibodies, or antigen-binding portions of the invention are expressed by inserting D ⁇ As encoding partial or full-length light and heavy chains, obtained as described above, into expression vectors such that the genes are operatively linked to necessary expression control sequences such as transcriptional and translational control sequences.
  • Expression vectors include plasmids, retroviruses, adenovirases, adeno-associated viruses (AAN), plant viruses such as cauliflower mosaic virus, tobacco mosaic virus, cosmids, YACs, EBN derived episomes, and the like.
  • the antibody gene is ligated into a vector such that transcriptional and translational control sequences within the vector serve their intended function of regulating the transcription and translation of the antibody gene.
  • the expression vector and expression control sequences are chosen to be compatible with the expression host cell used.
  • the antibody light chain gene and the antibody heavy chain gene can be inserted into separate vectors. In a prefened embodiment, both genes are inserted into the same expression vector.
  • a convenient vector is one that encodes a functionally complete human C H or C L immunoglobulin sequence, with appropriate restriction sites engineered so that any N ⁇ or N L sequence can easily be inserted and expressed, as described above.
  • splicing usually occurs between the splice donor site in the inserted J region and the splice acceptor site preceding the human C domain, and also at the splice regions that occur within the human C H exons. Polyadenylation and transcription termination occur at native chromosomal sites downstream of the coding regions.
  • the recombinant expression vector also can encode a signal peptide that facilitates secretion of the antibody chain from a host cell.
  • the antibody chain gene may be cloned into the vector such that the signal peptide is linked in-frame to the amino terminus of the immunoglobulin chain.
  • the signal peptide can be an immunoglobulin signal peptide or a heterologous signal peptide (i.e., a signal peptide from a non-immunoglobulin protein).
  • the recombinant expression vectors of the invention cany regulatory sequences that control the expression of the antibody chain genes in a host cell.
  • Prefened regulatory sequences for mammalian host cell expression include viral elements that direct high levels of protein expression in mammalian cells, such as promoters and/or enhancers derived from retroviral LTRs, cytomegalovirus (CMN) (such as the CMV promoter/enhancer), Simian Virus 40 (SV40) (such as the SV40 promoter/enhancer), adenovirus, (e.g., the adenovirus major late promoter (AdMLP)), polyoma and strong mammalian promoters such as native immunoglobulin and actin promoters.
  • CCN cytomegalovirus
  • SV40 Simian Virus 40
  • AdMLP adenovirus major late promoter
  • the recombinant expression vectors of the invention may cany additional sequences, such as sequences that regulate replication of the vector in host cells (e.g., origins of replication) and selectable marker genes.
  • the selectable marker gene facilitates selection of host cells into which the vector has been introduced (see e.g., U.S. Patent Nos. 4,399,216, 4,634,665 and 5,179,017).
  • the selectable marker gene confers resistance to drags, such as G418, hygromycin or methofrexate, on a host cell into which the vector has been introduced.
  • Prefened selectable marker genes include the dihydrofolate reductase (DHFR) gene (for use in dhfr-host cells with methofrexate selection/amplification), the neo gene (for G418 selection), and the glutamate synthetase gene.
  • DHFR dihydrofolate reductase
  • neo gene for G418 selection
  • glutamate synthetase gene for use in dhfr-host cells with methofrexate selection/amplification
  • Nucleic acid molecules encoding anti-CD40 antibodies and vectors comprising these nucleic acid molecules can be used for transfection of a suitable mammalian, plant, bacterial or yeast host cell. Transformation can be by any known method for introducing polynucleotides into a host cell.
  • Methods for introduction of heterologous polynucleotides into mammalian cells include dextran-mediated transfection, calcium phosphate precipitation, polybrene-mediated transfection, protoplast fusion, electroporation, encapsulation of the polynucleotide(s) in liposomes, and direct microinjection of the DNA into nuclei.
  • nucleic acid molecules may be introduced into mammalian cells by viral vectors.
  • Methods of transforming cells are well known in the art. See, e.g., U.S. Patent Nos. 4,399,216, 4,912,040, 4,740,461, and 4,959,455 (which patents are hereby incorporated herein by reference). Methods of transforming plant cells are well known in the art, including, e.g.,
  • Agrobacterium-mediated transformation biolistic transformation, direct injection, electroporation and viral transformation.
  • Methods of transforming b # acterial and yeast cells are also well known in the art.
  • Mammalian cell lines available as hosts for expression are well known in the art and include many immortalized cell lines available from the American Type
  • ATCC Culture Collection
  • CHO Chinese hamster ovary
  • NSO Chinese hamster ovary
  • SP2 cells
  • HeLa cells
  • BHK baby hamster kidney
  • COS monkey kidney cells
  • human hepatocellular carcinoma cells e.g., Hep G2
  • A549 cells A549 cells
  • cell lines of particular preference are selected through determining which cell lines have high expression levels.
  • Other cell lines that may be used are insect cell lines, such as Sf9 cells.
  • the antibodies When recombinant expression vectors encoding antibody genes are introduced into mammalian host cells, the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or, more preferably, secretion of the antibody into the culture medium in which the host cells are grown. Antibodies can be recovered from the culture medium using standard protein purification methods.
  • Plant host cells include, e.g., Nicotiana, Arabidopsis, duckweed, corn, wheat, potato, etc.
  • Bacterial host cells include E. coli and Streptomyces species.
  • Yeast host cells include Schizosaccharomyces pombe, Saccharomyces cerevisiae and Pichia pastoris.
  • GS system glutamine synthetase gene expression system
  • Anti-CD40 antibodies of the invention also can be produced transgenically through the generation of a mammal or plant that is transgenic for the immunoglobulin heavy and light chain sequences of interest and production of the antibody in a recoverable form therefrom.
  • anti-CD40 antibodies can be produced in, and recovered from, the milk of goats, cows, or other mammals. See, e.g., U.S. Patent Nos. 5,827,690, 5,756,687, 5,750,172, and 5,741,957.
  • non- human transgenic animals that comprise human immunoglobulin loci are immunized with CD40 or an immunogenic portion thereof, as described above. Methods for making antibodies in plants are described, e.g., in US patents 6,046,037 and US 5,959,177.
  • non-human transgenic animals or plants are produced by introducing one or more nucleic acid molecules encoding an anti- CD40 antibody of the invention into the animal or plant by standard transgenic techniques. See Hogan and United States Patent 6,417,429, supra.
  • the transgenic cells used for making the transgenic animal can be embryonic stem cells or somatic cells.
  • the transgenic non-human organisms can be chimeric, nonchimeric heterozygotes, and nonchimeric homozygotes.
  • the transgenic non-human animals have a targeted disruption and replacement by a targeting construct that encodes a heavy chain and/or a light chain of interest.
  • the transgenic animals comprise and express nucleic acid molecules encoding heavy and light chains that specifically bind to CD40, preferably human CD40.
  • the transgenic animals comprise nucleic acid molecules encoding a modified antibody such as a single-chain antibody, a chimeric antibody or a humanized antibody.
  • the anti- CD40 antibodies may be made in any transgenic animal.
  • the non-human animals are mice, rats, sheep, pigs, goats, cattle or horses.
  • the non-human transgenic animal expresses said encoded polypeptides in blood, milk, urine, saliva, tears, mucus and other bodily fluids.
  • the invention provides a method for producing an anti-CD40 antibody or antigen-binding portion thereof comprising the steps of synthesizing a library of human antibodies on phage, screening the library with CD40 or a portion thereof, isolating phage that bind CD40, and obtaining the antibody from the phage.
  • one method for preparing the library of antibodies for use in phage display techniques comprises the steps of immunizing a non-human animal comprising human immunoglobulin loci with CD40 or an antigenic portion thereof to create an immune response, extracting antibody producing cells from the immunized animal; isolating RNA from the extracted cells, reverse transcribing the RNA to produce cDNA, amplifying the cDNA using a primer, and inserting the cDNA into a phage display vector such that antibodies are expressed on the phage.
  • Recombinant anti-CD40 antibodies of the invention may be obtained in this way.
  • Recombinant anti-CD40 human antibodies of the invention can be isolated by screening a recombinant combinatorial antibody library.
  • the library is a scFv phage display library, generated using human N L and N ⁇ cD As prepared from mR ⁇ A isolated from B cells.
  • Methodologies for preparing and screening such libraries are known in the art.
  • kits for generating phage display libraries e.g., the Pharmacia Recombinant Phage Antibody System, catalog no. 27-9400-01; and the Stratagene SurfZAPTM phage display kit, catalog no. 240612).
  • kits for generating phage display libraries e.g., the Pharmacia Recombinant Phage Antibody System, catalog no. 27-9400-01; and the Stratagene SurfZAPTM phage display kit, catalog no. 240612).
  • methods and reagents that can be used in generating and screening antibody display libraries (see, e.g., U.S. Patent o. 5,223,409; PCT Publication Nos.
  • a human anti-CD40 antibody as described herein is first used to select human heavy and light chain sequences having similar binding activity toward CD40, using the epitope imprinting methods described in PCT Publication No. WO 93/06213.
  • the antibody libraries used in this method are preferably scFv libraries prepared and screened as described in PCT Publication No. WO 92/01047, McCafferty et al, Nature 348:552-554 (1990); and Griffiths et al, EMBOJ. 12:725-734 (1993).
  • the scFv antibody libraries preferably are screened using human CD40 as the antigen.
  • the N and N H segments of the prefened N L /N H pair(s) can be randomly mutated, preferably within the CDR3 region of N H and/or N L , in a process analogous to the in vivo somatic mutation process responsible for affinity maturation of antibodies during a natural immune response.
  • This in vitro affinity maturation can be accomplished by amplifying N H and N L domains using PCR primers complimentary to the N H CDR3 or N L CDR3, respectively, which primers have been "spiked” with a random mixture of the four nucleotide bases at certain positions such that the resultant PCR products encode N H and N segments into which random mutations have been introduced into the N H and/or N L CDR3 regions. These randomly mutated N ⁇ and N L segments can be rescreened for binding to CD40.
  • nucleic acids encoding the selected antibody can be recovered from the display package (e.g., from the phage genome) and subcloned into other expression vectors by standard recombinant D ⁇ A techniques. If desired, the nucleic acid can further be manipulated to create other antibody forms of the invention, as described below.
  • the D ⁇ A encoding the antibody is cloned into a recombinant expression vector and introduced into a mammalian host cells, as described above.
  • Another aspect of the invention provides a method for converting the class or subclass of an anti-CD40 antibody to another class or subclass.
  • a nucleic acid molecule encoding a N or N ⁇ that does not include any nucleic acid sequences encoding C L or C H is isolated using methods well- known in the art.
  • the nucleic acid molecule then is operatively linked to a nucleic acid sequence encoding a C L or C H from a desired immunoglobulin class or subclass. This can be achieved using a vector or nucleic acid molecule that comprises a C L or C H chain, as described above.
  • an anti-CD40 antibody that was originally IgM can be class switched to an IgG.
  • Another method for producing an antibody of the invention comprising a desired isotype comprises the steps of isolating a nucleic acid encoding a heavy chain of an anti-CD40 antibody and a nucleic acid encoding a light chain of an anti-CD40 antibody, isolating the sequence encoding the V H region, ligating the V H sequence to a sequence encoding a heavy chain constant domain of the desired isotype, expressing the light chain gene and the heavy chain construct in a cell, and collecting the anti-CD40 antibody with the desired isotype.
  • the antibody may be deimmunized using the techniques described in, e.g., PCT Publication Nos. WO98/52976 and WO00/34317 (which incorporated herein by reference in their entirety).
  • the nucleic acid molecules, vectors and host cells may be used to make mutated anti-CD40 antibodies.
  • the antibodies may be mutated in the variable domains of the heavy and/or light chains, e.g., to alter a binding property of the antibody.
  • a mutation may be made in one or more of the CDR regions to increase or decrease the K D of the antibody for CD40, to increase or decrease K off , or to alter the binding specificity of the antibody.
  • Techniques in site-directed mutagenesis are well-known in the art. See, e.g., Sambrook et al and Ausubel et al, supra.
  • mutations are made at an amino acid residue that is known to be changed compared to germline in a variable domain of an anti-CD40 antibody.
  • one or more mutations are made at an amino acid residue that is known to be changed compared to the germline in a CDR region or framework region of a variable domain, or in a constant domain of a monoclonal antibody 3.1.1, 3.1.1 H- A78T, 3.1.1H-A78T-V88A-V97A, 3.1.1L-L4M-L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K and 24.2.1.
  • one or more mutations are made at an amino acid residue that is known to be changed compared to the germline in a CDR region or framework region of a variable domain of an amino acid sequence selected from SEQ ID NOS: 4, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 94, 100, 102, 2, 10, 18, 26, 34, 42, 50, 58, 66, 74, 82, 90, 92, 96, 98, 100 or 102, or whose nucleic acid sequence is presented in SEQ ID NOS: 3, 11, 19, 27, 35, 43, 51, 59, 67, 75, 83, 93, 99, 101, 1, 9, 17, 25, 33, 41, 49, 57, 65, 73, 81, 89, 91, 95, 97, 99 or 101.
  • the framework region is mutated so that the resulting framework region(s) have the amino acid sequence of the conesponding germline gene.
  • a mutation may be made in a framework region or constant domain to increase the half-life of the anti-CD40 antibody. See, e.g., PCT Publication No. WO 00/09560, herein incorporated by reference.
  • a mutation in a framework region or constant domain also can be made to alter the immunogenicity of the antibody, to provide a site for covalent or non-covalent binding to another molecule, or to alter such properties as complement fixation, FcR binding and ADCC.
  • a single antibody may have mutations in any one or more of the framework regions, the constant domain and in the variable regions.
  • amino acid mutations there are from 1 to 18, including any number in between, amino acid mutations in either the V H or V L domains of the mutated anti- CD40 antibody compared to the anti-CD40 antibody prior to mutation.
  • the mutations may occur in one or more CDR regions.
  • any of the mutations can be conservative amino acid substitutions.
  • a fusion antibody or immunoadhesin may be made that comprises all or a portion of an anti-CD40 antibody of the invention linked to another polypeptide.
  • only the variable domains of the anti-CD40 antibody are linked to the polypeptide.
  • the V H domain of an anti-CD40 antibody is linked to a first polypeptide
  • the V L domain of an anti-CD40 antibody is linked to a second polypeptide that associates with the first polypeptide in a manner such that the V H and V L domains can interact with one another to form an antibody binding site.
  • the V H domain is separated from the V L domain by a linker such that the V H and V L domains can interact with one another (see below under Single Chain Antibodies).
  • the V H -linker-N antibody is then linked to the polypeptide of interest.
  • the fusion antibody is useful for directing a polypeptide to a CD40-ex ⁇ ressing cell or tissue.
  • the polypeptide may be a therapeutic agent, such as a toxin, growth factor or other regulatory protein, or may be a diagnostic agent, such as an enzyme that may be easily visualized, such as horseradish peroxidase.
  • fusion antibodies can be created in which two (or more) single-chain antibodies are linked to one another.
  • the N H - and N L -encoding D ⁇ A fragments are operatively linked to another fragment encoding a flexible linker, e.g., encoding the amino acid sequence (Gly 4 -Ser) 3 , such that the N H and N L sequences can be expressed as a contiguous single-chain protein, with the N and NH domains joined by the flexible linker.
  • a flexible linker e.g., encoding the amino acid sequence (Gly 4 -Ser) 3 , such that the N H and N L sequences can be expressed as a contiguous single-chain protein, with the N and NH domains joined by the flexible linker.
  • the single chain antibody may be monovalent, if only a single NH and N L are used, bivalent, if two N H and N L are used, or polyvalent, if more than two N H and N L are used. Bispecific or polyvalent antibodies may be generated that bind specifically to CD40 and to another molecule.
  • other modified antibodies may be prepared using anti-CD40 antibody-encoding nucleic acid molecules.
  • "Kappa bodies” 111 et al, Protein Eng. 10: 949-57 (1997)
  • “Minibodies” Martin et al, EMBO J. 13: 5303-9 (1994)
  • “Diabodies” HoUiger et al, Proc. Natl. Acad. Sci. USA 90: 6444-6448 (1993)
  • “Janusins” Traunecker et al, EMBOJ. 10:3655- 3659 (1991) and Traunecker et al, Int. J. Cancer (Suppl.) 7:51-52 (1992)
  • Bispecific antibodies or antigen-binding fragments can be produced by a variety of methods including fusion of hybridomas or linking of Fab' fragments. See, e.g., Songsivilai & Lachmann, Clin. Exp. Immunol. 79: 315-321 (1990), Kostelny et al, J Immunol. 148:1547-1553 (1992).
  • bispecific antibodies may be formed as "diabodies" or "Janusins.” In some embodiments, the bispecific antibody binds to two different epitopes of CD40.
  • the bispecific antibody has a first heavy chain and a first light chain from monoclonal antibody 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-N88A-N97A, 3.1.1L-L4M-L83N, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.28.1L-C92A, 23.29.1, 23.29.1L- R174K and 24.2.1, and an additional antibody heavy chain and light chain.
  • the additional light chain and heavy chain also are from one of the above-identified monoclonal antibodies, but are different from the first heavy and light chains.
  • the modified antibodies described above are prepared using one or more of the variable domains or CDR regions from a human anti-CD40 monoclonal antibody provided herein, from an amino acid sequence of said monoclonal antibody, or from a heavy chain or light chain encoded by a nucleic acid sequence encoding said monoclonal antibody.
  • An anti-CD40 antibody or antigen-binding portion of the invention can be derivatized or linked to another molecule (e.g., another peptide or protein).
  • another molecule e.g., another peptide or protein.
  • the antibodies or portion thereof is derivatized such that the CD40 binding is not affected adversely by the derivatization or labeling. Accordingly, the antibodies and antibody portions of the invention are intended to include both intact and modified forms of the human anti-CD40 antibodies described herein.
  • an antibody or antibody portion of the invention can be functionally linked (by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other molecular entities, such as another antibody (e.g., a bispecific antibody or a diabody), a detection agent, a cytotoxic agent, a pharmaceutical agent, and/or a protein or peptide that can mediate associate of the antibody or antibody portion with another molecule (such as a streptavidin core region or a polyhistidine tag).
  • another antibody e.g., a bispecific antibody or a diabody
  • a detection agent e.g., a cytotoxic agent, a pharmaceutical agent, and/or a protein or peptide that can mediate associate of the antibody or antibody portion with another molecule (such as a streptavidin core region or a polyhistidine tag).
  • One type of derivatized antibody is produced by crosslinking two or more antibodies (of the same type or of different types, e.g., to create bispecific antibodies).
  • Suitable crosslinkers include those that are heterobifunctional, having two distinctly reactive groups separated by an appropriate spacer (e.g., m-maleimidobenzoyl-N-hydroxysuccinimide ester) or homobifunctional (e.g., disuccinimidyl suberate).
  • Such linkers are available from Pierce Chemical Company, Rockford, 111.
  • Another type of derivatized antibody is a labeled antibody.
  • Useful detection agents with which an antibody or antigen-binding portion of the invention may be derivatized include fluorescent compounds, including fluorescein, fluorescein isothiocyanate, rhodamine, 5-dimethylamine-l-napthalenesulfonyl chloride, phycoerythrin, lanthanide phosphors and the like.
  • An antibody can also be labeled with enzymes that are useful for detection, such as horseradish peroxidase, j3-galactosidase, luciferase, alkaline phosphatase, glucose oxidase and the like.
  • an antibody When an antibody is labeled with a detectable enzyme, it is detected by adding additional reagents that the enzyme uses to produce a reaction product that can be discerned. For example, when the agent horseradish peroxidase is present, the addition of hydrogen peroxide and diaminobenzidine leads to a colored reaction product, which is detectable.
  • An antibody can also be labeled with biotin, and detected through indirect measurement of avidin or streptavidin binding.
  • An antibody can also be labeled with a predetermined polypeptide epitope recognized by a secondary reporter (e.g., leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags).
  • labels are attached by spacer arms of various lengths to reduce potential steric hindrance.
  • An anti-CD40 antibody can also be labeled with a radiolabeled amino acid.
  • the radiolabel can be used for both diagnostic and therapeutic purposes. For instance, the radiolabel can be used to detect CD40-expressing tumors by x-ray or other diagnostic techniques. Further, the radiolabel can be used therapeutically as a toxin for cancerous cells or tumors. Examples of labels for polypeptides include, but are not limited to, the following radioisotopes or radionuclides — 3 H, 14 C, 15 N,
  • An anti-CD40 antibody can also be derivatized with a chemical group such as polyethylene glycol (PEG), a methyl or ethyl group, or a carbohydrate group. These groups are useful to improve the biological characteristics of the antibody, e.g., to increase serum half-life or to increase tissue binding.
  • the invention also relates to compositions comprising a human anti- CD40 agonist antibody for the treatment of subjects in need of immunostimulation.
  • Such compositions are useful to treat, prevent, reduce the frequency of or severity of infection, including viral and bacterial infection, for treating a hyperproliferative disorder, including cancerous and pre-cancerous conditions, for treating genetic immunodeficiency conditions, such as hyper-IgM syndrome and for treating primary or combined immunodeficiency conditions, including conditions characterized by neutropenia, in a mammal, including humans.
  • Subjects for treatment with agonist anti-CD40 antibody therapy include any subject in need of immune enhancement, including but not limited to the elderly and individuals who are immunosuppressed, for example due to chemotherapy.
  • Hyperproliferative disorders that may be treated by an agonist anti-CD40 antibody of the invention can involve any tissue or organ and include but are not limited to brain, lung, squamous cell, bladder, gastric, pancreatic, breast, head, neck, liver, renal, ovarian, prostate, colorectal, esophageal, gynecological, nasopharynx, or thyroid cancers, melanomas, lymphomas, leukemias or multiple myelomas.
  • human agonist anti-CD40 antibodies of the invention are useful to treat carcinomas of the breast, prostate, colon and lung.
  • Treatment may involve administration of one or more agonist anti-CD40 monoclonal antibodies of the invention, or antigen-binding fragments thereof, alone or with a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier means any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • pharmaceutically acceptable carriers are water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof.
  • isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition.
  • additional examples of pharmaceutically acceptable substances are wetting agents or minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the antibody.
  • Agonist anti-CD40 antibodies of the invention and compositions comprising them can be administered in combination with one or more other therapeutic, diagnostic or prophylactic agents. Additional therapeutic agents include other anti-neoplastic, anti-tumor, anti-angiogenic or chemotherapeutic agents. Such additional agents may be included in the same composition or administered separately. In some embodiments, one or more agonist anti-CD40 antibodies of the invention can be used as a vaccine or as adjuvants to a vaccine.
  • compositions of this invention may be in a variety of forms, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, liposomes and suppositories.
  • liquid solutions e.g., injectable and infusible solutions
  • dispersions or suspensions tablets, pills, powders, liposomes and suppositories.
  • the prefened form depends on the intended mode of administration and therapeutic application. Typical prefened compositions are in the form of injectable or infusible solutions, such as compositions similar to those used for passive immunization of humans.
  • the prefened mode of administration is parenteral (e.g., intravenous, subcutaneous, intraperitoneal, intramuscular).
  • the antibody is administered by intravenous infusion or injection.
  • the antibody is administered by intramuscular or subcutaneous injection.
  • Therapeutic compositions typically must be sterile and stable under the conditions of manufacture and storage.
  • the composition can be formulated as a solution, microemulsion, dispersion, liposome, or other ordered stracture suitable to high drag concentration.
  • Sterile injectable solutions can be prepared by incorporating the anti-CD40 antibody in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the prefened methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prolonged absorption of injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, monostearate salts and gelatin.
  • the antibodies of the present invention can be administered by a variety of methods known in the art, although for many therapeutic applications, the prefened route/mode of administration is subcutaneous, intramuscular, or intravenous infusion. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results.
  • the antibody compositions active compound may be prepared with a carrier that will protect the antibody against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems.
  • a controlled release formulation including implants, transdermal patches, and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art.
  • an anti-CD40 antibody of the invention can be orally administered, for example, with an inert diluent or an assimilable edible carrier.
  • the compound (and other ingredients, if desired) can also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or incorporated directly into the subject's diet.
  • the anti-CD40 antibodies can be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • excipients for oral therapeutic administration, it may be necessary to coat the compound with, or co-administer the compound with, a material to prevent its inactivation.
  • an anti-CD40 antibody of the invention is co-formulated with and/or co-administered with one or more additional therapeutic agents.
  • agents include, without limitation, antibodies that bind other targets (e.g., antibodies that bind one or more growth factors or cytokines or their cell surface receptors, such as anti-CTL4-antibody), antineoplastic agents, antitumor agents, chemotherapeutic agents, peptide analogues that activate CD40, soluble CD40L, one or more chemical agents that activates CD40, and/or other agents known in the art that can enhance an immune response against tumor cells, e.g., IFN-jSl, IL-2, IL-8, IL-12, IL-15, IL-18, IL-23, IFN- ⁇ , and GM-CSF.
  • targets e.g., antibodies that bind one or more growth factors or cytokines or their cell surface receptors, such as anti-CTL4-antibody
  • antineoplastic agents e.g.,
  • Agonist anti-CD40 antibodies of the invention and compositions comprising them also may be administered in combination with other therapeutic regimens, in particular in combination with radiation treatment.
  • the compositions of the invention may include a "therapeutically effective amount” or a "prophylactically effective amount” of an antibody or antigen-binding portion of the invention.
  • a “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result.
  • a therapeutically effective amount of the antibody or antibody portion may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the antibody or antibody portion to elicit a desired response in the individual.
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the antibody or antibody portion are outweighed by the therapeutically beneficial effects.
  • a “prophylactically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.
  • Dosage regimens can be adjusted to provide the optimum desired response (e.g., a therapeutic or prophylactic response). For example, a single bolus can be administered, several divided doses can be administered over time or the dose can be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • An exemplary, non-limiting range for a therapeutically or prophylactically effective amount of an antibody or antibody portion of the invention is 0.025 to 50 mg/kg, more preferably 0.1 to 50 mg/kg, more preferably 0.1-25, 0.1 to 10 or 0.1 to 3 mg/kg. It is to be noted that dosage values may vary with the type and severity of the condition to be alleviated.
  • kits comprising an anti- CD40 antibody or antibody portion of the invention or a composition comprising such an antibody.
  • a kit may include, in addition to the antibody or composition, diagnostic or therapeutic agents.
  • a kit can also include instructions for use in a diagnostic or therapeutic method.
  • the kit includes the antibody or a composition comprising it and a diagnostic agent that can be used in a method described below.
  • the kit includes the antibody or a composition comprising it and one or more therapeutic agents that can be used in a method described below.
  • compositions for inhibiting abnormal cell growth in a mammal comprising an amount of an antibody of the invention in combination with an amount of a chemotherapeutic, wherein the amounts of the compound, salt, solvate, or prodrug, and of the chemotherapeutic are together effective in inhibiting abnormal cell growth.
  • chemotherapeutics are presently known in the art.
  • the chemotherapeutic is selected from the group consisting of mitotic inhibitors, alkylating agents, anti- metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biological response modifiers, anti-hormones, e.g. anti-androgens, and anti-angiogenesis agents.
  • Anti-angiogenic agents such as MMP-2 (matrix-metalloproteinase 2) inhibitors, MMP-9 (matrix-metalloproteinase 9) inhibitors, and COX-II (cyclooxygenase II) inhibitors, can be used in conjunction with an anti-CD40 antibody of the invention.
  • MMP-2 matrix-metalloproteinase 2
  • MMP-9 matrix-metalloproteinase 9
  • COX-II cyclooxygenase II
  • CELEBREXTM (alecoxib), valdecoxib, and rofecoxib.
  • Examples of useful matrix metalloproteinase inhibitors are described in WO 96/33172 (published October 24, 1996), WO 96/27583 (published March 7, 1996), European Patent Application No. 97304971.1 (filed July 8, 1997), European Patent Application No. 99308617.2 (filed October 29, 1999), WO 98/07697 (published February 26, 1998), WO
  • MMP inhibitors are those that do not demonstrate arthralgia. More prefened, are those that selectively inhibit MMP-2 and/or MMP- 9 relative to the other matrix-metalloproteinases (i.e.
  • MMP inhibitors useful in the present invention are AG- 3340, RO 32-3555, RS 13-0830, and the compounds recited in the following list: 3-[[4-(4-fluoro-phenoxy)-benzenesulfonyl]-(l-hydroxycarbamoyl-cyclopentyl)- amino] -propionic acid; 3-exo-3-[4-(4-fluoro-phenoxy)-benzenesulfonylamino]-8- oxa-bicyclo [3.2.
  • a compound of the invention can also be used with signal transduction inhibitors, such as agents that can inhibit EGF-R (epidermal growth factor receptor) responses, such as EGF-R antibodies, EGF antibodies, and molecules that are EGF-R inhibitors; NEGF (vascular endothelial growth factor) inhibitors, such as NEGF receptors and molecules that can inhibit NEGF; and erbB2 receptor inhibitors, such as organic molecules or antibodies that bind to the erbB2 receptor, for example, HERCEPTI ⁇ TM (Genentech, Inc.).
  • EGF-R epidermal growth factor receptor
  • NEGF vascular endothelial growth factor
  • erbB2 receptor inhibitors such as organic molecules or antibodies that bind to the erbB2 receptor, for example, HERCEPTI ⁇ TM (Genentech, Inc.).
  • EGF-R inhibitors are described in, for example in WO 95/19970 (published July 27, 1995), WO 98/14451 (published April 9, 1998), WO 98/02434 (published January 22, 1998), and United States Patent 5,747,498 (issued May 5, 1998), and such substances can be used in the present invention as described herein.
  • EGFR-inhibiting agents include, but are not limited to, the monoclonal antibodies C225 and anti-EGFR 22Mab (ImClone Systems Incorporated), ABX-EGF (Abgenix/Cell Genesys), EMD-7200 (Merck KgaA), EMD-5590 (Merck KgaA), MDX-447/H-477 (Medarex Inc. and Merck KgaA), and the compounds ZD-1834, ZD-1838 and ZD-1839 (AstraZeneca), PKI-
  • VEGF inhibitors for example SU-5416 and SU-6668 (Sugen Inc.), SH- 268 (Schering), and NX-1838 (NeXstar) can also be combined with the compound of the present invention.
  • VEGF inhibitors are described in, for example in WO 99/24440 (published May 20, 1999), PCT International Application PCT/IB99/00797 (filed May 3, 1999), in WO 95/21613 (published August 17, 1995), WO 99/61422 (published December 2, 1999), United States Patent 5,834,504 (issued November 10, 1998), WO 98/50356 (published November 12, 1998), United States Patent 5,883,113 (issued March 16, 1999), United States Patent 5,886,020 (issued March 23, 1999), United States Patent 5,792,783 (issued August 11, 1998), WO 99/10349 (published March 4, 1999), WO 97/32856
  • VEGF inhibitors useful in the present invention are IM862 (Cytran Inc.); anti-VEGF monoclonal antibody of Genentech, Inc.; and angiozyme, a synthetic ribozyme from Ribozyme and Chiron. These and other VEGF inhibitors can be used in the present invention as described herein.
  • ErbB2 receptor inhibitors such as GW-282974 (Glaxo Wellcome pic), and the monoclonal antibodies AR-209 (Aronex Pharmaceuticals Inc.) and 2B-1 (Chiron), can furthermore be combined with the compound of the invention, for example those indicated in WO 98/02434 (published January 22, 1998), WO 99/35146 (published July 15, 1999), WO 99/35132 (published July 15, 1999), WO 98/02437 (published January 22, 1998), WO 97/13760 (published April 17, 1997), WO 95/19970 (published July 27, 1995), United States Patent 5,587,458 (issued
  • Anti-survival agents include anti-IGF-ER antibodies and anti-integrin agents, such as anti-integrin antibodies.
  • the invention provides diagnostic methods.
  • the anti- CD40 antibodies can be used to detect CD40 in a biological sample in vitro or in vivo.
  • the invention provides a method for diagnosing the presence or location of an CD40-expressing tumor in a subject in need thereof, comprising the steps of injecting the antibody into the subject, determining the expression of CD40 in the subject by localizing where the antibody has bound, comparing the expression in the subject with that of a normal reference subject or standard, and diagnosing the presence or location of the tumor.
  • the anti-CD40 antibodies can be used in a conventional immunoassay, including, without limitation, an ELISA, an RIA, FACS, tissue immunohistochemistry, Western blot or immunoprecipitation.
  • the anti-CD40 antibodies of the invention can be used to detect CD40 from humans.
  • the anti-CD40 antibodies can be used to detect CD40 from Old World primates such as cynomolgus and rhesus monkeys, chimpanzees and apes.
  • the invention provides a method for detecting CD40 in a biological sample comprising contacting a biological sample with an anti-CD40 antibody of the invention and detecting the bound antibody.
  • the anti-CD40 antibody is directly labeled with a detectable label.
  • the anti-CD40 antibody (the first antibody) is unlabeled and a second antibody or other molecule that can bind the anti-CD40 antibody is labeled.
  • a second antibody is chosen that is able to specifically bind the particular species and class of the first antibody.
  • the anti-CD40 antibody is a human IgG
  • the secondary antibody could be an anti-human- IgG.
  • Other molecules that can bind to antibodies include, without limitation,
  • Suitable labels for the antibody or secondary antibody have been disclosed supra, and include various enzymes, prosthetic groups, fluorescent materials, luminescent materials and radioactive materials.
  • suitable enzymes include horseradish peroxidase, alkaline phosphatase, /3-galactosidase, or acetylcholinesterase;
  • suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin;
  • suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; and examples of suitable radioactive material include 125 1, 131 1, 35 S or 3 H.
  • CD40 can be assayed in a biological sample by a competition immunoassay utilizing CD40 standards labeled with a detectable substance and an unlabeled anti-CD40 antibody.
  • a competition immunoassay utilizing CD40 standards labeled with a detectable substance and an unlabeled anti-CD40 antibody.
  • the biological sample, the labeled CD40 standards and the anti-CD40 antibody are combined and the amount of labeled CD40 standard bound to the unlabeled antibody is determined.
  • the amount of CD40 in the biological sample is inversely proportional to the amount of labeled CD40 standard bound to the anti-CD40 antibody.
  • the anti-CD40 antibodies can be used to detect CD40 in cells in cell culture.
  • the anti-CD40 antibodies are used to determine the amount of CD40 on the surface of cells that have been treated with various compounds.
  • This method can be used to identify compounds that are useful to activate or inhibit CD40.
  • one sample of cells is treated with a test compound for a period of time while another sample is left untreated. If the total level of CD40 is to be measured, the cells are lysed and the total CD40 level is measured using one of the immunoassays described above. The total level of CD40 in the treated versus the untreated cells is compared to determine the effect of the test compound.
  • a prefened immunoassay for measuring total CD40 levels is an ELISA or
  • a prefened immunoassay for determining cell surface levels of CD40 includes the steps of labeling the cell surface proteins with a detectable label, such as biotin or 125 I, immunoprecipitating the CD40 with an anti-CD40 antibody and then detecting the labeled CD40.
  • Another prefened immunoassay for determining the localization of CD40, e.g., cell surface levels, is by using immunohistochemistry.
  • immunoassays can be scaled up for high throughput screening in order to test a large number of compounds for either activation or inhibition of CD40.
  • the anti-CD40 antibodies of the invention can also be used to determine the levels of CD40 in a tissue or in cells derived from the tissue.
  • the tissue is a diseased tissue.
  • the tissue is a tumor or a biopsy thereof, hi some embodiments of the method, a tissue or a biopsy thereof is excised from a patient. The tissue or biopsy is then used in an immunoassay to determine, e.g., total CD40 levels, cell surface levels of CD40 or localization of CD40 by the methods discussed above.
  • the above-described diagnostic method can be used to determine whether a tumor expresses high levels of CD40, which could be indicative that the tumor is a target for treatment with anti-CD40 antibody.
  • the same method can also be used to monitor the effect of the treatment with anti-CD40 antibody by detecting cell death in the tumor.
  • the diagnostic method can also be used to determine whether a tissue or cell expresses insufficient levels of CD40 or activated CD40, and thus is a candidate for treatment with activating anti-CD40 antibodies, CD40L and or other therapeutic agents for increasing CD40 levels or activity.
  • the antibodies of the present invention can also be used in vivo to identify tissues and organs that express CD40.
  • the anti-CD40 antibodies are used to identify CD40-expressing tumors.
  • One advantage of using the human anti-CD40 antibodies of the present invention is that they may safely be used in vivo without eliciting an immune response to the antibody upon administration, unlike antibodies of non-human origin or with humanized antibodies.
  • the method comprises the steps of administering a detectably labeled an anti-CD40 antibody or a composition comprising them to a patient in need of such a diagnostic test and subjecting the patient to imaging analysis to determine the location of the CD40-expressing tissues.
  • Imaging analysis is well known in the medical art, and includes, without limitation, x-ray analysis, magnetic resonance imaging (MRI) or computed tomography (CE).
  • the antibody can be labeled with any agent suitable for in vivo imaging, for example a contrast agent, such as barium, which can be used for x-ray analysis, or a magnetic contrast agent, such as a gadolinium chelate, which can be used for MRI or CE.
  • labeling agents include, without limitation, radioisotopes, such as 99 Tc.
  • the anti-CD40 antibody will be unlabeled and will be imaged by administering a second antibody or other molecule that is detectable and that can bind the anti- CD40 antibody.
  • a biopsy is obtained from the patient to determine whether the tissue of interest expresses CD40.
  • invention provides therapeutic methods of using an anti-CD40 antibody of the invention.
  • a human agonist anti-CD40 antibody of the invention can be administered to a human or to a non-human mammal that expresses a cross- reacting CD40.
  • the antibody can be administered to such a non-human mammal (i.e., a primate, cynomolgus or rhesus monkey) for veterinary purposes or as an animal model of human disease.
  • a non-human mammal i.e., a primate, cynomolgus or rhesus monkey
  • Such animal models are useful for evaluating the therapeutic efficacy of antibodies of this invention.
  • the anti-CD40 antibody is administered to a subject who suffers from primary and/or combined immunodeficiencies, including CD40- dependent immunodeficiency with Hyper-IgM syndrome, Common Variable Immunodeficiency, Braton's Agammaglobulinemia, IgG subclass deficiencies, and X-linked SCID (common gamma chain mutations).
  • the anti-CD40 antibody is administered to treat a subject who is immunosuppressed, for example due to chemotherapy, or has an immune- debilitating disease, including any acquired immune deficiency disease, such as HIV.
  • the anti-CD40 antibody is administered to enhance the immunity of an elderly subject.
  • the anti-CD40 antibody is administered to treat a subject who has a bacterial, viral, fungal or parasitic infection.
  • a human agonist anti-CD40 antibody of the invention may be administered prophylactically to a subject who, because of age, illness or general poor health is susceptible to infection to prevent or to reduce the number or severity of infections.
  • the anti-CD40 antibody is administered to a subject who has a hyperproliferative disorder.
  • the anti-CD40 antibody is administered to treat a subject who has a tumor, i some embodiments, the tumor is CD40 positive. In some embodiments, the tumor is a CD40 negative. The tumor can be a solid tumor or a non-solid tumor such as lymphoma. In some embodiments, an anti-CD40 antibody is administered to a patient who has a tumor that is cancerous. In some embodiments, the antibody inhibits cancer cell proliferation, inhibits or prevents an increase in tumor weight or volume, and/or causes a decrease in tumor weight or volume.
  • Patients that can be treated with anti-CD40 antibodies or antibody portions of the invention include, but are not limited to, patients that have been diagnosed as having brain cancer, lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head and neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, gastric cancer, colorectal cancer, colon cancer, breast cancer, gynecologic tumors (e.g., uterine sarcomas, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina or carcinoma of the vulva), cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system (e.g., cancer of the thyroid, parathyroid or adrenal glands), sarcomas of soft tissues, leukemia, myeloma, multiple myeloma, cancer of the urethr
  • the antibody may be administered from three times daily to once every six months, and preferably may be admimstered via an oral, mucosal, buccal, intranasal, inhalable, intravenous, subcutaneous, intramuscular, parenteral, intratumor, transdermal or topical route.
  • the antibody can also be administered continuously via a minipump.
  • the antibody generally will be administered for as long as the tumor is present provided that the antibody causes the tumor or cancer to stop growing or to decrease in weight or volume.
  • the dosage of antibody generally will be in the range of 0.025 to 50 mg/kg, more preferably 0.1 to 50 mg/kg, more preferably 0.1-20 mg/kg, 0.1-10 mg/kg, 0.1-5 mg/kg or even more preferable 0.1-2 mg/kg..
  • the anti-CD40 antibody is administered as part of a therapeutic regimen that includes one or more additional antineoplastic drags or molecules to a patient who has a hyperproliferative disorder, such as cancer or a tumor.
  • exemplary antitumor agents include, but are not limited to, mitotic inhibitors, alkylating agents, anti-metabolites, intercalating agents, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biological response modifiers, anti-hormones, kinase inhibitors, matrix metalloprotease inhibitors, genetic therapeutics and anti-androgens.
  • the anti-CD40 antibody is administered with an antineoplastic agent, such as adriamycin or taxol
  • the anti-CD40 therapy is performed along with radiotherapy, chemotherapy, photodynamic therapy, surgery or other immunotherapy.
  • the anti-CD40 antibody is administered with one or more additional antibodies.
  • the anti-CD40 antibody can be administered with antibodies that are known to inhibit tumor or cancer cell proliferation. Such antibodies include, but are -not limited to, an antibody that inhibits CTLA4, erbB2 receptor, EGF-R, IGF-1R, CD20 or VEGF.
  • the anti-CD40 antibody is labeled with a radiolabel, an immunotoxin or a toxin, or is a fusion protein comprising a toxic peptide.
  • the anti-CD40 antibody or anti-CD40 antibody fusion protein directs the radiolabel, immunotoxin, toxin or toxic peptide to the tumor or cancer cell, hi a prefened embodiment, the radiolabel, immunotoxin, toxin or toxic peptide is internalized by the tumor or cancer cell after the anti-CD40 antibody binds to the CD40 on the surface of the cell.
  • the anti-CD40 antibody can be used therapeutically to induce apoptosis of specific cells in a patient.
  • the cells targeted for apoptosis are cancerous or tumor cells.
  • the invention provides a method of inducing apoptosis by administering an anti-CD40 antibody to a patient in need thereof.
  • the invention provides a method of administering an activating anti-CD40 antibody to a patient to increase CD40 activity.
  • An anti- CD40 antibody is administered with one or more other factors that increase CD40 activity.
  • factors include CD40L, and/or analogues of CD40L that activate CD40.
  • the anti-CD40 antibody is administered with one or more additional immune enhancing agents, including, without limitation IFN- ⁇ l, IL-2, IL-8, IL-12, IL-15, IL-18, IL-23, IFN- ⁇ , and GM-CSF.
  • additional immune enhancing agents including, without limitation IFN- ⁇ l, IL-2, IL-8, IL-12, IL-15, IL-18, IL-23, IFN- ⁇ , and GM-CSF.
  • a human agonist anti-CD40 antibody of the invention is used as an adjuvant to enhance the efficacy of a vaccine.
  • the anti-CD-40 antibody activates CD40 on antigen presenting cells, including B cells, dendritic cells and monocytes as well as enhancing the production of immunomodulatory molecules, such as cytokines and chemokines. The immunostimulatory effect of the antibody enhances the immune response of the vaccinated subject to the vaccine antigen.
  • the invention provides a method for generating a dendritic cell vaccine for cancer or for dendritic cell immunotherapy.
  • dendritic cells from a cancer patient axe cultured for 1-5 -days with tumor lysate or homogenate, tumor cells killed by inadiation or other means, or tumor specific antigens (e.g., peptides, idiotypes) and 1-10 ⁇ g ml of an anti-CD40 antibody.
  • the tumor antigen-pulsed dendritic cells are re-injected into the patient to stimulate anti-tumor immune responses, particularly anti-tumor CTL responses.
  • Monocyte-derived dendritic cells for use in the method can be obtained from a peripheral blood sample by culture in IL-4 and GM-CSF.
  • Dendritic cells also can be derived from the bone manow of a patient by magnetic purification or sorting of CD34 positive cells, followed by culture in IL-4 and GM-CSF.
  • the nucleic acid molecules of the instant invention can be administered to a patient in need thereof via gene therapy.
  • the therapy may be either in vivo or ex vivo.
  • nucleic acid molecules encoding both a heavy chain and a light chain are administered to a patient.
  • the nucleic acid molecules are administered such that they are stably integrated into chromosomes of B cells because these cells are specialized for producing antibodies.
  • precursor B cells are transfected or infected ex vivo and re-transplanted into a patient in need thereof.
  • precursor B cells or other cells are infected in vivo using a virus known to infect the cell type of interest.
  • Typical vectors used for gene therapy include liposomes, plasmids and viral vectors.
  • Exemplary viral vectors are retroviruses, adenoviruses and adeno-associated viruses. After infection either in vivo or ex vivo, levels of antibody expression can be monitored by taking a sample from the treated patient and using any immunoassay known in the art or discussed herein.
  • the gene therapy method comprises the steps of administering an isolated nucleic acid molecule encoding the heavy chain or an antigen-binding portion thereof of an anti-CD40 antibody and expressing the nucleic acid molecule.
  • the gene therapy method comprises the steps of administering an isolated nucleic acid molecule encoding the light chain or an antigen-binding portion thereof of an anti-CD40 antibody and expressing the nucleic acid molecule.
  • the gene therapy method comprises the steps of administering of an isolated nucleic acid molecule encoding the heavy chain or an antigen-binding portion thereof and an isolated nucleic acid molecule encoding the light chain or the antigen-binding portion thereof of an anti-CD40 antibody of the invention and expressing the nucleic acid molecules.
  • the gene therapy method may also comprise the step of administering another anti-cancer agent, such as taxol or adriamycin.
  • Antibodies of the invention were prepared, selected, and assayed as follows:
  • CAGGTGCAGCTGGAGCAGTCIGG (SEQ ID NO: 118), in conjunction with primers specific for the human Cj2 constant region , MG-40d, 5'-GCTGAGGGAGTAGAGTCCTGAGGA-3' (SEQ ID NO: 119) or CK constant region (hcP2; as previously described in Green et al, 1994).
  • MG-40d 5'-GCTGAGGGAGTAGAGTCCTGAGGA-3'
  • CK constant region CK constant region
  • RNA from approximately 4 X 10 6 hybridoma cells using QIAGEN RNeasy RNA isolation kit (QIAGEN).
  • QIAGEN QIAGEN RNeasy RNA isolation kit
  • Table 1 lists the forward amplification primers used to sequence the antibody clones.
  • Table 2 sets forth the gene utilization evidenced by selected hybridoma clones of antibodies in accordance with the invention: TABLE 2
  • Table A provides the sequence identifiers for each of the nucleotide and predicted amino acid sequences of the sequenced antibodies.
  • Tables 3-7 provide the nucleotide and predicted amino acid sequences of the heavy and kappa light chains of antibodies 3.1.1 (Table 3), 7.1.2 (Table 4), 10.8.3 (Table 5), 15.1.1 (Table 6) and 21.4.1 (Table 7).
  • Tables 8-13 provide the nucleotide and predicted amino acid sequences of the variable domain of the heavy chain and kappa light chain of antibodies 21.2.1 (Table 8), 22.1.1 (Table 9), 23.5.1 (Table 10), 23.28.1 (Table 11), 23.29.1 (Table 12) and 24.2.1 (Table l3).
  • the DNA sequence from the full-length sequencing of monoclonal antibody 23.28.1 differs from DNA sequences obtained from sequencing the NH region of the initial PCR product by one base pair (C to G), resulting in a change of residue 16 of the natural heavy chain from D to E.
  • Tables 14-19 provide the nucleotide and predicted amino acid sequences of the heavy and kappa light chains of antibodies 21.2.1 (Table 14), 22.1.1 (Table 15), 23.5.1 (Table 16), 23.28.1 (Table 17), 23.29.1 (Table 18) and 24.2.1 (Table 19).
  • the signal peptide sequence (or the bases encoding the same) are underlined.
  • Table 20 provides the nucleotide and amino acid sequences of the mutated heavy chain of antibody 22.1.1H-C109A.
  • Table 21 provides the nucleotide and amino acid sequences of the mutated light chain of antibody 23.28.1. The mutated D ⁇ A codons are shown in italics. The mutated amino acid residue is in bold.
  • Table 3 DNA and protein sequences of antibody 3.1.1
  • VYACEVTHQGLSSPVTKSF ⁇ RGEC Table 4 DNA and protein sequences of antibody 7.1.2
  • Table 8 DNA and protein sequences of mature variable domains of 21.2.1 antibody
  • Table 10 DNA and protein sequences of mature variable domains of 23.5.1 antibody
  • Table 12 DNA and protein sequences of mature variable domains of 23.29.1 antibody
  • Table 13 DNA and protein sequences of mature variable domains of 24.2.1 antibody
  • Table 18 DNA and protein sequences of antibody 23.29.1

Abstract

The present invention relates to antibodies and antigen-binding portions thereof that specifically bind to CD40, preferably human CD40, and that function as CD40 agonists. The invention also relates to human anti-CD40 antibodies and antigen-binding portions thereof. The invention also relates to antibodies that are chimeric, bispecific, derivatized, single chain antibodies or portions of fusion proteins. The invention also relates to isolated heavy and light chain immunoglobulins derived from human anti-CD40 antibodies and nucleic acid molecules encoding such immunoglobulins. The present invention also relates to methods of making human anti-CD40 antibodies, compositions comprising these antibodies and methods of using the antibodies and compositions for diagnosis and treatment. The invention also provides gene therapy methods using nucleic acid molecules encoding the heavy and/or light immunoglobulin molecules that comprise the human anti-CD40 antibodies. The invention also relates to transgenic animals comprising nucleic acid molecules of the present invention.

Description

ANTIBODIES TO CD40
[0001] This application claims the benefit of United States Provisional Application 60/348,980, filed November 9, 2001.
BACKGROUND OF THE INVENTION
[0002] The CD40 antigen is a 50 kDa cell surface glycoprotein which belongs to the Tumor Necrosis Factor Receptor (TNF-R) family. (Stamenkovic et al., EMBO J. 8:1403-10 (1989).) CD40 is expressed in many normal and tumor cell types, including B lymphocytes, dendritic cells, monocytes, macrophages, thymic epithelium, endothelial cells, fibroblasts, and smooth muscle cells. (Paulie S. et al., Cancer Immunol. Immunother. 20:23-8 (1985); Banchereau J. et al., Adv. Exp.
Med. & Biol. 378:79-83 (1995); Alderson M.R. et al., J. of Exp. Med. 178:669-74 (1993); Ruggiero G. et al., J of Immunol. 156:3737-46 (1996); HoUenbaugh D. et al., J. of Exp. Med. 182:33-40 (1995); Yellin M.j. et al, J of Leukocyte Biol. 58:209-16 (1995); and Lazaar A.L. et al., J of Immunol. 161:3120-7 (1998).) CD40 is expressed in all B-lymphomas and in 70% of all solid tumors. Although constitutively expressed, CD40 is up-regulated in antigen presenting cells by maturation signals, such as LPS, IL-lj8, -FN-γand GM-CSF. [0003] CD40 activation plays a critical role in regulating humoral and cellular immune responses. Antigen presentation without CD40 activation can lead to tolerance, while CD40 signaling can reverse such tolerance, enhance antigen presentation by all antigen presenting cells (APCs), lead to secretion of helper cytokines and chemokines, increase co-stimulatory molecule expression and signaling, and stimulate cytolytic activity of immune cells. [0004] CD40 plays a critical role in B cell proliferation, maturation and class switching. (Foy T.M. et a\., Ann. Rev. of Immunol. 14:591-617 (1996).) Disruption of the CD40 signaling pathway leads to abnormal serum immunoglobulin isotype distribution, lack of CD4+ T cell priming, and defects in secondary humoral responses. For example, the X-linked hyper-IgM syndrome is a disease associated with a mutation in the human CD40L gene, and it is characterized by the inability of affected individuals to produce antibodies other than those of the IgM isotype, indicating that the productive interaction between CD40 and CD40L is required for an effective immune response. [0005] CD40 engagement by CD40L leads to the association of the CD40 cytoplasmic domain with TRAFs (TNF-R associated factors). (Lee H.H. et al., Proc. Nαtl. Acαd. Sci. USA 96:1421-6 (1999); PuUen S.S. et al., Biochemistry 37:11836-45 (1998); Grammar A.C. et al., J of Immunol. 161:1183-93 (1998);
Ishida T.K. et al., Proc. Nαtl. Acαd. Sci. USA 93:9437-42 (1996); Pullen S.S. et al., J. of Biol. Chem. 274:14246-54 (1999)). The interaction with TRAFs can culminate in the activation of both NFKB and Jun/APl pathways. (Tsukamoto N. et al., Proc. Nαtl. Acαd. Sci. USA 96:1234-9 (1999); Sutherland C.L. et al, J. of Immunol. 162:4720-30 (1999).) Depending on cell type, this signaling leads to enhanced secretion of cytokines such as IL-6 (Jeppson LD. et al., J. of Immunol. 161:1738-42 (1998); Uejima Y. et al., Int. Arch, of Allergy & Immunol. 110:225-32, (1996), IL-8 (Grass H.J. et al., Blood 84:2305-14 (1994); von Leoprechting A. et al., Cancer Res. 59:1287-94 (1999); Denfeld R.W. et al., Europ. J. of Immunol. 26:2329-34 (1996)), IL-12 (Cella M. et al., J. of Exp . Med.
184:747-52 (1996); Ferlin W.G. et al., Europ. J. of Immunol. 28:525-31 (1998); Armant M. et al., Europ. J. of Immunol. 26:1430-4 (1996); Koch F. et al., J. of Exp. Med. 184:741-6 (1996); Seguin R. and L.H. Kasper, J. of Infect. Diseases 179:467-74 (1999); Chaussabel D. et al., Infection & Immunity 67:1929-34 (1999)), IL-15 (Kuniyoshi J.S. et al, Cellular Immunol. 193:48-58 (1999)) and chemokines (MIPlo MIP1/3, RANTES, and others) (McDyer LF. et al, J. of Immunol. 162:3711-7 (1999); Schaniel C. et al., J of Exp. Med. 188:451-63 (1998); Altenburg A. et al., J of Immunol. 162:4140-7 (1999); Deckers LG. et al, J of the Am. Society ofNephrology 9:1187-93 (1998)), increased expression of MHC class I and II (Santos-Argumedo L. et al., Cellular Immunol. 156:272-85 (1994)), and increased expression of adhesion molecules (e.g., ICAM) (Lee H.H. et al., Proc. Natl. Acad. Sci. USA. 96:1421-6 (1999); Grousson L et al., Archives of Dermatol. Res. 290:325-30 (1998); Katada Y. et al., Europ. J. of Immunol. 26:192-200 (1996); Mayumi M. et al., J. of Allergy & Clin. Immunol. 96:1136-44 (1995); Flores-Romo L. et al., Immunol. 79:445-51 (1993)) and costimulatory molecules (e.g., B7) (Roy M. et al., Europ. J. of Immunol. 25:596-603 (1995); Jones K.W. and C.J. Hackett, Cellular Immunol. 174:42-53 (1996); Caux C. et al., Journal of Exp. Med. 180:1263-72 (1994); Kiener P.A. et al., J. of Immunol. 155:4917-25 (1995)). Cytokines induced by CD40 engagement enhance T cell survival and activation. [0006] In addition to enhancement of cellular and immune function, the effects of CD40 activation include: cell recruitment and differentiation by chemokines and cytokines; activation of monocytes; increased cytolytic activity of cytolytic T lymphocyte (CTL) and natural killer (NK) cells; induction of apoptosis in CD40 positive tumors; enhancement of immunogenicity of CD40 positive tumors; and tumor-specific antibody production. The role of CD40 activation in cell-mediated immune responses is also well established, and it is reviewed in: Grewal et al.,
Ann. Rev. of Immunol. 16:111-35 (1998); Mackey et al., J. of Leukocyte Biol. 63:418-28 (1998); and Noelle RJ., -4ge..ts & Actions - Suppl. 49:17-22 (1998). [0007] Studies using a cross-priming model system showed that CD40 activation of APCs can replace helper T cell requirement for the generation of cytolytic T lymphocyte (CTL). (Bennett et al, Nature 393:478-480 (1998).) Evidence from CD40L deficient mice indicates a clear requirement for CD40 signaling in helper T cell priming. (Grewal LS. et al., Science 273:1864-7 (1996); Grewal LS. et al., Nature 378:617-20 (1995).) CD40 activation converts otherwise tolerogenic, antigen bearing B cells into competent APCs. (Buhlmann J.E. et al., Immunity 2:645-53 (1995).) CD40 activation induces maturation and differentiation of cord blood progenitors into dendritic cells. (Flores-Romo L. et al, J. of Exp. Med. 185:341-9 (1997); Mackey M.F. et al., J. of Immunol. 161 :2094-8 (1998).) CD40 activation also induces differentiation of monocytes into functional dendritic cells. (Brossart P. et al., Blood 92:4238-47 (1998).) Further, CD40 activation enhances cytolytic activity of NK cells through APC-CD40 induced cytokines. (Carbone E. et al., J. of Exp. Med. 185:2053-60 (1997); Martin-Fontecha A. et al., J. of Immunol. 162:5910-6 (1999).) These observations indicate that CD40 plays an essential role in the initiation and enhancement of immune responses by inducing maturation of APCs, secretion of helper cytokines, upregulation of costimulatory molecules, and enhancement of effector functions. [0008] The critical role of CD40 signaling in the initiation and maturation of humoral and cytotoxic immune responses makes this system an ideal target for immune enhancement. Such enhancement can be particularly important for mounting effective immune responses to tumor antigens, which are generally presented to the immune system through cross-priming of activated APCs. (Huang AN. et al., Ciba Foundation Symp. 187:229-44 (1994); Toes R.E.M. et al, Seminars in Immunol. 10:443-8 (1998); Albert M.L. et al., N-.t-.re 392:86-9 (1998); Bennett S.R. et al., J of Exp. Med. 186:65-70 (1997).) [0009] Several groups have demonstrated the effectiveness of CD40 activation for antitumor responses in vitro and in vivo. (Toes R.E.M. et al., Seminars in Immunol. 10:443-8 (1998).) Two groups, using lung metastatic model of renal cell carcinoma and subcutaneous tumors by virally transformed cells, have independently demonstrated that CD40 activation can reverse tolerance to tumor- specific antigens, resulting in efficient antitumor priming of T cells. (Sotomayor E.M. et al., Nature Medicine 5:780-787 (1999); Diehl L. et al., Nature Medicine 5:774-9 (1999).) Antitumor activity in the absence of immune cells was also reported by CD40L and anti-CD40 antibody treatment in a human breast cancer line model in SCID mice. (Hirano A. et al, Blood 93:2999-3007 (1999).) CD40 activation by anti-CD40 antibody was recently shown to eradicate CD40+ and CD40- lymphoma in mouse models. (French R.R. et al., Nature Medicine 5:548-53 (1999).) Furthermore, previous studies by Glennie and co-workers conclude that signaling activity by anti-CD40 antibodies is more effective for inducing in vivo tumor clearance than other anti-surface marker antibodies capable of recruiting effectors. (Tutt A.L. et al., J. of Immunol. 161:3176-85 (1998).) Consistent with these observations, when anti-CD40 antibodies were tested for activity against CD40+ tumor cells in vivo, most but not all of the tumoricidal activity was associated with CD40 signaling rather than ADCC. (Funakoshi S. et al., J. of Immunotherapy with Emphasis on Tumor Immunol. 19:93-101 (1996).) In another study, bone marrow dendritic cells were treated ex vivo with a variety of agents, and tested for in vivo antitumor activity. These studies demonstrated that CD40L stimulated DCs were the most mature and most effective cells that mounting an antitumor response. [0010] The essential role of CD40 in antitumor immunity has also been demonstrated by comparing responses of wild-type and CD40-/- mice to tumor vaccines. These studies show that CD40-/- mice are incapable of achieving the tumor immunity observed in normal mice. (Mackey M.F. et al., Cancer Research 51:2569-14 (1997).) In another study, splenocytes from tumor bearing mice were stimulated with tumor cells and treated with activating anti-CD40 antibodies ex vivo, and were shown to have enhanced tumor specific CTL activity. (Donepudi M. et al., Cancer Immunol. Immunother. 48:153-164 (1999).) These studies demonstrate that CD40 occupies a critical position in antitumor immunity, in both CD40 positive and negative tumors. Since CD40 is expressed in lymphomas, leukemias, multiple myeloma, a majority of carcinomas of nasopharynx, bladder, ovary, and liver, and some breast and colorectal cancers, activation of CD40 can have a broad range of clinical applications.
[0011] Anti-CD40 activating monoclonal antibodies can contribute to tumor eradication via several important mechanisms. Foremost among these is activation of host dendritic cells for enhanced tumor antigen processing and presentation, as well as enhanced antigen presentation or immunogenicity of CD40 positive tumor cells themselves, leading to activation of tumor specific CD4+ and CD8+ lymphocytes. Additional antitumor activity can be mediated by other immune- enhancing effects of CD40 signaling (production of chemokines and cytokines, recruitment and activation monocytes, and enhanced CTL and NK cytolytic activity), as well as direct killing of CD40+ tumors by induction of apoptosis or by stimulating a humoral response leading to ADCC. Apoptotic and dying tumor cells can also become an important source of tumor-specific antigens that are processed and presented by CD40 activated APCs.
Accordingly, there is a critical need for therapeutic, clinically relevant anti-CD40 agonist antibodies.
BRIEF DESCRIPTION OF THE DRAWINGS
[0012] Figures 1A-1H are sequence alignments of predicted amino acid sequences of isolated anti-CD40 monoclonal antibody light and heavy chain variable domains with the germline amino acid sequences of the corresponding light and heavy chain genes.). Differences between the clones and the germline sequence are indicated by shading. The germline CDR1, CDR2, and CDR3 sequences are underlined. In alignments of heavy chain sequences, apparent insertions to the CDR3 region are indicated by a dash (-) in the germline sequence and apparent deletions in the CDR3 region are indicated by a dash (-) in the clone sequence. [0013] Figure IA: the predicted kappa light chain variable region amino acid sequences of mAbs 3.1.1 and 7.1.2 with the Vκ=A3/A19 and J=J/<1 gene germline amino acid sequences.
[0014] Figure IB: the predicted kappa light chain variable region amino acid sequence from clone 15.1.1 and the germline amino acid sequence (Nκ=A3/A19 and J=Jκ2);
[0015] Figure IC: the predicted kappa light chain variable region amino acid sequences from mAbs 10.8.3 and 21.4.1 and the germline amino acid sequence
Figure imgf000007_0001
[0016] Figure ID: the predicted heavy chain variable region amino acid sequence from mAb 3.1.1 and the germline amino acid sequence (NH-3-30+ (DP -49), D=D4+DIR3 and J=JH6);
[0017] Figure IE: the predicted heavy chain variable region aminό acid sequence from mAb 7.1.2 and the germline amino acid sequence (NH=3-30+ (DP-49), D=DIR5+Dl-26 and J-JH6); [0018] Figure IF: the predicted heavy chain amino acid sequences from mAb 10.8.3 and the germline amino acid sequence (Nπ=4.35 (NIN-4), D=DIR3 and J=JH6);
[0019] Figure 1G: the predicted heavy chain variable region amino acid sequences from mAb 15.1.1 and the germline amino acid sequence (Nπ=4-59 (DP- 71), D=D4-23 and J=JH4); and
[0020] Figure 1H: the predicted heavy variable region chain amino acid sequences from mAb 21.4.1 and the germline amino acid sequence (NH=l-02 (DP- 75), D=DLR1 and J=JH4). [0021] Figure 2A-2H are sequence alignments of predicted amino acid sequences of isolated anti-CD40 monoclonal antibody light and heavy chain variable domains with the germline amino acid sequences of the corresponding light and heavy chain genes.). Differences between the clones and the germline sequence are indicated in bold. The germline CDR1, CDR2, and CDR3 sequences are underlined. In alignments of heavy chain sequences, apparent insertions to the CDR3 region are indicated by a dash (-) in the germline sequence and apparent deletions in the CDR3 region are indicated by a dash (-) in the clone sequence. [0022] Figure 2A: the predicted kappa light chain amino acid sequences from mAbs 22.1.1, 23.5.1 and 23.29.1 and the germline amino acid sequence (Vκ=A3/Al 9 and J=Jκ 1);
[0023] Figure 2B: the predicted kappa light chain amino acid sequence from mAb 21.2.1 and the germline amino acid sequence
Figure imgf000008_0001
[0024] Figure 2C: the predicted kappa light chain amino acid sequences from mAbs 23.28.1, 23.28.1L-C92A and 24.2.1 and the germline amino acid sequence (Nκ=A27 and J=Jκ3);
[0025] Figure 2D: the predicted heavy chain amino acid sequence from mAb
21.2.1 and the germline amino acid sequence (VH=3-30+, D=DIR3+D6-19 and
J=JH4);
[0026] Figure 2E: the predicted heavy chain amino acid sequence from mAbs 22.1.1, 22.1.1H-C109A and the germline amino acid sequence (NH=3-30+, D=D1-
1 and J=JH6); [0027] Figure 2F: the predicted heavy chain amino acid sequence from mAb 23.5.1 and the germline amino acid sequence (NH=3-30+, D=D4-17 and J=JH6); [0028] Figure 2G: the predicted heavy chain amino acid sequence from mAb 23.29.1 and the germline amino acid sequence (VH=3-30.3, D=D4-17 and J=JH6); and
[0029] Figure 2H: the predicted heavy chain amino acid sequences from mAb 23.28.1, 23.28.1H-D16E and 24.2.1 and the germline amino acid sequence (VH=4- 59, D=DIR1+D4-17 and J=JH5). [0030] Figure 3 is a dose-response curve that illustrates the ability of an anti- CD40 antibody of the invention (21.4.1) to enhance IL-12p40 production by human dendritic cells.
[0031] Figure 4 is a dose-response curve that illustrates the ability of an anti- CD40 antibody of the invention (21.4.1) to enhance IL-12p70 production by human dendritic cells. [0032] Figure 5 is a graph that illustrates the ability of an anti-CD40 antibody of the invention (21.4.1) to increase immunogenicity of Jy stimulator cells and enhance CTL activity against Jy target cells.
[0033] Figure 6 is a tumor growth inhibition curve that illustrates the reduced growth of CD40 positive Daudi tumors in SCID-beige mice treated with an anti- CD40 antibody of the invention (21.4.1).
[0034] Figure 7 is a tumor growth inhibition curve that illustrates the reduced growth of CD40 negative K562 tumors in SCID-beige mice treated with an anti- CD40 antibody of the invention (21.4.1) and human dendritic cells and T cells. [0035] Figure 8 shows inhibition in the growth of CD40 negative K562 tumors in SCID mice by different concentrations of anti-CD40 agonist mAb 23.29.1.
[0036] Figure 9 shows inhibition in the growth of CD40 negative K562 tumors in SCID mice by different concentrations of anti-CD40 agonist mAb 3.1.1. [0037] Figure 10 shows inhibition in the growth of CD40 positive Raji tumors in the presence and absence of T cells and dendritic cells in SCID mice by an anti- CD40 agonist mAb.
[0038] Figure 11 shows inhibition in the growth of CD40 positive Raji tumors in SCID mice by anti-CD40 agonist antibodies. [0039] Figure 12 shows inhibition in the growth of BT 474 breast cancer cells in SCID-beige mice by anti-CD40 agonist antibodies.
[0040] Figure 13 shows inhibition in the growth of PC-3 prostate tumors in SCID-beige mice by anti-CD40 agonist antibodies. [0041] Figure 14 is a survival curve for SCID-beige mice injected (iv) with Daudi tumor cells and treated with anti-CD40 agonist antibodies. [0042] Figure 15 is a Western blot analysis of anti-CD40 agonist antibodies to reduced (R) and non-reduced (NR) human CD40.
[0043] Figure 16 is an alignment of the D1-D4 domains of mouse and human CD40.
[0044] Figure 17 is an alignment of the mouse and human CD40 amino acid sequences showing the location of the fusion sites of the chimeras. [0045] Figure 18 is a group of schematic diagrams of the chimeric CD40 constructs.
SUMMARY OF THE INVENTION
[0046] The present invention provides an isolated antibody or antigen-binding portion thereof that binds CD40 and acts as a CD40 agonist. [0047] The invention provides a composition comprising the anti-CD40 antibody, or antigen binding portion thereof, and a pharmaceutically acceptable carrier. The composition may further comprise another component, such as an anti-tumor agent or an imaging agent. Diagnostic and therapeutic methods are also provided by the invention.
[0048] The invention provides an isolated cell line, such as a hybridoma, that produces an anti-CD40 antibody or antigen binding portion thereof. [0049] The invention also provides nucleic acid molecules encoding the heavy and/or light chain, or antigen-binding portions thereof, of an anti-CD40 antibody. [0050] The invention provides vectors and host cells comprising the nucleic acid molecules, as well as methods of recombinantly producing the polypeptides encoded by nucleic acid molecules. [0051] Non-human transgenic animals that express the heavy and/or light chain, or antigen-binding portions thereof, of an anti-CD40 antibody are also provided. [0052] The invention also provides a method for treating a subject in need thereof with an effective amount of a nucleic acid molecule encoding the heavy and/or light chain, or antigen-binding portions thereof, of an anti-CD40 antibody.
DETAILED DESCRIPTION OF THE INVENTION
Definitions and General Techniques
[0053] Unless otherwise defined herein, scientific and technical terms used in connection with the present invention shall have the meanings that are commonly understood by those of ordinary skill in the art. Further, unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular. Generally, nomenclatures used in connection with, and techniques of, cell and tissue culture, molecular biology, immunology, microbiology, genetics and protein and nucleic acid chemistry and hybridization described herein are those well known and commonly used in the art. [0054] The methods and techniques of the present invention are generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification unless otherwise indicated. See, e.g., Sambrook et al. Molecular Cloning: A Laboratory Manual, 2d ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989) and Ausubel et al., Current Protocols in Molecular Biology. Greene Publishing Associates (1992), and Harlow and Lane Antibodies: A Laboratory Manual Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1990), which are incorporated herein by reference. Enzymatic reactions and purification techniques are performed according to manufacturer's specifications, as commonly accomplished in the art or as described herein. The nomenclatures used in connection with, and the laboratory procedures and techniques of, analytical chemistry, synthetic organic
* chemistry, and medicinal and pharmaceutical chemistry described herein are those well known and commonly used in the art. Standard techniques are used for chemical syntheses, chemical analyses, pharmaceutical preparation, formulation, and delivery, and treatment of patients. [0055] The following terms, unless otherwise indicated, shall be understood to have the following meanings:
[0056] The term "polypeptide" encompasses native or artificial proteins, protein fragments and polypeptide analogs of a protein sequence. A polypeptide may be monomeric or polymeric.
[0057] The term "isolated protein", "isolated polypeptide" or "isolated antibody" is a protein, polypeptide or antibody that by virtue of its origin or source of derivation (1) is not associated with naturally associated components that accompany it in its native state, (2) is free of other proteins from the same species, (3) is expressed by a cell from a different species, or (4) does not occur in nature. Thus, a polypeptide that is chemically synthesized or synthesized in a cellular system different from the cell from which it naturally originates will be "isolated" from its naturally associated components. A protein may also be rendered substantially free of naturally associated components by isolation, using protein purification techniques well known in the art.
[0058] Examples of isolated antibodies include an anti-CD40 antibody that has been affinity purified using CD40, an anti-CD40 antibody that has been synthesized by a hybridoma or other cell line in vitro, and a human anti-CD40 antibody derived from a transgenic mouse. [0059] A protein or polypeptide is "substantially pure," "substantially homogeneous," or "substantially purified" when at least about 60 to 75% of a sample exhibits a single species of polypeptide. The polypeptide or protein may be monomeric or multimeric. A substantially pure polypeptide or protein will typically comprise about 50%, 60%, 70%, 80% or 90% W/W of a protein sample, more usually about 95%, and preferably will be over 99% pure. Protein purity or homogeneity may be indicated by a number of means well known in the art, such as polyacrylamide gel electrophoresis of a protein sample, followed by visualizing a single polypeptide band upon staining the gel with a stain well known in the art. For certain purposes, higher resolution may be provided by using HPLC or other means well known in the art for purification.
[0060] The term "polypeptide fragment" as used herein refers to a polypeptide that has an amino-terminal and/or carboxy-terminal deletion, but where the remaining amino acid sequence is identical to the corresponding positions in the naturally-occurring sequence. In some embodiments, fragments are at least 5, 6, 8 or 10 amino acids long. In other embodiments, the fragments are at least 14 , at least 20, at least 50, or at least 70, 80, 90, 100, 150 or 200 amino acids long. [0061] The term "polypeptide analog" as used herein refers to a polypeptide that comprises a segment that has substantial identity to a portion of an amino acid sequence and that has at least one of the following properties: (1) specific binding to CD40 under suitable binding conditions, (2) ability to activate CD40, (3) the ability to upregulate the expression of cell surface molecules such as ICAM, MHC- π, B7-1, B7-2, CD71, CD23 and CD83, or (4) the ability to enhance the secretion of cytokines such as IFN-/31, IL-2, IL-8, IL-12, IL-15, IL-18 and IL-23. Typically, polypeptide analogs comprise a conservative amino acid substitution (or insertion or deletion) with respect to the naturally-occurring sequence. Analogs typically are at least 20 or 25 amino acids long, preferably at least 50, 60, 70, 80, 90, 100, 150 or 200 amino acids long or longer, and can often be as long as a full-length naturally-occurring polypeptide.
[0062] Preferred amino acid substitutions are those which: (1) reduce susceptibility to proteolysis, (2) reduce susceptibility to oxidation, (3) alter binding affinity for forming protein complexes, and (4) confer or modify other physicochemical or functional properties of such analogs. Analogs can include various muteins of a sequence other than the naturally-occurring peptide sequence. For example, single or multiple amino acid substitutions (preferably conservative amino acid substitutions) may be made in the naturally-occurring sequence (preferably in the portion of the polypeptide outside the domain(s) forming intermolecular contacts). A conservative amino acid substitution should not substantially change the structural characteristics of the parent sequence (e.g., a replacement amino acid should not tend to break a helix that occurs in the parent sequence, or disrupt other types of secondary stracture that characterizes the parent sequence). Examples of art-recognized polypeptide secondary and tertiary structures are described in Proteins, Structures and Molecular Principles
(Creighton, Ed., W. H. Freeman and Company, New York (1984)); Introduction to Protein Structure (C. Branden and J. Tooze, eds., Garland Publishing, New York, NN. (1991)); and Thornton et al, Nαtwre 354:105 (1991), which are each incorporated herein by reference.
[0063] Νon-peptide analogs are commonly used in the pharmaceutical industry as drags with properties analogous to those of the template peptide. These types of non-peptide compound are termed "peptide mimetics" or "peptidomimetics." Fauchere, J. Adv. Drug Res. 15:29 (1986); Neber and Freidinger, TINSp.392 (1985); and Evans et al, J. Med. Chem. 30:1229 (1987), which are incorporated herein by reference. Such compounds are often developed with the aid of computerized molecular modeling. Peptide mimetics that are structurally similar to therapeutically useful peptides may be used to produce an equivalent therapeutic or prophylactic effect. Generally, peptidomimetics are structurally similar to a paradigm polypeptide (i.e., a polypeptide that has a desired biochemical property or pharmacological activity), such as a human antibody, but have one or more peptide linkages optionally replaced by a linkage selected from the group consisting of: -CH2ΝH-, -CH2S~, ~CH2-CH2~, -CH=CH-(cis and trans), ~COCH2-, ~CH(OH)CH2-, and -CH2SO-, by methods well known in the art. Systematic substitution of one or more amino acids of a consensus sequence with a D-amino acid of the same type (e.g., D-lysine in place of L-lysine) may also be used to generate more stable peptides. In addition, constrained peptides comprising a consensus sequence or a substantially identical consensus sequence variation may be generated by methods known in the art (Rizo and Gierasch, Ann. Rev. Biochem. 61:387 (1992), incorporated herein by reference); for example, by adding internal cysteine residues capable of forming intramolecular disulfide bridges which cyclize the peptide. [0064] An "antibody" refers to a complete antibody or to an antigen-binding portion thereof, that competes with the intact antibody for specific binding. See generally, Fundamental Immunology. Ch. 7 (Paul, W., ed., 2nd ed. Raven Press, N.Y. (1989)) (incorporated by reference in its entirety for all purposes). Antigen- binding portions may be produced by recombinant DNA techniques or by enzymatic or chemical cleavage of intact antibodies. Antigen-binding portions include, z.zter alia, Fab, Fab', F(ab')2, Fd, Fv, dAb, and complementarity determining region (CDR) fragments, single-chain antibodies (scFv), chimeric antibodies, diabodies and polypeptides that contain at least a portion of an antibody that is sufficient to confer specific antigen binding to the polypeptide. [0065] From N-terminus to C-terminus, both light and heavy chain variable domains comprise the regions FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4. The assignment of amino acids to each domain is in accordance with the definitions of Kabat, Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md. (1987 and 1991)), or Chothia & Lesk, J. Mol. Biol. 196:901-917 (1987); Chothia et al., Nature 342:878-883 (1989). [0066] As used herein, an antibody that is referred to by number is a monoclonal antibody that is obtained from the hybridoma of the same number. For example, monoclonal antibody 3.1.1 is obtained from hybridoma 3.1.1. [0067] As used herein, a Fd fragment means an antibody fragment that consists of the VH and CH 1 domains; an Fv fragment consists of the N and VH domains of a single arm of an antibody; and a dAb fragment (Ward et al., Nature 341:544-546 (1989)) consists of a VH domain.
[0068] In some embodiments, the antibody is a single-chain antibody (scFv) in which a VL and VH domains are paired to form a monovalent molecules via a synthetic linker that enables them to be made as a single protein chain. (Bird et al., Science 242:423-426 (1988) and Huston et al, Proc. Natl. Acad. Sci. USA 85:5879-5883 (1988).) In some embodiments, the antibodies are diabodies, i.e., are bivalent antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites. (See e.g., HoUiger P. et al., Proc. Natl. Acad. Sci. USA 90:6444-6448 (1993), and
Poljak R. J. et al., Structure 2:1121-1123 (1994).) In some embodiments, one or more CDRs from an antibody of the invention may be incorporated into a molecule either covalently or noncovalently to make it an immunoadhesin that specifically binds to CD40. In such embodiments, the CDR(s) may be incorporated as part of a larger polypeptide chain, may be covalently linked to another polypeptide chain, or may be incorporated noncovalently. [0069] hi embodiments having one or more binding sites, the binding sites may be identical to one another or may be different.
[0070] As used herein, the term "human antibody" means any antibody in which all of the variable and constant domain sequences are human sequences. These antibodies may be prepared in a variety of ways, as described below.
[0071] The term "chimeric antibody" as used herein means an antibody that comprises regions from two or more different antibodies. In one embodiment, one or more of the CDRs are derived from a human anti-CD40 antibody. In another embodiment, all of the CDRs are derived from a human anti-CD40 antibody. In another embodiment, the CDRs from more than one human anti-CD40 antibodies are combined in a chimeric antibody. For instance, a chimeric antibody may comprise a CDR1 from the light chain of a first human anti-CD40 antibody, a CDR2 from the light chain of a second human anti-CD40 antibody and a CDR3 and CDR3 from the light chain of a third human anti-CD40 antibody, and the CDRs from the heavy chain may be derived from one or more other anti-CD40 antibodies. Further, the framework regions may be derived from one of the same anti-CD40 antibodies or from one or more different human. [0072] An "activating antibody" (also referred to herein as an "agonist antibody" as used herein means an antibody that increases one or more CD40 activities by at least about 20% when added to a cell, tissue or organism expressing CD40. In some embodiments, the antibody activates CD40 activity by at least 40%, 50%, 60%, 70%), 80%), 85%. In some embodiments, the activating antibody is added in the presence of CD40L. In some embodiments, the activity of the activating antibody is measured using a whole blood surface molecule upregulation assay. See Example VII. In another embodiment, the activity of the activating antibody is measured using a dendritic cell assay to measure IL-12 release. See Example VIII. In another embodiment the activity of the activating antibody is measured using an in vivo tumor model. See Example X. *
[0073] Fragments or analogs of antibodies or immunoglobulin molecules can be readily prepared by those of ordinary skill in the art following the teachings of this specification. Preferred amino- and carboxy-termini of fragments or analogs occur near boundaries of functional domains. Structural and functional domains can be identified by comparison of the nucleotide and/or amino acid sequence data to public or proprietary sequence databases. Preferably, computerized comparison methods are used to identify sequence motifs or predicted protein conformation domains that occur in other proteins of known stracture and/or function. Methods to identify protein sequences that fold into a known three-dimensional stracture are known. See Bowie et al., Science 253:164 (1991).
[0074] The term "surface plasmon resonance", as used herein, refers to an optical phenomenon that allows for the analysis of real-time biospecific interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BIAcore system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, N.J.). For further descriptions, see Jonsson U. et al., Ann. Biol. Clin. 51:19-26 (1993); Jonsson U. et al., Biotechniques 11 :620-627 (1991); Jonsson B. et al., J. Mol. Recognit. 8:125-131 (1995); and Johnsson B. et al., Anal. Biochem. 198:268-277 (1991). [0075] The term "KD" refers to the equilibrium dissociation constant of a particular antibody-antigen interaction.
[0076] The term "epitope" includes any protein determinant capable of specific binding to an immunoglobulin or T-cell receptor. Epitopic determinants usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics. An antibody is said to specifically bind an antigen when the equilibrium dissociation constant is -.1 μM, preferably --100 nM and most preferably --10 nM. [0077] As used herein, the twenty conventional amino acids and their abbreviations follow conventional usage. See Immunology - A Synthesis (2nd
Edition, E.S. Golub and D.R. Gren, Eds., Sinauer Associates, Sunderland, Mass. (1991)), which is incorporated herein by reference.
[0078] The term "polynucleotide" as referred to herein means a polymeric form of nucleotides of at least 10 bases in length, either ribonucleotides or deoxynucleotides or a modified form of either type of nucleotide. The term includes single and double stranded forms. [0079] The term "isolated polynucleotide" as used herein means a polynucleotide of genomic, cDNA, or synthetic origin or some combination thereof, which by virtue of its origin the "isolated polynucleotide" (1) is not associated with all or a portion of a polynucleotides with which the "isolated polynucleotide" is found in nature, (2) is operably linked to a polynucleotide to which it is not linked in nature, or (3) does not occur in nature as part of a larger sequence. [0080] The term "oligonucleotide" as used herein includes naturally occurring, and modified nucleotides linked together by naturally occurring and non-naturally occurring oligonucleotide linkages. Oligonucleotides are a polynucleotide subset generally comprising a length of 200 bases or fewer. Preferably oligonucleotides are 10 to 60 bases in length and most preferably 12, 13, 14, 15, 16, 17, 18, 19, or 20 to 40 bases in length. Oligonucleotides are usually single stranded, e.g. for primers and probes; although oligonucleotides may be double stranded, e.g. for use in the construction of a gene mutant. Oligonucleotides of the invention can be either sense or antisense oligonucleotides.
[0081] The term "naturally occurring nucleotides" as used herein includes deoxyribonucleotides and ribonucleotides. The term "modified nucleotides" as used herein includes nucleotides with modified or substituted sugar groups and the like. The term "oligonucleotide linkages" referred to herein includes oligonucleotides linkages such as phosphorothioate, phosphorodithioate, phosphoroselenoate, phosphorodiselenoate, phosphoroanilothioate, phoshoraniladate, phosphoroamidate, and the like. See e.g., LaPlanche et al., Nucl. Acids Res. 14:9081 (1986); Stec et al., J. Am. Chem. Soc. 106:6077 (1984); Stein et al., Nucl. Acids Res. 16:3209 (1988); Zon et al., Anti-Cancer Drug Design 6:539 (1991); Zon et al.. Oligonucleotides and Analogues: A Practical Approach. pp. 87-108 (F. Eckstein, Ed., Oxford University Press, Oxford England (1991)); U.S. Patent No. 5,151,510; Uhlmann and Peyman, Chemical Reviews 90:543 (1990), the disclosures of which are hereby incorporated by reference. An oligonucleotide can include a label for detection, if desired. [0082] "Operably linked" sequences include both expression control sequences that are contiguous with the gene of interest and expression control sequences that act in trans or at a distance to control the gene of interest. The term "expression control sequence" as used herein means polynucleotide sequences that are necessary to effect the expression and processing of coding sequences to which they are ligated. Expression control sequences include appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation signals; sequences that stabilize cytoplasmic mRNA; sequences that enhance translation efficiency (i.e., Kozak consensus sequence); sequences that enhance protein stability; and when desired, sequences that enhance protein secretion. The nature of such control sequences differs depending upon the host organism; in prokaryotes, such control sequences generally include promoter, ribosomal binding site, and transcription termination sequence; in eukaryotes, generally, such control sequences include promoters and transcription termination sequence. The term "control sequences" is intended to include, at a minimum, all components whose presence is essential for expression and processing, and can also include additional components whose presence is advantageous, for example, leader sequences and fusion partner sequences.
[0083] The term "vector", as used herein, means a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. In some embodiments, the vector is a plasmid, i.e., a circular double stranded DNA loop into which additional DNA segments may be ligated. In some embodiments, the vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome. In some embodiments, the vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). In other embodiments, the vectors (e.g., non-episomal mammalian vectors) can be integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as "recombinant expression vectors" (or simply, "expression vectors").
[0084] The term "recombinant host cell" (or simply "host cell"), as used herein, means a cell into which a recombinant expression vector has been introduced. It should be understood that "recombinant host cell" and "host cell" mean not only the particular subject cell but also the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term "host cell" as used herein. [0085] The term "selectively hybridize" referred to herein means to detectably and specifically bind. Polynucleotides, oligonucleotides and fragments thereof in accordance with the invention selectively hybridize to nucleic acid strands under hybridization and wash conditions that minimize appreciable amounts of detectable binding to nonspecific nucleic acids. "High stringency" or "highly stringent" conditions can be used to achieve selective hybridization conditions as known in the art and discussed herein. One example of "high stringency" or "highly stringent" conditions is the incubation of a polynucleotide with another polynucleotide, wherein one polynucleotide may be affixed to a solid surface such as a membrane, in a hybridization buffer of 6X SSPE or SSC, 50% formamide, 5X Denhardt's reagent, 0.5% SDS, 100 j-ig/ml denatured, fragmented salmon sperm DNA at a hybridization temperature of 42°C for 12-16 hours, followed by twice washing at 55°C using a wash buffer of IX SSC, 0.5% SDS. See also Sambrook et al., supra, pp. 9.50-9.55. [0086] The term "percent sequence identity" in the context of nucleic acid sequences means the residues in two sequences that are the same when aligned for maximum correspondence. The length of sequence identity comparison may be over a stretch of at least about nine nucleotides, usually at least about 18 nucleotides, more usually at least about 24 nucleotides, typically at least about 28 nucleotides, more typically at least about 32 nucleotides, and preferably at least about 36, 48 or more nucleotides. There are a number of different algorithms known in the art which can be used to measure nucleotide sequence identity. For instance, polynucleotide sequences can be compared using FASTA,' Gap or Bestfit, which are programs in Wisconsin Package Version 10.0, Genetics Computer Group (GCG), Madison, Wisconsin. FASTA, which includes, e.g., the programs
FASTA2 and FASTA3, provides alignments and percent sequence identity of the regions of the best overlap between the query and search sequences (Pearson, Methods Enzymol. 183:63-98 (1990); Pearson, Methods Mol. Biol. 132:185-219 (2000); Pearson, Methods Enzymol. 266:227-258 (1996); Pearson, J. Mol. Biol. 276:71-84 (1998); herein incorporated by reference). Unless otherwise specified, default parameters for a particular program or algorithm are used. For instance, percent sequence identity between nucleic acid sequences can be deteraiined using FASTA with its default parameters (a word size of 6 and the NOP AM factor for the scoring matrix) or using Gap with its default parameters as provided in GCG Version 6.1, herein incorporated by reference. [0087] A reference to a nucleotide sequence encompasses its complement unless otherwise specified. Thus, a reference to a nucleic acid having a particular sequence should be understood to encompass its complementary strand, with its complementary sequence.
[0088] In the molecular biology art, researchers use the terms "percent sequence identity", "percent sequence similarity" and "percent sequence homology" interchangeably. In this application, these terms shall have the same meaning with respect to nucleic acid sequences only.
[0089] The term "substantial similarity" or "substantial sequence similarity," when referring to a nucleic acid or fragment thereof, means that when optimally aligned with appropriate nucleotide insertions or deletions with another nucleic acid (or its complementary strand), there is nucleotide sequence identity in at least about 85%, preferably at least about 90%, and more preferably at least about 95%, 96%, 97%, 98% or 99% of the nucleotide bases, as measured by any well-known algorithm of sequence identity, such as FASTA, BLAST or Gap, as discussed above. [0090] As applied to polypeptides, the term "substantial identity" means that two peptide sequences, when optimally aligned, such as by the programs GAP or BESTFIT using default gap weights, share at least 70, 75 or 80 percent sequence identity, preferably at least 90 or 95 percent sequence identity, and more preferably at least 97, 98 or 99 percent sequence identity. Preferably, residue positions that are not identical differ by conservative amino acid substitutions. A "conservative amino acid substitution" is one in which an amino acid residue is substituted by another amino acid residue having a side chain R group) with similar chemical properties (e.g., charge or hydrophobicity). In general, a conservative amino acid substitution will not substantially change the functional properties of a protein. In cases where two or more amino acid sequences differ from each other by conservative substitutions, the percent sequence identity or degree of similarity may be adjusted upwards to correct for the conservative nature of the substitution. Means for making this adjustment are well-known to those of skill in the art. See, e.g., Pearson, Methods Mol. Biol. 243:307-31 (1994). Examples of groups of amino acids that have side chains with similar chemical properties include 1) aliphatic side chains: glycine, alanine, valine, leucine, and isoleucine; 2) aliphatic-hydroxyl side chains: serine and threonine; 3) amide-containing side chains: asparagine and glutamine; 4) aromatic side chains: phenylalanine, tyrosine, and tryptophan; 5) basic side chains: lysine, arginine, and histidine; 6) acidic side chains: aspartic acid and glutamic acid; and 7) sulfur-containing side chains: cysteine and methionine. Preferred conservative amino acids substitution groups are: valine-leucine-isoleucine, phenylalanine-tyrosine, lysine-arginine, alanine-valine, glutamate-aspartate, and asparagine-glutamine. [0091] Alternatively, a conservative replacement is any change having a positive value in the PAM250 log-likelihood matrix disclosed in Gonnet et al., Science 256:1443-45 (1992), herein incorporated by reference. A "moderately conservative" replacement is any change having a nonnegative value in the
PAM250 log-likelihood matrix.
[0092] Sequence similarity for polypeptides, which is also referred to as sequence identity, is typically measured using sequence analysis software. Protein analysis software matches similar sequences using measures of similarity assigned to various substitutions, deletions and other modifications, including conservative amino acid substitutions. For instance, GCG contains programs such as "Gap" and "Bestfit" which can be used with default parameters to determine sequence homology or sequence identity between closely related polypeptides, such as homologous polypeptides from different species of organisms or between a wild type protein and a mutein thereof. See, e.g., GCG Version 6.1. Polypeptide sequences also can be compared using FASTA using default or recommended parameters, a program in GCG Version 6.1. FASTA (e.g., FASTA2 and FASTA3) provides alignments and percent sequence identity of the regions of the best overlap between the query and search sequences (Pearson, Methods Enzymol. 183:63-98 (1990); Pearson, Methods Mol. Biol. 132:185-219 (2000)). Another preferred algorithm when comparing a sequence of the invention to a database containing a large number of sequences from different organisms is the computer program BLAST, especially blastp or tblastn, using default parameters. See, e.g., Altschul et al., J. Mol. Biol. 215:403-410 (1990); Altschul et al., Nucleic Acids Res. 25:3389-402 (1997); herein incorporated by reference. [0093] The length of polypeptide sequences compared for homology will generally be at least about 16 amino acid residues, usually at least about 20 residues, more usually at least about 24 residues, typically at least about 28 residues, and preferably more than about 35 residues. When searching a database containing sequences from a large number of different organisms, it is preferable to compare amino acid sequences. [0094] As used herein, the terms "label" or "labeled" refers to incorporation of another molecule in the antibody. In one embodiment, the label is a detectable marker, e.g., incorporation of a radiolabeled amino acid or attachment to a polypeptide of biotinyl moieties that can be detected by marked avidin (e.g., streptavidin containing a fluorescent marker or enzymatic activity that can be detected by optical or colorimetric methods). In another embodiment, the label or marker can be therapeutic, e.g., a drag conjugate or toxin. Various methods of labeling polypeptides and glycoproteins are known in the art and may be used. Examples of labels for polypeptides include, but are not limited to, the following: radioisotopes or radionuclides (e.g., 3H, 14C, 15N, 35S, 90Y, 99Tc, n lIn, 125I, I31I), fluorescent labels (e.g., FITC, rhodamine, lanthanide phosphors), enzymatic labels
(e.g., horseradish peroxidase, /5-galactosidase, luciferase, alkaline phosphatase), chemiluminescent markers, biotinyl groups, predetermined polypeptide epitopes recognized by a secondary reporter (e.g., leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags), magnetic agents, such as gadolinium chelates, toxins such as pertussis toxin, taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorabicin, daunorabicin, dihydroxy anthracin dione, mitoxantrone, mithrarnycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof. In some embodiments, labels are attached by spacer arms of various lengths to reduce potential steric hindrance.
[0095] The term patient includes human and veterinary subjects. [0096] Throughout this specification and claims, the word "comprise," or variations such as "comprises" or "comprising," will be understood to imply the inclusion of a stated integer or group of integers but not the exclusion of any other integer or group of integers.
Human Anti-CD40 Antibodies and Characterization Thereof
[0097] Human antibodies avoid certain of the problems associated with antibodies that possess non-human (e.g., rodent) variable and/or constant regions. Such problems include the rapid clearance of the antibodies or immune response against the antibody. Therefore, in one embodiment, the invention provides humanized anti-CD40 antibodies. In another embodiment, the invention provides human anti-CD40 antibodies. In some embodiments, human anti-CD40 antibodies are produced by immunizing a rodent whose genome comprises human immunoglobulin genes so that the rodent produces human antibodies. Human anti- CD40 antibodies are expected to minimize the immunogenic and allergic responses intrinsic to non-human or non-human-derivatized monoclonal antibodies (Mabs) and thus to increase the efficacy and safety of the administered antibodies. The use of fully human antibodies can be expected to provide a substantial advantage in the treatment of chronic and recurring human diseases, such as inflammation and cancer, which may require repeated antibody administrations.
[0098] The invention provides eleven activating human anti-CD40 monoclonal antibodies (mAbs) and the hybridoma cell lines that produce them. able A lists the sequence identifiers (SEQ ID NOS:) of the nucleic acids encoding the full- length heavy and light chains (including leader sequence), the corresponding full- length deduced amino acid sequences, and the nucleotide and deduced amino acid sequence of the heavy and light chain variable regions. Table A
Figure imgf000025_0001
[0099] The invention further provides human anti-CD40 mAb 23.25.1 and the hybridoma cell line that produces it. [0100] The invention further provides heavy and/or light chain variants of certain of the above-listed human anti-CD40 mAbs, comprising one or more amino acid substitutions. The invention provides two variant heavy chains of mAb 3.1.1. In one, the alanine at residue 78 is changed to threonine. In the second, the alanine at residue 78 is changed to threonine, and the valines at residues 88 and 97 are changed to alanines. The invention also provides a variant light chain of mAb
3.1.1 in which the leucine at residue 4 and the leucine at residue 83 are changed to methionine and valine, respectively. Combination with a variant heavy or light chain with a wild type light or heavy chain, respectively is designated by the mutant chain. Thus, an antibody containing a wild type light chain and a heavy chain comprising the alanine to threonine mutation at residue 78 is designated as 3.1.1H-A78T. However, in other embodiments of the invention, antibodies containing any combination of a variant heavy chain and the variant light chain of 3.1.1 are included. [0101] Further, the invention provides a variant of the heavy chain of mAb 22.1.1 in which the cysteine at residue 109 is changed to an alanine. A monoclonal antibody comprising the variant heavy chain and the 22.1.1 light chain chain is designated mAb 22.1.1 H-C 109 A. The invention further provides two variant heavy chains and a variant light chain of mAb 23.28.1. In one heavy chain variant, the aspartic acid at residue 16 is changed to glutamic acid. A mAb comprising the variant heavy chain variant and the 23.28.1 light chain is designated 23.28.1 H- D16E. The invention also includes a 23.28.1 light chain variant in which the cysteine at residue 92 is changed to an alanine. A mAb comprising the 23.28.1 heavy chain and the variant light chain is designated 23.28.1 L C92A. The invention also provides mAbs comprising either of the 23.28.1 heavy chain variants with the 23.28.1 light chain variant.
[0102] The light chain produced by hybridoma 23.29.1 contains a mutation in the constant region at residue 174. The light chain produced by the hybridoma has arginine at this position instead of the canonical lysine. Accordingly, the invention also provides a 23.29.1 light chain with the canonical lysine at residue 174 and a mAb, designated 23.29.1L-R174K, comprising the 23.29.1 heavy chain and the variant light chain. [0103] In a preferred embodiment, the anti-CD40 antibody is 3.1.1, 3.1.1H- A78T, 3.1.1H-A78T-V88A-V97A, 3.1.1L-L4M-L83V, 7.1.2, 10.8.3, 15.1.1,
21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K and 24.2.1. In some embodiments, the anti-CD40 antibody comprises a light chain comprising an amino acid sequence selected from SEQ ID NO: 8, 16, 24, 32, 40, 48, 56, 64, 72, 80, 88, 94, 100 or 102 or the variable region therefrom, or encoded by a nucleic acid sequence selected from SEQ ID NO: 7, 15, 23, 31, 39, 47, 55, 63, 71, 79, 87, 93, 99 or 101. In some embodiments, the anti-CD40 antibody comprises a light chain comprising at least the CDR2 from one of listed antibodies, one of the above-identified amino acid sequences (as shown in Figs. 1A-1C and 2A-2C) or encoded by one of the above- identified nucleic acid sequences. In another embodiment, the light chain further comprises a CDR1 and CDR3 independently selected from a light chain variable region that comprises no more than ten amino acids from the amino acid sequence encoded by a germline VK A3/A19, L5 or A27 gene, or comprises a CDRl and CDR3 independently selected from one of a CDRl and CDR3 of (1) an antibody selected from 3.1.1, 3.1.1L-L4M-L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K or 24.2.1 ; (2) the amino acid sequence of SEQ ED NO: 4, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 94, 100 or 102 or (3) encoded by the nucleic acid sequence of SEQ ID NO: 3, 11, 19, 27, 35, 43, 51, 59, 67, 75, 83, 93, 99 or 101. [0104] In another preferred embodiment, the anti-CD40 antibody comprises a heavy chain comprising an amino acid sequence selected from SEQ ID NOS: 6, 14, 22, 30, 38, 46, 54, 62, 70, 78 or 86 or the variable region therefrom or encoded by a nucleic acid sequence selected from SEQ ID NOS: 5, 13, 21, 29, 37, 45, 53, 61, 69, 77 or 85. In some embodiments, the anti-CD40 antibody comprises a heavy chain comprising at least the CDR3 from one of listed antibodies, one of the above-identified amino acid sequences (as shown in Figs. 1A-1C and 2A-2C) or encoded by one of the above-identified nucleic acid sequences. In another embodiment, the heavy chain further comprises a CDRl and CDR2 independently selected from a heavy chain variable region that comprises no more than eighteen amino acids from the amino acid sequence encoded by a germline VH 3-30+, 4-59, 1-02, 4.35 or 3-30.3 gene, or comprises a CDRl and CDR2 independently selected from one of a CDRl and CDR2 of ( 1 ) an antibody selected from 3.1.1, 3.1.1 H-
A78T, 3.1.1H-A78T-V88A-V97A, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.29.1 and 24.2.1; (2) the amino acid sequence of SEQ ID NO: 2, 10, 18, 26, 34, 42, 50, 58, 66, 74, 82, 90, 92, 96 or 98 or (3) encoded by the nucleic acid sequence of SEQ ID NO: 1, 9, 17, 25, 33, 41, 49, 57, 65, 73, 81, 89, 91, 95 or 97. In another embodiment, the anti-CD40 antibody comprises a heavy chain and a light chain as defined above. [0105] As used herein, antibody 3.1.1H-A78T is identical to that of 3.1.1 except that residue 78 of the heavy chain is threonine instead of alanine. Similarly, in antibody 3.1.1H-A78T-V88A-V97A, residue 78 is changed to A, and residues 88 and 97 are changed from valine to alanine in the heavy chain. Antibody 3.1.1L-
L4M-L83V is identical to that of 3.1.1 except that residue 4 is methionine instead of leucine and residue 83 is valine instead of leucine in the light chain. Antibody 22.1.1H-C109A is identical to that of 22.1.1 except that residue 109 of the heavy chain is changed from a cysteine to an alanine. Antibodies 23.28.1H-D16E and 23.28.1L-C92A are identical to that of 23.28.1 except that residue 16 of the heavy chain is changed from aspartate to glutamate, and residue 92 of the light chain is changed from cysteine to alanine, respectively. Antibody 23.29.1L-R174K is identical to that of 23.29.1 except that residue 174 of the light chain is changed from arginine to lysine.
Class and Subclass of Anti-CD4Q Antibodies
[0106] The class and subclass of anti-CD40 antibodies may be determined by any method known in the art. In general, the class and subclass of an antibody may be determined using antibodies that are specific for a particular class and subclass of antibody. Such antibodies are available commercially. The class and subclass can be determined by ELISA, or Western Blot as well as other techniques. Alternatively, the class and subclass may be determined by sequencing all or a portion of the constant domains of the heavy and/or light chains of the antibodies, comparing their amino acid sequences to the known amino acid sequences of various class and subclasses of immunoglobulins, and determining the class and subclass of the antibodies.
[0107] In some embodiments, the anti-CD40 antibody is a monoclonal antibody. The anti-CD40 antibody can be an IgG, an IgM, an IgE, an IgA or an IgD molecule. In a preferred embodiment, the anti-CD40 antibody is an IgG and is an IgGl, IgG2, IgG3 or IgG4 subclass. In another preferred embodiment, the anti- CD40 antibodies are subclass IgG2.
Species and Molecule Selectivity [0108] In another aspect of the invention, the anti-CD40 antibodies demonstrate both species and molecule selectivity. In some embodiments, the anti-CD40 antibody binds to primate and human CD40. In some embodiments, the anti-CD40 antibody binds to human, cynomolgus or rhesus CD40. In other embodiments, the anti-CD40 antibody does not bind to mouse, rat, dog or rabbit CD40. Following the teachings of the specification, one can determine the species selectivity for the anti-CD40 antibody using methods well known in the art. For instance, one can determine species selectivity using Western blot, FACS, ELISA or RIA. (See, e.g., Example IV.)
[0109] In some embodiments, the anti-CD40 antibody has a selectivity for CD40 that is more than 100 times greater than its selectivity for RANK (receptor activator of nuclear factor-kappa B), 4- IBB (CD 137), TNFR- 1 (Tumor Necrosis Factor Receptor- 1) and TNFR-2 (Tumor Necrosis Factor Receptor-2). In some embodiments, the anti-CD40 antibody does not exhibit any appreciable specific binding to any other protein other than CD40. One can determine the selectivity of the anti-CD40 antibody for CD40 using methods well known in the art following the teachings of the specification. For instance, one can determine the selectivity using Western blot, FACS, ELISA or RIA. (See, e.g., Example V.)
Identification of CD40 Epitopes Recognized by Anti-CD40 Antibody
[0110] Further, the invention provides a human anti-CD40 monoclonal antibody that binds CD40 and cross-competes with and/or binds the same epitope and or binds to CD40 with the same KD as a human anti-CD40 antibody selected from an antibody 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-V88A-V97A, 3.1.1L-L4M-L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K or 24.2.1; or a human anti-CD40 antibody that comprises a heavy chain variable region having an amino acid sequence of SEQ ID NO: 2, 10, 18, 26, 34, 42, 50, 58, 66, 74, 82, 90, 92, 96 or 98 or a human anti-CD40 antibody that comprises a light chain variable region having an amino acid sequence of SEQ ID NO: 4, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 94, 100 or 102. [0111] One can determine whether an antibody binds to the same epitope as or cross competes for binding with an anti-CD40 antibody by using any method known in the art. In one embodiment, one can allow the anti-CD40 antibody of the invention to bind to CD40 under saturating conditions and then measure the ability of the test antibody to bind to CD40. If the test antibody is able to bind to the CD40 at the same time as the anti-CD40 antibody, then the test antibody binds to a different epitope as the anti-CD40 antibody. However, if the test antibody is not able to bind to the CD40 at the same time, then the test antibody binds to the same epitope, an overlapping epitope, or an epitope that is in close proximity to the epitope bound by the human anti-CD40 antibody. This experiment can be performed using ELISA, RIA, FACS or surface plasmon resonance. (See, e.g., Example NI.) In a prefened embodiment, the experiment is performed using surface plasmon resonance. In a more preferred embodiment, BIAcore is used.
Binding Affinity of Anti-CD40 Antibodies to CD40
[0112] In some embodiments of the invention, the anti-CD40 antibody binds to CD40 with high affinity. In some embodiments, the anti-CD40 antibody binds to CD40 with a KD of 2 x 10" M or less. In another preferred embodiments, the antibody binds to CD40 with a KD of 2 x 10"9, 2 x 10"10, 4.0 x 10"11 M or less. In an even more prefened embodiment, the antibody binds to CD40 with a KD of 2.5 x 10"12 M or less. In some embodiments, the antibody binds to CD40 with substantially the same KD as an antibody selected from 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-N88A-N97A, 3.1.1L-L4M-L83N, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.28.1L-C92A, 23.29.1 , 23.29.1L-R174K or 24.2.1. In another preferred embodiment, the antibody binds to CD40 with substantially the same KD as an antibody that comprises a CDR2 of a light chain, and/or a CDR3 of a heavy chain from an antibody selected from 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-N88A-N97A, 3.1.1L-L4M-L83N, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, . 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.28.1L-C92A, 23.29.1, 23.29.1L- Rl 74K and 24.2.1. In still another preferred embodiment, the antibody binds to CD40 with substantially the same KD as an antibody that comprises a heavy chain variable region having an amino acid sequence of SEQ ID NO: 2, 10, 18, 26, 34, 42, 50, 58, 66, 74, 82, 90, 92, 96 or 98 or that comprises a light chain having an amino acid sequence of SEQ ID NO: 4, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 94, 100 or 102. In another preferred embodiment, the antibody binds to CD40 with substantially the same KD as an antibody that comprises a CDR2 of" a light chain variable region having an amino acid sequence of SEQ ID NO: 4, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 94, 100 or 102 or a CDR3 of a heavy chain variable region having an amino acid sequence of SEQ ID NO: 2, 10, 18, 26, 34, 42, 50, 58, 66, 74, 82, 90, 92, 96 or 98. [0113] In some embodiments, the anti-CD40 antibody has a low dissociation rate. In some embodiments, the anti-CD40 antibody has an Koff of 2.0 x 10"4 or lower. In some embodiments, the Koff is 2.0 x 10"7 or lower. In some embodiments, the Koff is substantially the same as an antibody described herein, including an antibody selected from 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-N88A- N97A, 3.1.1L-L4M-L83N, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H- C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K and 24.2.1. In some embodiments, the antibody binds to CD40 with substantially the same Koff as an antibody that comprises a CDR3 of a heavy chain or a CDR2 of a light chain from an antibody selected from 3.1.1, 3.1.1H- A78T, 3.1.1H-A78T-N88A-N97A, 3.1.1L-L4M-L83N, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K and 24.2.1. In some embodiments, the antibody binds to CD40 with substantially the same Koff as an antibody that comprises a heavy chain variable region having an amino acid sequence of SEQ ID NO: 2, 10, 18, 26, 34, 42, 50, 58, 66, 74, 82, 90, 92, 96 or 98 or that comprises a light chain variable region having an amino acid sequence of SEQ ID NO: 4, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 94, 100 or 102. In another preferred embodiment, the antibody binds to CD40 with substantially the same Koff as an antibody that comprises a CDR2 of a light chain variable region having an amino acid sequence of SEQ ID NO: 4, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 94, 100 or 102 or a CDR3 of a heavy chain variable region having an amino acid sequence of SEQ ID NO: 6, 14, 22, 30, 38, 46, 54, 62, 70, 78, 86, 90, 92, 96 or 98. [0114] The binding affinity and dissociation rate of an anti-CD40 antibody to CD40 can be determined by any method known in the art. The binding affinity can be measured by competitive ELIS As, RIAs or surface plasmon resonance, such as BIAcore. The dissociation rate also can be measured by surface plasmon resonance. Preferably, the binding affinity and dissociation rate is measured by surface plasmon resonance. More preferably, the binding affinity and dissociation rate are measured using a BIAcore™. See, e.g., Example XIV. Light and Heavy Chain Gene Usage
[0115] An anti-CD40 antibody of the invention can comprise a human kappa or a human lambda light chain or an amino acid sequence derived therefrom. In some embodiments comprising a kappa light chain, the light chain variable domain (VL) is encoded in part by a human A3/A19 (DPK- 15), L5 (DP5), or A27 (DPK-22) V/c gene.
[0116] In some embodiments, the NL of the anti-CD40 antibody contains one or more amino acid substitutions relative to the germline amino acid sequence. In some embodiments, the NL of the anti-CD40 antibody comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid substitutions relative to the germline amino acid sequence. In some embodiments, one or more of those substitutions from germline is in the CDR regions of the light chain. In some embodiments, the amino acid substitutions relative to germline are at one or more of the same positions as the substitutions relative to germline in any one or more of the NL of antibodies 3.1.1, 3.1.1L-L4M-L83N, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K and 24.2.1. For example, the NL of the anti-CD40 antibody may contain one or more amino acid substitutions compared to germline found in antibody 21.4.1, and other amino acid substitutions compared to germline found in antibody 10.8.3 which utilizes the same N gene as antibody 21.4.1. In some embodiments, the amino acid changes are at one or more . of the same positions but involve a different mutation than in the reference antibody.
[0117] In some embodiments, amino acid changes relative to germline occur at one or more of the same positions as in any of the NL of antibodies 3.1.1, 3.1.1 L- L4M-L83N, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K and 24.2.1, but the changes may represent conservative amino acid substitutions at such position(s) relative to the amino acid in the reference antibody. For example, if a particular position in one of these antibodies is changed relative to germline and is glutamate, one may conservatively substitute aspartate at that position. Similarly, if an amino acid substitution compared to germline is serine, one may conservatively substitute threonine for serine at that position. Conservative amino acid substitutions are discussed supra.
[0118] In some embodiments, the light chain of the human anti-CD40 antibody comprises the amino acid sequence that is the same as the amino acid sequence of the VL of antibody 3.1.1 (SEQ. LD NO: 4), 3.1.1L-L4M-L83V (SEQ ID NO: 94), 7.1.2 (SEQ. ID NO: 12), 10.8.3 (SEQ. ID NO: 20), 15.1.1 (SEQ. ID NO: 28), 21.4.1(SEQ. ID NO:), 21.2.1 (SEQ. ID NO: 36), 21.4.1 (SEQ ID NO: 44), 22.1.1 (SEQ. ID NO: 52), 23.5.1 (SEQ. ID NO: 60), 23.28.1 (SEQ. ID NO: 68), 23.28.1L-C92A (SEQ. ID NO: 100), 23.29.1 (SEQ. ID NO: 76), 23.29.1L-R174K (SEQ ID NO: 102) or 24.2.1 (SEQ. ]D NO: 84), or said amino acid sequence having up to 1, 2, 3, 4, 6, 8 or 10 conservative amino acid substitutions and/or a total of up to 3 non-conservative amino acid substitutions. [0119] h some embodiments, the light chain of the anti-CD40 antibody comprises at least the light chain CDR2, and may also comprise the CDRl and CDR3 regions of a germline sequence, as described herein. In another embodiment, the light chain may comprise a CDRl and CDR2 of an antibody independently selected from 3.1.1, 3.1.1L-L4M-L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.29.1 and 24.2.1, or CDR regions each having less than 8, less than 6, less than 4 or less than 3 conservative amino acid substitutions and/or a total of three or fewer non- conservative amino acid substitutions. In other embodiments, the light chain of the anti-CD40 antibody comprises at least the light chain CDR2, and may also comprise the CDRl and CDR3 regions, each of which are independently selected from the CDRl and CDR3 regions of an antibody having a light chain variable region comprising the amino acid sequence selected from SEQ ID NOS: 4, 12, 20,
28, 36, 44, 52, 60, 68, 76, 84, 94 or 100, or encoded by a nucleic acid molecule selected from SEQ ID NOS: 3, 11, 19, 27, 35, 43, 51, 59, 67, 75, 83, 93 or 99. [0120] With regard to the heavy chain, in some embodiments, the variable region of the heavy chain amino acid sequence is encoded in part by a human VH 3-30+, VH 4-59, VH 1-02, VH 4.35 or VH 3-30.3 gene. In some embodiments, the VH of the anti-CD40 antibody contains one or more amino acid substitutions, deletions or insertions (additions) relative to the germline amino acid sequence. In some embodiments, the variable domain of the heavy chain comprises 1, 2, 3, 4, 5, 6, 1, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17 or 18 mutations from the germline amino acid sequence. In some embodiments, the mutation(s) are non-conservative substitutions compared to the germline amino acid sequence. In some embodiments, the mutations are in the CDR regions of the heavy chain. In some embodiments, the amino acid changes are made at one or more of the same positions as the mutations from germline in any one or more of the VH of antibodies 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-V88A-V97A, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.29.1 and 24.2.1. In other embodiments, the amino acid changes are at one or more of the same positions but involve a different mutation than in the reference antibody.
[0121] In some embodiments, the heavy chain comprises an amino acid sequence of the variable domain (VH) of antibody 3.1.1 (SEQ JD NO: 2), 3.1.1H-A78T (SEQ ID NO: 90), 3.1.1H-A78T-V88A-V97A (SEQ ID NO: 92), 7.1.2 (SEQ ID NO: 10), 10.8.3 (SEQ ID NO: 18), 15.1.1 (SEQ ID NO: 26), 21.2.1 (SEQ ID NO: 34), 21.4.1 (SEQ ID NO: 42), 22.1.1 (SEQ ID NO: 50), 22.1.1H-C109A (SEQ ID NO: 96), 23.5.1 (SEQ ID NO: 58), 23.28.1 (SEQ ID NO: 66), 23.28.1H-D16E (SEQ ID NO: 98), 23.29.1 (SEQ ID NO: 74) and 24.2.1 (SEQ ID NO: 82), or said amino acid sequence having up to 1, 2, 3, 4, 6, 8 or 10 conservative amino acid substitutions and/or a total of up to 3 non-conservative amino acid substitutions. [0122] In some embodiments, the heavy chain comprises the heavy chain CDRl, CDR2 and CDR3 regions of antibody 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-N88A- N97A, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.29.1 and 24.2.1 (as shown in Figs. lD-lH or 2D-2H), or said CDR regions each having less than 8, less than 6, less than 4, or less than 3 conservative amino acid substitutions and/or a total of three or fewer non-conservative amino acid substitutions. [0123] In some embodiments, the heavy chain comprises a CDR3, and may also comprise the CDRl and CDR2 regions of a germline sequence, as described above, or may comprise a CDRl and CDR2 of an antibody, each of which are independently selected from an antibody comprising a heavy chain of an antibody selected from 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-N88A-N97A, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H- D16E, 23.29.1 and 24.2.1. In another embodiment, the heavy chain comprises a CDR3, and may also comprise the CDRl and CDR2 regions, each of which are
J5 independently selected from a CDRl and CDR2 region of a heavy chain variable region comprising an amino acid sequence selected from SEQ ID ΝOS: 2, 10, 18, 26, 34, 42, 50, 58, 66, 74, 82, 90, 92, 96 or 98 (as shown in Figs. 1D-1H or Figs. 2D-2H)or encoded by a nucleic acid sequence selected from SEQ ID ΝOS: 1, 9, 17, 25, 33, 41, 49, 57, 65, 73, 81, 89, 91, 95 or 97. In another embodiment, the 0 antibody comprises a heavy chain as disclosed above and a light chain as disclosed above.
[0124] One type of amino acid substitution that may be made is to change one or more cysteines in the antibody, which may be chemically reactive, to another residue, such as, without limitation, alanine or serine. In one embodiment, the 5 cysteine substitution is made in a framework region of a variable domain or in the constant domain of an antibody. In another embodiment, the cysteine is in a non- canonical region of the antibody. Another type of amino acid substitution that may be made is to change any potential proteolytic sites in the antibody, particularly those that are in a framework region of a variable domain, in the constant domain 0 of an antibody, or in a non-canonical region of the antibody Substitution of cysteine residues and removal of proteolytic sites may decrease the risk of any heterogeneity in the antibody product and thus increase its homogeneity. Another type of amino acid substitution is to eliminate asparagine-glycine pairs, which form potential deamidation sites, by altering one or both of the residues. This is 5 preferably done in framework regions, the constant domain or non-canonical regions of the antibody.
Activation of CD40 by Anti-CD40 Antibody
*
[0125] Another aspect of the present invention involves an anti-CD40 antibody that is an activating antibody, i.e., a CD40 agonist. An activating antibody 0 amplifies or substitutes for the effects of CD40L on CD40. In some embodiments, the activating antibody is essentially a mimic of CD40L, and competes with CD40L for binding to CD40. In some embodiments, the antibody does not compete with CD40L for binding to CD40, but amplifies the effect of CD40L binding to CD40. In some embodiments, the anti-CD40 antibody activates CD40 in the presence or absence of CD40L.
Inhibition of Tumor Growth In Vivo by Anti-CD40 Antibodies [0126] According to some embodiments, the invention provides an anti-CD40 antibody that inhibits the proliferation of tumor cells in vitro or tumor growth in vivo.
[0127] In some embodiments, the antibody inhibits tumor growth by at least 50%, 55%, 60%, 65%, 70%, 75%. In some embodiments, the antibody inhibits tumor growth by 75%. In one embodiment, the inhibition of tumor growth is detectable 14 days after initial treatment with the antibody. In other embodiments, the inhibition of tumor growth is detectable 7 days after initial treatment with the antibody. In some embodiments, another antineoplastic agent is administered to the animal with the anti-CD40 antibody. In some embodiments, the antineoplastic agent further inhibits tumor growth. In some embodiments, the antineoplastic agent is adriamycin or taxol. In some embodiments, the co-admimstration of an antineoplastic agent and the anti-CD40 antibody inhibits tumor growth by at least 50%, after a period of 22-24 days from initiation of treatment compared to tumor growth on an untreated animal.
Induction of Apoptosis by Anti-CD40 Antibodies
[0128] Another aspect of the invention provides an anti-CD40 antibody that induces cell death of CD40 positive cells. In some embodiments, the antibody causes apoptosis of CD40 positive cells either in vivo or in vitro.
Enhancement of Expression of Cell Surface Molecules [0129] In some embodiments, the anti-CD40 antibody enhances the expression of B cell surface molecules, including but not limited to ICAM, MHC-II, B7-2, CD71, CD23 and CD83. In some embodiments, 1 μg/ml of the antibody enhances ICAM expression in a whole blood B-cell surface molecule up-regulation assay by at least 2 fold, or more preferably by at least 4 fold. In some embodiments, 1 μg/ml of the antibody enhances MHC-II expression in a whole blood B-cell surface molecule upregulation assay by at least 2 fold, or more preferably by at least 3 fold. In some embodiments, 1 μg ml of the antibody enhances CD23 expression in whole blood B-cell surface molecule up-regulation assay by at least 2 fold, or more preferably by at least 5 fold. See, e.g., Example VII, Table 25. [0130] In some embodiments, the anti-CD40 antibody enhances the expression of dendritic cell surface molecules including but not limited to MHC-II, ICAM, B7-2, CD83 and B7-1. In some embodiments the range of upregulation is similar to the range of upregulation observed in B cells. See, e.g., Tables 25 and 26, infra. In some embodiments, the antibody preferentially upregulates the expression of dendritic cell surface molecules, such as B7-2 and MHC-II, compared to B cell expression of these molecules. See, e.g., Table 27.
Enhancement of Secretion of Cellular Cytokines
[0131] In some embodiments the antibody enhances cellular secretion of cytokines including but not limited to IL-8, IL-12, IL-15, IL-18 and IL-23. [0132] In some embodiments the antibody enhances cytokine secretion by dendritic cells and adherent monocytes. In some embodiments cytokine production is further enhanced by co-stimulation with one or more of LPS, IFN-γ or IL-ljS. In yet another aspect of the invention, the antibody with LPS co- stimulation enhances IL-12p70 production in a dendritic cell assay with an EC50 of about 0.48 μg/ml. In some embodiments, the antibody enhances IL-12p40 production in dendritic cells with an EC50 of about 0.21 μg/ml. (See, e.g., Example
VIII.)
[0133] In some embodiments, the antibody enhances secretion of IFN-gamma by
T cells in an allogenic T cell/dendritic cell assay, as described in Example VIII. In some embodiments, the antibody enhances IFN-gamma secretion in an allogenic T cell dendritic cell assay with an EC50 of about 0.3 μg/ml. In some embodiments, the antibody enhances IFN-gamma secretion in an allogenic T cel .dendritic cell assay with an EC50 of about 0.2 μg/ml. In one embodiment, the antibody enhances IFN-gamma secretion in an allogenic T cell/dendritic cell assay with an EC50 of about 0.03 μg/ml. Methods of Producing Antibodies and Antibody-Producing Cell Lines
Immunization
[0134] In some embodiments, human antibodies are produced by immunizing a non-human animal comprising in its genome some or all of human immunoglobulin heavy chain and light chain loci with a CD40 antigen. In a preferred embodiment, the non-human animal is a XenoMouse™ animal. [0135] XenoMouse™ mice are engineered mouse strains that comprise large fragments of human immunoglobulin heavy chain and light chain loci and are deficient in mouse antibody production. See, e.g., Green et al., Nature Genetics 7:13-21 (1994) and U.S. Patents 5,916,771, 5,939,598, 5,985,615, 5,998,209,
6,075,181, 6,091,001, 6,114,598, 6,130,364, 6,162,963 and 6,150,584. See also WO 91/10741, WO 94/02602, WO 96/34096, WO 96/33735, WO 98/16654, WO 98/24893, WO 98/50433, WO 99/45031, WO 99/53049, WO 00/09560, and WO 00/037504. [0136] In another aspect, the invention provides a method for making anti-CD40 antibodies from non-human, non-mouse animals by immunizing non-human transgenic animals that comprise human immunoglobulin loci with a CD40 antigen. One can produce such animals using the methods described in the above- cited documents. The methods disclosed in these documents can be modified as described in U.S. Patent 5,994,619. In preferred embodiments, the non-human animals are rats, sheep, pigs, goats, cattle or horses.
[0137] XenoMouse™ mice produce an adult-like human repertoire of fully human antibodies and generate antigen-specific human antibodies. In some embodiments, the XenoMouse™ mice contain approximately 80% of the human antibody V gene repertoire through introduction of megabase sized, germline configuration yeast artificial chromosome (YAC) fragments of the human heavy chain loci and kappa light chain loci. See Mendez et al., Nature Genetics 15:146- 156 (1997), Green and Jakobovits, J. Exp. Med. 188:483-495 (1998), and WO 98/24893, the disclosures of which are hereby incorporated by reference. [0138] In some embodiments, the non-human animal comprising human immunoglobulin genes are animals that have a human immunoglobulin "minilocus". In the minilocus approach, an exogenous Ig locus is mimicked through the inclusion of individual genes from the Ig locus. Thus, one or more NH genes, one or more DH genes, one or more JH genes, a mu constant domain, and a second constant domain (preferably a gamma constant domain) are formed into a construct for insertion into an animal. This approach is described, inter alia, in U.S. Patent os. 5,545,807, 5,545,806, 5,569,825, 5,625,126, 5,633,425,
5,661,016, 5,770,429, 5,789,650, 5,814,318, 5,591,669, 5,612,205, 5,721,367, 5,789,215, and 5,643,763, hereby incorporated by reference. [0139] An advantage of the minilocus approach is the rapidity with which constructs including portions of the Ig locus can be generated and introduced into animals. However, a potential disadvantage of the minilocus approach is that there may not be sufficient immunoglobulin diversity to support full B-cell development, such that there may be lower antibody production.
[0140] In another aspect, the invention provides a method for making humanized anti-CD40 antibodies. In some embodiments, non-human animals are immunized with a CD40 antigen as described below under conditions that permit antibody production. Antibody-producing cells are isolated from the animals, fused with myelomas to produce hybridomas, and nucleic acids encoding the heavy and light chains of an anti-CD40 antibody of interest are isolated. These nucleic acids are subsequently engineered using techniques known to those of skill in the art and as described further below to reduce the amount of non-human sequence, i.e., to humanize the antibody to reduce the immune response in humans [0141] In some embodiments, the CD40 antigen is isolated and/or purified CD40. In a prefened embodiment, the CD40 antigen is human CD40. In some embodiments, the CD40 antigen is a fragment of CD40. In some embodiments, the CD40 fragment is the extracellular domain of CD40. In some embodiments, the CD40 fragment comprises at least one epitope of CD40. In other embodiments, the CD40 antigen is a cell that expresses or overexpresses CD40 or an immunogenic fragment thereof on its surface. In some embodiments, the CD40 antigen is a CD40 fusion protein. [0142] Immunization of animals can be by any method known in the art. See, e.g., Harlow and Lane, Antibodies: A Laboratory Manual New York: Cold Spring Harbor Press, 1990. Methods for immunizing non-human animals such as mice, rats, sheep, goats, pigs, cattle and horses are well known in the art. See, e.g., Harlow and Lane, supra, and U.S. Patent 5,994,619. In a prefened embodiment, the CD40 antigen is administered with an adjuvant to stimulate the immune response. Exemplary adjuvants include complete or incomplete Freund's adjuvant, RIBI (muramyl dipeptides) or ISCOM (immunostimulating complexes). Such adjuvants may protect the polypeptide from rapid dispersal by sequestering it in a local deposit, or they may contain substances that stimulate the host to secrete factors that are chemotactic for macrophages and other components of the immune system. Preferably, if a polypeptide is being administered, the immunization schedule will involve two or more administrations of the polypeptide, spread out over several weeks. [0143] Example I describes the production of anti-CD40 monoclonal antibodies.
Production of Antibodies and Antibody-Producing Cell Lines
[0144] After immunization of an animal with a CD40 antigen, antibodies and/or antibody-producing cells can be obtained from the animal. In some embodiments, anti-CD40 antibody-containing serum is obtained from the animal by bleeding or sacrificing the animal. The serum may be used as it is obtained from the animal, an immunoglobulin fraction may be obtained from the serum, or the anti-CD40 antibodies may be purified from the serum. It is well known to one of ordinary skill in the art that serum or immunoglobulins obtained in this manner will be polyclonal The disadvantage is using polyclonal antibodies prepared from serum is that the amount of antibodies that can be obtained is limited and the polyclonal antibody has a heterogeneous anay of properties. [0145] In some embodiments, antibody-producing immortalized cell lines are prepared from cells isolated from the immunized animal. After immunization, the animal is sacrificed and lymph node and/or splenic B cells are immortalized. Methods of immortalizing cells include, but are not limited to, transferring them with oncogenes, inflecting them with the oncogenic virus cultivating them under conditions that select for immortalized cells, subjecting them to carcinogenic or mutating compounds, fusing them with an immortalized cell, e.g., a myeloma cell, and inactivating a tumor suppressor gene. See, e.g., Harlow and Lane, supra. If fusion with myeloma cells is used, the myeloma cells preferably do not secrete immunoglobulin polypeptides (a non-secretory cell line). Immortalized cells are screened using CD40, a portion thereof, or a cell expressing CD40. In a prefened embodiment, the initial screening is performed using an enzyme-linked immunoassay (ELISA) or a radioimmunoassay. An example of ELISA screening is provided in WO 00/37504, herein incorporated by reference.
[0146] Anti-CD40 antibody-producing cells, e.g., hybridomas, are selected, cloned and further screened for desirable characteristics, including robust growth, high antibody production and desirable antibody characteristics, as discussed further below. Hybridomas can be expanded in vivo in syngeneic animals, in animals that lack an immune system, e.g., nude mice, or in cell culture in vitro.
Methods of selecting, cloning and expanding hybridomas are well known to those of ordinary skill in the art.
[0147] In a prefened embodiment, the immunized animal is a non-human animal that expresses human immunoglobulin genes and the splenic B cells are fused to a myeloma cell line from the same species as the non-human animal. In a more prefened embodiment, the immunized animal is a XENOMOUSE™ animal and the myeloma cell line is a non-secretory mouse myeloma. In an even more prefened embodiment, the myeloma cell line is P3-X63-AG8.653. See, e.g., Example I. [0148] In another aspect, the invention provides hybridomas that produce an human anti-CD40 antibody. In a prefened embodiment, the hybridomas are mouse hybridomas, as described above. In other embodiments, the hybridomas are produced in a non-human, non-mouse species such as rats, sheep, pigs, goats, cattle or horses. In another embodiment, the hybridomas are human hybridomas.
Nucleic Acids. Vectors, Host Cells and Recombinant Methods of Making Antibodies
Nucleic Acids
[0149] The present invention also encompasses nucleic acid molecules encoding anti-CD40 antibodies. In some embodiments, different nucleic acid molecules encode a heavy chain and a light chain of an anti-CD40 immunoglobulin. In other embodiments, the same nucleic acid molecule encodes a heavy chain and a light chain of an anti-CD40 immunoglobulin. [0150] In some embodiments, the nucleic acid molecule encoding the variable domain of the light chain comprises a human A3/A19 (DPK-15), L5 (DP5) or A27 (DPK-22) V/c gene sequence or a sequence derived therefrom. In some embodiments, the nucleic acid molecule comprises a nucleotide sequence of a A3/A19 V/c gene and a J/cl, Jc2 or Jc3 gene or sequences derived therefrom. In some embodiments, the nucleic acid molecule comprises a nucleotide sequence of an L5 V c gene and a J/c4 gene. In some embodiments, the nucleic acid molecule comprises a nucleotide sequence of a A27 V/c gene and a Jκ3 gene. [0151] In some embodiments, the nucleic acid molecule encoding the light chain, encodes an amino acid sequence comprising 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 mutations from the germline amino acid sequence. In some embodiments, the nucleic acid molecule comprises a nucleotide sequence that encodes a VL amino acid sequence comprising 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 non-conservative amino acid substitutions and/or 1, 2 or 3 non-conservative substitutions compared to the germline sequence. Substitutions may be in the CDR regions, the framework regions or in the constant domain.
[0152] In some embodiments, the nucleic acid molecule encoding the variable domain of the light chain (VL) encodes a VL amino acid sequence comprising one or more mutations compared to the germline sequence that are identical to the mutations found in the VL of one of the antibodies 3.1.1, 3.1.1L-L4M-L83V, 7.1.2,
10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.29.1 and 24.2.1. In some embodiments, the nucleic acid molecule encodes at least three amino acid mutations compared to the germline sequence found in the V of one of the antibodies 3.1.1, 3.1.1L-L4M-L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.29.1 and 24.2.1.
[0153] In some embodiments, the nucleic acid molecule comprises a nucleotide sequence that encodes the VL amino acid sequence of monoclonal antibody 3.1.1 (SEQ ID NO: 4), 3.1.1L-L4M-L83V (SEQ ID NO: 94), 7.1.2 (SEQ'ID NO: 12), 10.8.3 (SEQ ID NO: 20), 15.1.1 (SEQ ID NO: 28), 21.2.1 (SEQ ID NO: 36), 2.1.4.1 (SEQ ID NO: 44), 22.1.1 (SEQ ID NO: 52), 23.5.1 (SEQ ID NO: 60),
23.28.1 (SEQ ID NO: 68), 23.28.1L-C92A (SEQ ID NO: 100), 23.29.1 (SEQ ID NO: 76) or 24.2.1 (SEQ ID NO: 84), or a portion thereof. In some embodiments, said portion comprises at least the CDR3 region. In some embodiments, the nucleic acid encodes the amino acid sequence of the light chain CDRs of said antibody. In some embodiments, said portion is a contiguous portion comprising CDR1-CDR3. [0154] In some embodiments, the nucleic acid molecule comprises a nucleotide sequence that encodes the amino acid sequence of one of SEQ ID NOS: 4, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 94 or 100, or said sequence lacking the signal sequence. In some prefened embodiments, the nucleic acid molecule comprises the nucleotide sequence of SEQ ID NOS: 3, 11, 19, 27, 35, 43, 51, 59, 67, 75, 83, 93 or 99, or a portion thereof, said sequences optionally lacking the signal sequence.
[0155] In some embodiments, said portion encodes a VL region. In some embodiments, said portion encodes at least the CDR2 region. In some embodiments, the nucleic acid encodes the amino acid sequence of the light chain CDRs of said antibody. In some embodiments, said portion encodes a contiguous region from CDR1-CDR3.
[0156] In some embodiments, the nucleic acid molecule encodes a VL amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98% or 99% identical to a VL amino acid sequence of any one of antibodies 3.1.1, 3.1.1L-L4M- L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1,
23.28.1L-C92A, 23.29.1 or 24.2.1, or a VL amino acid sequence of any one of SEQ ID NOS: 4, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 94or 100. Nucleic acid molecules of the invention include nucleic acids that hybridize under highly stringent conditions, such as those described above, to a nucleic acid sequence encoding the amino acid sequence of SEQ ID NOS: 4, 12, 20, 28, 36, 44, 52, 60,
68, 76, 84, 94 or 100, or that has the nucleic acid sequence of SEQ ID NOS: 3, 11, 19, 27, 35, 43, 51, 59, 67, 75, 83, 93 or 99.
[0157] In another embodiment, the nucleic acid encodes a full-length light chain of an antibody selected from 3.1.1, 3.1.1L-L4M-L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.29.1, 23.29.1L-
R174K or 24.2.1, or a light chain comprising the amino acid sequence of SEQ ID NOS: 8, 16, 24, 32, 40, 48, 56, 64, 72, 80, 88, 94, 100 or 102, or a light chain comprising a mutation, such as one disclosed herein. Further, the nucleic acid may comprise the nucleotide sequence of SEQ ID NOS: 7, 15, 23, 31, 39, 47, 55, 63, 71, 79 or 87, or a nucleic acid molecule encoding a light chain comprise a mutation, such as one disclosed herein. [0158] In another prefened embodiment, the nucleic acid molecule encodes the variable domain of the heavy chain (VH) that comprises a human 3-30+, 4-59, 1- 02, 4.35 or 3-30.3 VH gene sequence or a sequence derived therefrom. In various embodiments, the nucleic acid molecule comprises a human 3-30+ VH gene, a D4 (DIR3) gene and a human JH6 gene; a human 3-30+ VH gene, a human Dl-26 (DIR5) gene and a human JH6 gene; a human 4.35 VH gene, a human DIR3 gene and a human H6 gene; a human 4-59 VH gene, a human D4-23 gene and a human JH4 gene; a human 1-02 VH gene, a human DLR1 gene and a human JH4 gene; a human 3-30+ VH gene, a human D6-19 (DIR3) gene and a human JH4 gene; a human 3-30+ VH gene, a human Dl-1 gene and a human JH6 gene; a human 3-30+ VH gene, a human D4-17 gene and a human JH6 gene; a human 3-30.3 VH gene, a human D4-17 gene and a human JH6 gene; a human 4-59 VH gene, a human D4-17 (DJR1) gene and a human JH5 gene, or sequence derived from the human genes. [0159] In some embodiments, the nucleic acid molecule encodes an amino acid sequence comprising 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17 or 18 mutations compared to the germline amino acid sequence of the human V, D or J genes. In some embodiments, said mutations are in the VH region. In some embodiments, said mutations are in the CDR regions.
[0160] In some embodiments, the nucleic acid molecule encodes one or more amino acid mutations compared to the germline sequence that are identical to amino acid mutations found in the VH of monoclonal antibody 3.1.1, 3.1.1 H-A78T,
3.1.1H-A78T-V88A-V97A, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H- C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.29.1 or 24.2.1. In some embodiments, the nucleic acid encodes at least three amino acid mutations compared to the germline sequences that are identical to at least three amino acid mutations found in one of the above-listed monoclonal antibodies.
[0161] In some embodiments, the nucleic acid molecule comprises a nucleotide sequence that encodes at least a portion of the VH amino acid sequence of antibody 3.1.1 (SEQ ID NO: 2), 3.1.1H-A78T (SEQ ID NO: 90), 3.1.1H-A78T-V88A- V97A (SEQ ID NO: 92), 7.1.2 (SEQ ID NO: 10), 10.8.3 (SEQ ID NO: 18), 15.1.1 (SEQ ID NO: 26), 21.2.1 (SEQ ID NO: 34), 21.4.1 (SEQ ID NO: 42), 22.1.1 (SEQ ID NO: 50), 22.1.1H-C109A (SEQ ID NO: 96), 23.5.1 (SEQ ID NO: 58), 23.28.1 (SEQ ID NO: 66), 23.28.1H-D16E (SEQ ID NO: 98), 23.29.1 (SEQ ID NO: 74) or 24.2.1 (SEQ ID NO: 82), or said sequence having conservative amino acid mutations and/or a total of three or fewer non-conservative amino acid substitutions. In various embodiments the sequence encodes one or more CDR regions, preferably a CDR3 region, all three CDR regions, a contiguous portion including CDR1-CDR3, or the entire VH region, with or without a signal sequence. [0162] In some embodiments, the nucleic acid molecule comprises a nucleotide sequence that encodes the amino acid sequence of one of SEQ ID NOS: 2, 10, 18, 26, 34, 42, 50, 58, 66, 74, 82, 90, 92, 96 or 98, or said sequence lacking the signal sequence. In some prefened embodiments, the nucleic acid molecule comprises at least aportion of the nucleotide sequence of SEQ ID NO: 1, 9, 17, 25, 33, 41, 49, 57, 65, 73, 81, 89, 91, 95 or 97, or said sequence lacking the signal sequence. In some embodiments, said portion encodes the VH region (with or without a signal sequence), a CDR3 region, all three CDR regions, or a contiguous region including CDR1-CDR3. [0163] In some embodiments, the nucleic acid molecule encodes a VH amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98% or 99% identical to the VH amino acid sequences shown in FIGS. 1A-1C or 2A-2C or to a VH amino acid sequence of any one of SEQ ID NOS: 2, 10, 18, 26, 34, 42, 50, 58, 66, 74, 82, 90, 92, 96 or 98. Nucleic acid molecules of the invention include nucleic acids that hybridize under highly stringent conditions, such as those described above, to a nucleic acid sequence encoding the amino acid sequence of SEQ ID NOS: 2, 10, 18, 26, 34, 42, 50, 58, 66, 74, 82, 90, 92, 96 or 98, or that has the nucleic acid sequence of SEQ ID NOS: 1, 9, 17, 25, 33, 41, 49, 57, 65, 73, 81, 89, 91, 95 or 97. Nucleic acid molecule of the invention include nucleic acid molecule that hybridize under highly stringent conditions, such as those described above, to a nucleic acid sequence encoding a VH described immediately above. [0164] In another embodiment, the nucleic acid encodes a full-length heavy chain of an antibody selected from 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-V88A- V97A, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.29.1 and 24.2.1, or a heavy chain having the amino acid sequence of SEQ ID NOS: 6, 14, 22, 30, 38, 46, 54, 62, 70, 78 or 86, or a heavy chain comprising a mutation, such as one of the mutations discussed herein Further, the nucleic acid may comprise the nucleotide sequence of SEQ ID NOS: 5, 13, 21, 29, 37, 45, 53, 61, 69, 77, 85 or 89, or a nucleic acid molecule encoding a heavy chain comprising a mutation, such as one of the mutations discussed herein.
[0165] A nucleic acid molecule encoding the heavy or entire light chain of an anti-CD40 antibody or portions thereof can be isolated from any source that produces such antibody. In various embodiments, the nucleic acid molecules are isolated from a B cell isolated from an animal immunized with CD40 or from an immortalized cell derived from such a B cell that expresses an anti-CD40 antibody. Methods of isolating mRNA encoding an antibody are well-known in the art. See, e.g., Sambrook et al. The mRNA may be used to produce cDNA for use in the polymerase chain reaction (PCR) or cDNA cloning of antibody genes. In a prefened embodiment, the nucleic acid molecule is isolated from a hybridoma that has as one of its fusion partners a human immunoglobulin-producing cell from a non-human transgenic animal. In an even more prefened embodiment, the human immunoglobulin producing cell is isolated from a XenoMouse animal. In another embodiment, the human immunoglobulin-producing cell is from a non- human, non-mouse transgenic animal, as described above. In another embodiment, the nucleic acid is isolated from a non-human, non-transgenic animal The nucleic acid molecules isolated from a non-human, non-transgenic animal may be used, e.g., for humanized antibodies.
[0166] In some embodiments, a nucleic acid encoding a heavy chain of an anti- CD40 antibody of the invention can comprise a nucleotide sequence encoding a VH domain of the invention joined in-frame to a nucleotide sequence encoding a heavy chain constant domain from any source. Similarly, a nucleic acid molecule encoding a light chain of an anti-CD40 antibody of the invention can comprise a nucleotide sequence encoding a VL domain of the invention joined in-frame to a nucleotide sequence encoding a light chain constant domain from any source. [0167] In a further aspect of the invention, nucleic acid molecules encoding the variable domain of the heavy (VH) and light (VL) chains are "converted" to full- length antibody genes. In one embodiment, nucleic acid molecules encoding the VH or VL domains are converted to full-length antibody genes by insertion into an expression vector already encoding heavy chain constant or light chain constant domains, respectively, such that the VH segment is operatively linked to the CH segment(s) within the vector, and the VL segment is operatively linked to the CL segment within the vector. In another embodiment, nucleic acid molecules encoding the VH and/or VL domains are converted into full-length antibody genes by linking, e.g., ligating, a nucleic acid molecule encoding a VH and/or VL domains to a nucleic acid molecule encoding a CH and/or CL domain using standard molecular biological techniques. Nucleic acid sequences of human heavy and light chain immunoglobulin constant domain genes are known in the art. See, e.g.,
Kabat et al, Sequences of Proteins of Immunological Interest. 5th Ed., NJJff Publ No. 91-3242, 1991. Nucleic acid molecules encoding the full-length heavy and/or light chains may then be expressed from a cell into which they have been introduced and the anti-CD40 antibody isolated. [0168] The nucleic acid molecules may be used to recombinantly express large quantities of anti-CD40 antibodies. The nucleic acid molecules also may be used to produce chimeric antibodies, bispecific antibodies, single chain antibodies, immunoadhesins, diabodies, mutated antibodies and antibody derivatives, as described further below. If the nucleic acid molecules are derived from a non- human, non-transgenic animal, the nucleic acid molecules may be used for antibody humanization, also as described below.
[0169] In another embodiment, a nucleic acid molecule of the invention is used as a probe or PCR primer for a specific antibody sequence. For instance, the nucleic acid can be used as a probe in diagnostic methods or as a PCR primer to amplify regions of DNA that could be used, inter alia, to isolate additional nucleic acid molecules encoding variable domains of anti-CD40 antibodies. In some embodiments, the nucleic acid molecules are oligonucleotides. In some embodiments, the oligonucleotides are from highly variable regions of the heavy and light chains of the antibody of interest, hi some embodiments, the oligonucleotides encode all or a part of one or more of the CDRs of antibody 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-V88A-V97A, 3.1.1L-L4M-L83N, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H- D16E, 23.28.1L-C92A, 23.29.1 or 24.2.1.
Vectors
[0170] The invention provides vectors comprising nucleic acid molecules that encode the heavy chain of an anti-CD40 antibody of the invention or an antigen- binding portion thereof. The invention also provides vectors comprising nucleic acid molecules that encode the light chain of such antibodies or antigen-binding portion thereof. The invention further provides vectors comprising nucleic acid molecules encoding fusion proteins, modified antibodies, antibody fragments, and probes thereof. [0171] In some embodiments, the anti-CD40 antibodies, or antigen-binding portions of the invention are expressed by inserting DΝAs encoding partial or full-length light and heavy chains, obtained as described above, into expression vectors such that the genes are operatively linked to necessary expression control sequences such as transcriptional and translational control sequences. Expression vectors include plasmids, retroviruses, adenovirases, adeno-associated viruses (AAN), plant viruses such as cauliflower mosaic virus, tobacco mosaic virus, cosmids, YACs, EBN derived episomes, and the like. The antibody gene is ligated into a vector such that transcriptional and translational control sequences within the vector serve their intended function of regulating the transcription and translation of the antibody gene. The expression vector and expression control sequences are chosen to be compatible with the expression host cell used. The antibody light chain gene and the antibody heavy chain gene can be inserted into separate vectors. In a prefened embodiment, both genes are inserted into the same expression vector. The antibody genes are inserted into the expression vector by standard methods (e.g., ligation of complementary restriction sites on the antibody gene fragment and vector, or blunt end ligation if no restriction sites are present). [0172] A convenient vector is one that encodes a functionally complete human CH or CL immunoglobulin sequence, with appropriate restriction sites engineered so that any Nπ or NL sequence can easily be inserted and expressed, as described above. In such vectors, splicing usually occurs between the splice donor site in the inserted J region and the splice acceptor site preceding the human C domain, and also at the splice regions that occur within the human CH exons. Polyadenylation and transcription termination occur at native chromosomal sites downstream of the coding regions. The recombinant expression vector also can encode a signal peptide that facilitates secretion of the antibody chain from a host cell. The antibody chain gene may be cloned into the vector such that the signal peptide is linked in-frame to the amino terminus of the immunoglobulin chain. The signal peptide can be an immunoglobulin signal peptide or a heterologous signal peptide (i.e., a signal peptide from a non-immunoglobulin protein). [0173] In addition to the antibody chain genes, the recombinant expression vectors of the invention cany regulatory sequences that control the expression of the antibody chain genes in a host cell. It will be appreciated by those skilled in the art that the design of the expression vector, including the selection of regulatory sequences may depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc. Prefened regulatory sequences for mammalian host cell expression include viral elements that direct high levels of protein expression in mammalian cells, such as promoters and/or enhancers derived from retroviral LTRs, cytomegalovirus (CMN) (such as the CMV promoter/enhancer), Simian Virus 40 (SV40) (such as the SV40 promoter/enhancer), adenovirus, (e.g., the adenovirus major late promoter (AdMLP)), polyoma and strong mammalian promoters such as native immunoglobulin and actin promoters. For further description of viral regulatory elements, and sequences thereof, see e.g., U.S. Patent No. 5,168,062, U.S. Patent No. 4,510,245 and U.S. Patent No. 4,968,615. Methods for expressing antibodies in plants, including a description of promoters and vectors, as well as transformation of plants is known in the art. See, e.g., United States Patents
6,517,529, herein incorporated by reference. Methods of expressing polypeptides in bacterial cells or fungal cells, e.g., yeast cells, are also well known in the art. [0174] In addition to the antibody chain genes and regulatory sequences, the recombinant expression vectors of the invention may cany additional sequences, such as sequences that regulate replication of the vector in host cells (e.g., origins of replication) and selectable marker genes. The selectable marker gene facilitates selection of host cells into which the vector has been introduced (see e.g., U.S. Patent Nos. 4,399,216, 4,634,665 and 5,179,017). For example, typically the selectable marker gene confers resistance to drags, such as G418, hygromycin or methofrexate, on a host cell into which the vector has been introduced. Prefened selectable marker genes include the dihydrofolate reductase (DHFR) gene (for use in dhfr-host cells with methofrexate selection/amplification), the neo gene (for G418 selection), and the glutamate synthetase gene.
Non-Hybridoma Host Cells and Methods of Recombinantly Producing Protein [0175] Nucleic acid molecules encoding anti-CD40 antibodies and vectors comprising these nucleic acid molecules can be used for transfection of a suitable mammalian, plant, bacterial or yeast host cell. Transformation can be by any known method for introducing polynucleotides into a host cell. Methods for introduction of heterologous polynucleotides into mammalian cells are well known in the art and include dextran-mediated transfection, calcium phosphate precipitation, polybrene-mediated transfection, protoplast fusion, electroporation, encapsulation of the polynucleotide(s) in liposomes, and direct microinjection of the DNA into nuclei. In addition, nucleic acid molecules may be introduced into mammalian cells by viral vectors. Methods of transforming cells are well known in the art. See, e.g., U.S. Patent Nos. 4,399,216, 4,912,040, 4,740,461, and 4,959,455 (which patents are hereby incorporated herein by reference). Methods of transforming plant cells are well known in the art, including, e.g.,
Agrobacterium-mediated transformation, biolistic transformation, direct injection, electroporation and viral transformation. Methods of transforming b # acterial and yeast cells are also well known in the art.
[0176] Mammalian cell lines available as hosts for expression are well known in the art and include many immortalized cell lines available from the American Type
Culture Collection (ATCC). These include, inter alia, Chinese hamster ovary (CHO) cells, NSO, SP2 cells, HeLa cells, baby hamster kidney (BHK) cells, monkey kidney cells (COS), human hepatocellular carcinoma cells (e.g., Hep G2), A549 cells, and a number of other cell lines. Cell lines of particular preference are selected through determining which cell lines have high expression levels. Other cell lines that may be used are insect cell lines, such as Sf9 cells. When recombinant expression vectors encoding antibody genes are introduced into mammalian host cells, the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or, more preferably, secretion of the antibody into the culture medium in which the host cells are grown. Antibodies can be recovered from the culture medium using standard protein purification methods. Plant host cells include, e.g., Nicotiana, Arabidopsis, duckweed, corn, wheat, potato, etc. Bacterial host cells include E. coli and Streptomyces species. Yeast host cells include Schizosaccharomyces pombe, Saccharomyces cerevisiae and Pichia pastoris. [0177] Further, expression of antibodies of the invention (or other moieties therefrom) from production cell lines can be enhanced using a number of known techniques. For example, the glutamine synthetase gene expression system (the GS system) is a common approach for enhancing expression under certain conditions. The GS system is discussed in whole or part in connection with European Patent Nos. 0 216 846, 0 256 055, and 0 323 997 and European Patent Application No. 89303964.4.
[0178] It is likely that antibodies expressed by different cell lines or in transgenic animals will have different glycosylation from each other. However, all antibodies encoded by the nucleic acid molecules provided herein, or comprising the amino acid sequences provided herein are part of the instant invention, regardless of the glycosylation of the antibodies.
Transgenic Animals and Plants
[0179] Anti-CD40 antibodies of the invention also can be produced transgenically through the generation of a mammal or plant that is transgenic for the immunoglobulin heavy and light chain sequences of interest and production of the antibody in a recoverable form therefrom. In connection with the transgenic production in mammals, anti-CD40 antibodies can be produced in, and recovered from, the milk of goats, cows, or other mammals. See, e.g., U.S. Patent Nos. 5,827,690, 5,756,687, 5,750,172, and 5,741,957. In some embodiments, non- human transgenic animals that comprise human immunoglobulin loci are immunized with CD40 or an immunogenic portion thereof, as described above. Methods for making antibodies in plants are described, e.g., in US patents 6,046,037 and US 5,959,177.
[0180] In some embodiments, non-human transgenic animals or plants are produced by introducing one or more nucleic acid molecules encoding an anti- CD40 antibody of the invention into the animal or plant by standard transgenic techniques. See Hogan and United States Patent 6,417,429, supra. The transgenic cells used for making the transgenic animal can be embryonic stem cells or somatic cells. The transgenic non-human organisms can be chimeric, nonchimeric heterozygotes, and nonchimeric homozygotes. See, e.g., Hogan et al, Manipulating the Mouse Embryo: A Laboratory Manual 2ed., Cold Spring Harbor Press (1999); Jackson et al, Mouse Genetics and Transgenics: A Practical Approach, Oxford University Press (2000); and Pinkert, Transgenic Animal Technology: A Laboratory Handbook, Academic Press (1999). In some embodiments, the transgenic non-human animals have a targeted disruption and replacement by a targeting construct that encodes a heavy chain and/or a light chain of interest. In a prefened embodiment, the transgenic animals comprise and express nucleic acid molecules encoding heavy and light chains that specifically bind to CD40, preferably human CD40. In some embodiments, the transgenic animals comprise nucleic acid molecules encoding a modified antibody such as a single-chain antibody, a chimeric antibody or a humanized antibody. The anti- CD40 antibodies may be made in any transgenic animal. In a prefened embodiment, the non-human animals are mice, rats, sheep, pigs, goats, cattle or horses. The non-human transgenic animal expresses said encoded polypeptides in blood, milk, urine, saliva, tears, mucus and other bodily fluids.
Phage Display Libraries
[0181] The invention provides a method for producing an anti-CD40 antibody or antigen-binding portion thereof comprising the steps of synthesizing a library of human antibodies on phage, screening the library with CD40 or a portion thereof, isolating phage that bind CD40, and obtaining the antibody from the phage. By way of example, one method for preparing the library of antibodies for use in phage display techniques comprises the steps of immunizing a non-human animal comprising human immunoglobulin loci with CD40 or an antigenic portion thereof to create an immune response, extracting antibody producing cells from the immunized animal; isolating RNA from the extracted cells, reverse transcribing the RNA to produce cDNA, amplifying the cDNA using a primer, and inserting the cDNA into a phage display vector such that antibodies are expressed on the phage. Recombinant anti-CD40 antibodies of the invention may be obtained in this way. [0182] Recombinant anti-CD40 human antibodies of the invention can be isolated by screening a recombinant combinatorial antibody library. Preferably the library is a scFv phage display library, generated using human NL and Nπ cD As prepared from mRΝA isolated from B cells. Methodologies for preparing and screening such libraries are known in the art. There are commercially available kits for generating phage display libraries (e.g., the Pharmacia Recombinant Phage Antibody System, catalog no. 27-9400-01; and the Stratagene SurfZAP™ phage display kit, catalog no. 240612). There also are other methods and reagents that can be used in generating and screening antibody display libraries (see, e.g., U.S. Patent o. 5,223,409; PCT Publication Nos. WO 92/18619, WO 91/17271, WO 92/20791, WO 92/15679, WO 93/01288, WO 92/01047, WO 92/09690; Fuchs et al, Bio/Technology 9: 1370-1372 (1991); Hay et al, Hum. Antibod. Hybridomas
3:81-85 (1992); Huse et al, Science 246:1275-1281 (1989); McCafferty et al, Nature 348:552-554 (1990); Griffiths et al, EMBOJ. 12:725-734 (1993); Hawkins et al, J Mol. Biol. 226:889-896 (1992); Clackson et al, Nature 352:624-628 (1991); Gram et al, Proc. Natl. Acad. Sci. USA 89:3576-3580 (1992); Ganad et al, Bio/Technology 9:1373-1377 (1991); Hoogenboom et al, Nuc. Acid Res.
19:4133-4137 (1991); and Barbas et al, Proc. Natl. Acad. Sci. USA 88:7978-7982
(1991).
[0183] In one embodiment, to isolate a human anti-CD40 antibodies with the desired characteristics, a human anti-CD40 antibody as described herein is first used to select human heavy and light chain sequences having similar binding activity toward CD40, using the epitope imprinting methods described in PCT Publication No. WO 93/06213. The antibody libraries used in this method are preferably scFv libraries prepared and screened as described in PCT Publication No. WO 92/01047, McCafferty et al, Nature 348:552-554 (1990); and Griffiths et al, EMBOJ. 12:725-734 (1993). The scFv antibody libraries preferably are screened using human CD40 as the antigen. [0184] Once initial human VL and VH domains are selected, "mix and match" experiments are performed, in which different pairs of the initially selected VL and NH segments are screened for CD40 binding to select prefened Ni/NH pair combinations. Additionally, to further improve the quality of the antibody, the N and NH segments of the prefened NL/NH pair(s) can be randomly mutated, preferably within the CDR3 region of NH and/or NL, in a process analogous to the in vivo somatic mutation process responsible for affinity maturation of antibodies during a natural immune response. This in vitro affinity maturation can be accomplished by amplifying NH and NL domains using PCR primers complimentary to the NH CDR3 or NL CDR3, respectively, which primers have been "spiked" with a random mixture of the four nucleotide bases at certain positions such that the resultant PCR products encode NH and N segments into which random mutations have been introduced into the NH and/or NL CDR3 regions. These randomly mutated Nπ and NL segments can be rescreened for binding to CD40. [0185] Following screening and isolation of an anti-CD40 antibody of the invention from a recombinant immunoglobulin display library, nucleic acids encoding the selected antibody can be recovered from the display package (e.g., from the phage genome) and subcloned into other expression vectors by standard recombinant DΝA techniques. If desired, the nucleic acid can further be manipulated to create other antibody forms of the invention, as described below.
To express a recombinant human antibody isolated by screening of a combinatorial library, the DΝA encoding the antibody is cloned into a recombinant expression vector and introduced into a mammalian host cells, as described above.
Class Switching [0186] Another aspect of the invention provides a method for converting the class or subclass of an anti-CD40 antibody to another class or subclass. In some embodiments, a nucleic acid molecule encoding a N or Nπ that does not include any nucleic acid sequences encoding CL or CH is isolated using methods well- known in the art. The nucleic acid molecule then is operatively linked to a nucleic acid sequence encoding a CL or CH from a desired immunoglobulin class or subclass. This can be achieved using a vector or nucleic acid molecule that comprises a CL or CH chain, as described above. For example, an anti-CD40 antibody that was originally IgM can be class switched to an IgG. Further, the class switching may be used to convert one IgG subclass to another, e.g., from IgGl to IgG2. Another method for producing an antibody of the invention comprising a desired isotype comprises the steps of isolating a nucleic acid encoding a heavy chain of an anti-CD40 antibody and a nucleic acid encoding a light chain of an anti-CD40 antibody, isolating the sequence encoding the VH region, ligating the VH sequence to a sequence encoding a heavy chain constant domain of the desired isotype, expressing the light chain gene and the heavy chain construct in a cell, and collecting the anti-CD40 antibody with the desired isotype.
Deimmunized Antibodies
[0187] Another way of producing antibodies with reduced immunogenicity is the deimmunization of antibodies. In another aspect of the invention, the antibody may be deimmunized using the techniques described in, e.g., PCT Publication Nos. WO98/52976 and WO00/34317 (which incorporated herein by reference in their entirety).
Mutated Antibodies
[0188] In another embodiment, the nucleic acid molecules, vectors and host cells may be used to make mutated anti-CD40 antibodies. The antibodies may be mutated in the variable domains of the heavy and/or light chains, e.g., to alter a binding property of the antibody. For example, a mutation may be made in one or more of the CDR regions to increase or decrease the KD of the antibody for CD40, to increase or decrease Koff, or to alter the binding specificity of the antibody. Techniques in site-directed mutagenesis are well-known in the art. See, e.g., Sambrook et al and Ausubel et al, supra. In a prefened embodiment, mutations are made at an amino acid residue that is known to be changed compared to germline in a variable domain of an anti-CD40 antibody. In another embodiment, one or more mutations are made at an amino acid residue that is known to be changed compared to the germline in a CDR region or framework region of a variable domain, or in a constant domain of a monoclonal antibody 3.1.1, 3.1.1 H- A78T, 3.1.1H-A78T-V88A-V97A, 3.1.1L-L4M-L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K and 24.2.1. In another embodiment, one or more mutations are made at an amino acid residue that is known to be changed compared to the germline in a CDR region or framework region of a variable domain of an amino acid sequence selected from SEQ ID NOS: 4, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 94, 100, 102, 2, 10, 18, 26, 34, 42, 50, 58, 66, 74, 82, 90, 92, 96, 98, 100 or 102, or whose nucleic acid sequence is presented in SEQ ID NOS: 3, 11, 19, 27, 35, 43, 51, 59, 67, 75, 83, 93, 99, 101, 1, 9, 17, 25, 33, 41, 49, 57, 65, 73, 81, 89, 91, 95, 97, 99 or 101. [0189] In one embodiment, the framework region is mutated so that the resulting framework region(s) have the amino acid sequence of the conesponding germline gene. A mutation may be made in a framework region or constant domain to increase the half-life of the anti-CD40 antibody. See, e.g., PCT Publication No. WO 00/09560, herein incorporated by reference. A mutation in a framework region or constant domain also can be made to alter the immunogenicity of the antibody, to provide a site for covalent or non-covalent binding to another molecule, or to alter such properties as complement fixation, FcR binding and ADCC. According to the invention, a single antibody may have mutations in any one or more of the framework regions, the constant domain and in the variable regions. [0190] In some embodiments, there are from 1 to 18, including any number in between, amino acid mutations in either the VH or VL domains of the mutated anti- CD40 antibody compared to the anti-CD40 antibody prior to mutation. In any of the above, the mutations may occur in one or more CDR regions. Further, any of the mutations can be conservative amino acid substitutions. In some embodiments, there are no more than 5, 4, 3, 2, or 1 amino acid changes in the constant domains.
Modified Antibodies [0191] In another embodiment, a fusion antibody or immunoadhesin may be made that comprises all or a portion of an anti-CD40 antibody of the invention linked to another polypeptide. In a prefened embodiment, only the variable domains of the anti-CD40 antibody are linked to the polypeptide. In another prefened embodiment, the VH domain of an anti-CD40 antibody is linked to a first polypeptide, while the VL domain of an anti-CD40 antibody is linked to a second polypeptide that associates with the first polypeptide in a manner such that the VH and VL domains can interact with one another to form an antibody binding site. In another prefened embodiment, the VH domain is separated from the VL domain by a linker such that the VH and VL domains can interact with one another (see below under Single Chain Antibodies). The VH-linker-N antibody is then linked to the polypeptide of interest. The fusion antibody is useful for directing a polypeptide to a CD40-exρressing cell or tissue. The polypeptide may be a therapeutic agent, such as a toxin, growth factor or other regulatory protein, or may be a diagnostic agent, such as an enzyme that may be easily visualized, such as horseradish peroxidase. In addition, fusion antibodies can be created in which two (or more) single-chain antibodies are linked to one another. This is useful if one wants to create a divalent or polyvalent antibody on a single polypeptide chain, or if one wants to create a bispecific antibody. [0192] To create a single chain antibody, (scFv) the NH- and NL-encoding DΝA fragments are operatively linked to another fragment encoding a flexible linker, e.g., encoding the amino acid sequence (Gly4 -Ser)3, such that the NH and NL sequences can be expressed as a contiguous single-chain protein, with the N and NH domains joined by the flexible linker. See, e.g., Bird et al, Science 242:423-426 (1988); Huston et al, Proc. Natl. Acad. Sci. USA 85:5879-5883
(1988); McCafferty et al, N-.t-.re 348:552-554 (1990). The single chain antibody may be monovalent, if only a single NH and NL are used, bivalent, if two NH and NL are used, or polyvalent, if more than two NH and NL are used. Bispecific or polyvalent antibodies may be generated that bind specifically to CD40 and to another molecule.
[0193] In other embodiments, other modified antibodies may be prepared using anti-CD40 antibody-encoding nucleic acid molecules. For instance, "Kappa bodies" (111 et al, Protein Eng. 10: 949-57 (1997)), "Minibodies" (Martin et al, EMBO J. 13: 5303-9 (1994)), "Diabodies" (HoUiger et al, Proc. Natl. Acad. Sci. USA 90: 6444-6448 (1993)), or "Janusins" (Traunecker et al, EMBOJ. 10:3655- 3659 (1991) and Traunecker et al, Int. J. Cancer (Suppl.) 7:51-52 (1992)) maybe prepared using standard molecular biological techniques following the teachings of the specification.
[0194] Bispecific antibodies or antigen-binding fragments can be produced by a variety of methods including fusion of hybridomas or linking of Fab' fragments. See, e.g., Songsivilai & Lachmann, Clin. Exp. Immunol. 79: 315-321 (1990), Kostelny et al, J Immunol. 148:1547-1553 (1992). In addition, bispecific antibodies may be formed as "diabodies" or "Janusins." In some embodiments, the bispecific antibody binds to two different epitopes of CD40. In some embodiments, the bispecific antibody has a first heavy chain and a first light chain from monoclonal antibody 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-N88A-N97A, 3.1.1L-L4M-L83N, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.28.1L-C92A, 23.29.1, 23.29.1L- R174K and 24.2.1, and an additional antibody heavy chain and light chain. In some embodiments, the additional light chain and heavy chain also are from one of the above-identified monoclonal antibodies, but are different from the first heavy and light chains.
[0195] In some embodiments, the modified antibodies described above are prepared using one or more of the variable domains or CDR regions from a human anti-CD40 monoclonal antibody provided herein, from an amino acid sequence of said monoclonal antibody, or from a heavy chain or light chain encoded by a nucleic acid sequence encoding said monoclonal antibody.
Derivatized and Labeled Antibodies
[0196] An anti-CD40 antibody or antigen-binding portion of the invention can be derivatized or linked to another molecule (e.g., another peptide or protein). In general, the antibodies or portion thereof is derivatized such that the CD40 binding is not affected adversely by the derivatization or labeling. Accordingly, the antibodies and antibody portions of the invention are intended to include both intact and modified forms of the human anti-CD40 antibodies described herein. For example, an antibody or antibody portion of the invention can be functionally linked (by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other molecular entities, such as another antibody (e.g., a bispecific antibody or a diabody), a detection agent, a cytotoxic agent, a pharmaceutical agent, and/or a protein or peptide that can mediate associate of the antibody or antibody portion with another molecule (such as a streptavidin core region or a polyhistidine tag).
[0197] One type of derivatized antibody is produced by crosslinking two or more antibodies (of the same type or of different types, e.g., to create bispecific antibodies). Suitable crosslinkers include those that are heterobifunctional, having two distinctly reactive groups separated by an appropriate spacer (e.g., m-maleimidobenzoyl-N-hydroxysuccinimide ester) or homobifunctional (e.g., disuccinimidyl suberate). Such linkers are available from Pierce Chemical Company, Rockford, 111. [0198] Another type of derivatized antibody is a labeled antibody. Useful detection agents with which an antibody or antigen-binding portion of the invention may be derivatized include fluorescent compounds, including fluorescein, fluorescein isothiocyanate, rhodamine, 5-dimethylamine-l-napthalenesulfonyl chloride, phycoerythrin, lanthanide phosphors and the like. An antibody can also be labeled with enzymes that are useful for detection, such as horseradish peroxidase, j3-galactosidase, luciferase, alkaline phosphatase, glucose oxidase and the like. When an antibody is labeled with a detectable enzyme, it is detected by adding additional reagents that the enzyme uses to produce a reaction product that can be discerned. For example, when the agent horseradish peroxidase is present, the addition of hydrogen peroxide and diaminobenzidine leads to a colored reaction product, which is detectable. An antibody can also be labeled with biotin, and detected through indirect measurement of avidin or streptavidin binding. An antibody can also be labeled with a predetermined polypeptide epitope recognized by a secondary reporter (e.g., leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags). In some embodiments, labels are attached by spacer arms of various lengths to reduce potential steric hindrance. [0199] An anti-CD40 antibody can also be labeled with a radiolabeled amino acid. The radiolabel can be used for both diagnostic and therapeutic purposes. For instance, the radiolabel can be used to detect CD40-expressing tumors by x-ray or other diagnostic techniques. Further, the radiolabel can be used therapeutically as a toxin for cancerous cells or tumors. Examples of labels for polypeptides include, but are not limited to, the following radioisotopes or radionuclides — 3H, 14C, 15N,
35S, 90Y, 99Tc, n ιIn, 125I, 131I.
[0200] An anti-CD40 antibody can also be derivatized with a chemical group such as polyethylene glycol (PEG), a methyl or ethyl group, or a carbohydrate group. These groups are useful to improve the biological characteristics of the antibody, e.g., to increase serum half-life or to increase tissue binding.
Pharmaceutical Compositions and Kits
[0201] The invention also relates to compositions comprising a human anti- CD40 agonist antibody for the treatment of subjects in need of immunostimulation. Such compositions are useful to treat, prevent, reduce the frequency of or severity of infection, including viral and bacterial infection, for treating a hyperproliferative disorder, including cancerous and pre-cancerous conditions, for treating genetic immunodeficiency conditions, such as hyper-IgM syndrome and for treating primary or combined immunodeficiency conditions, including conditions characterized by neutropenia, in a mammal, including humans. Subjects for treatment with agonist anti-CD40 antibody therapy include any subject in need of immune enhancement, including but not limited to the elderly and individuals who are immunosuppressed, for example due to chemotherapy. [0202] Hyperproliferative disorders that may be treated by an agonist anti-CD40 antibody of the invention can involve any tissue or organ and include but are not limited to brain, lung, squamous cell, bladder, gastric, pancreatic, breast, head, neck, liver, renal, ovarian, prostate, colorectal, esophageal, gynecological, nasopharynx, or thyroid cancers, melanomas, lymphomas, leukemias or multiple myelomas. In particular, human agonist anti-CD40 antibodies of the invention are useful to treat carcinomas of the breast, prostate, colon and lung.
[0203] Treatment may involve administration of one or more agonist anti-CD40 monoclonal antibodies of the invention, or antigen-binding fragments thereof, alone or with a pharmaceutically acceptable carrier. As used herein, "pharmaceutically acceptable carrier" means any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Some examples of pharmaceutically acceptable carriers are water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition. Additional examples of pharmaceutically acceptable substances are wetting agents or minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the antibody.
[0204] Agonist anti-CD40 antibodies of the invention and compositions comprising them, can be administered in combination with one or more other therapeutic, diagnostic or prophylactic agents. Additional therapeutic agents include other anti-neoplastic, anti-tumor, anti-angiogenic or chemotherapeutic agents. Such additional agents may be included in the same composition or administered separately. In some embodiments, one or more agonist anti-CD40 antibodies of the invention can be used as a vaccine or as adjuvants to a vaccine. [0205] The compositions of this invention may be in a variety of forms, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, liposomes and suppositories. The prefened form depends on the intended mode of administration and therapeutic application. Typical prefened compositions are in the form of injectable or infusible solutions, such as compositions similar to those used for passive immunization of humans. The prefened mode of administration is parenteral (e.g., intravenous, subcutaneous, intraperitoneal, intramuscular). In a prefened embodiment, the antibody is administered by intravenous infusion or injection. In another prefened embodiment, the antibody is administered by intramuscular or subcutaneous injection. [0206] Therapeutic compositions typically must be sterile and stable under the conditions of manufacture and storage. The composition can be formulated as a solution, microemulsion, dispersion, liposome, or other ordered stracture suitable to high drag concentration. Sterile injectable solutions can be prepared by incorporating the anti-CD40 antibody in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the prefened methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. The proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prolonged absorption of injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, monostearate salts and gelatin. [0207] The antibodies of the present invention can be administered by a variety of methods known in the art, although for many therapeutic applications, the prefened route/mode of administration is subcutaneous, intramuscular, or intravenous infusion. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results. [0208] In certain embodiments, the antibody compositions active compound may be prepared with a carrier that will protect the antibody against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art.
See, e.g., Sustained and Controlled Release Drag Delivery Systems (J. R. Robinson, ed., Marcel Dekker, Inc., New York, 1978). [0209] In certain embodiments, an anti-CD40 antibody of the invention can be orally administered, for example, with an inert diluent or an assimilable edible carrier. The compound (and other ingredients, if desired) can also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or incorporated directly into the subject's diet. For oral therapeutic administration, the anti-CD40 antibodies can be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. To administer a compound of the invention by other than parenteral administration, it may be necessary to coat the compound with, or co-administer the compound with, a material to prevent its inactivation.
[0210] Additional active compounds also can be incorporated into the compositions. In certain embodiments, an anti-CD40 antibody of the invention is co-formulated with and/or co-administered with one or more additional therapeutic agents. These agents include, without limitation, antibodies that bind other targets (e.g., antibodies that bind one or more growth factors or cytokines or their cell surface receptors, such as anti-CTL4-antibody), antineoplastic agents, antitumor agents, chemotherapeutic agents, peptide analogues that activate CD40, soluble CD40L, one or more chemical agents that activates CD40, and/or other agents known in the art that can enhance an immune response against tumor cells, e.g., IFN-jSl, IL-2, IL-8, IL-12, IL-15, IL-18, IL-23, IFN-γ, and GM-CSF. Such combination therapies may require lower dosages of the anti-CD40 antibody as well as the co-administered agents, thus avoiding possible toxicities or complications associated with the various monotherapies. [0211 ] Agonist anti-CD40 antibodies of the invention and compositions comprising them also may be administered in combination with other therapeutic regimens, in particular in combination with radiation treatment. [0212] The compositions of the invention may include a "therapeutically effective amount" or a "prophylactically effective amount" of an antibody or antigen-binding portion of the invention. A "therapeutically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result. A therapeutically effective amount of the antibody or antibody portion may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the antibody or antibody portion to elicit a desired response in the individual. A therapeutically effective amount is also one in which any toxic or detrimental effects of the antibody or antibody portion are outweighed by the therapeutically beneficial effects. A "prophylactically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount. [0213] Dosage regimens can be adjusted to provide the optimum desired response (e.g., a therapeutic or prophylactic response). For example, a single bolus can be administered, several divided doses can be administered over time or the dose can be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the anti-CD40 antibody or portion and the particular therapeutic or prophylactic effect to be achieved, and (b) the limitations inherent in the art of compounding such an antibody for the treatment of sensitivity in individuals. [0214] An exemplary, non-limiting range for a therapeutically or prophylactically effective amount of an antibody or antibody portion of the invention is 0.025 to 50 mg/kg, more preferably 0.1 to 50 mg/kg, more preferably 0.1-25, 0.1 to 10 or 0.1 to 3 mg/kg. It is to be noted that dosage values may vary with the type and severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that dosage ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition.
[0215] Another aspect of the present invention provides kits comprising an anti- CD40 antibody or antibody portion of the invention or a composition comprising such an antibody. A kit may include, in addition to the antibody or composition, diagnostic or therapeutic agents. A kit can also include instructions for use in a diagnostic or therapeutic method. In a prefened embodiment, the kit includes the antibody or a composition comprising it and a diagnostic agent that can be used in a method described below. In another prefened embodiment, the kit includes the antibody or a composition comprising it and one or more therapeutic agents that can be used in a method described below.
[0216] This invention also relates to compositions for inhibiting abnormal cell growth in a mammal comprising an amount of an antibody of the invention in combination with an amount of a chemotherapeutic, wherein the amounts of the compound, salt, solvate, or prodrug, and of the chemotherapeutic are together effective in inhibiting abnormal cell growth. Many chemotherapeutics are presently known in the art. In some embodiments, the chemotherapeutic is selected from the group consisting of mitotic inhibitors, alkylating agents, anti- metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biological response modifiers, anti-hormones, e.g. anti-androgens, and anti-angiogenesis agents.
[0217] Anti-angiogenic agents, such as MMP-2 (matrix-metalloproteinase 2) inhibitors, MMP-9 (matrix-metalloproteinase 9) inhibitors, and COX-II (cyclooxygenase II) inhibitors, can be used in conjunction with an anti-CD40 antibody of the invention. Examples of useful COX-II inhibitors include
CELEBREX™ (alecoxib), valdecoxib, and rofecoxib. Examples of useful matrix metalloproteinase inhibitors are described in WO 96/33172 (published October 24, 1996), WO 96/27583 (published March 7, 1996), European Patent Application No. 97304971.1 (filed July 8, 1997), European Patent Application No. 99308617.2 (filed October 29, 1999), WO 98/07697 (published February 26, 1998), WO
98/03516 (published January 29, 1998), WO 98/34918 (published August 13, 1998), WO 98/34915 (published August 13, 1998), WO 98/33768 (published August 6, 1998), WO 98/30566 (published July 16, 1998), European Patent Publication 606,046 (published July 13, 1994), European Patent Publication 931,788 (published July 28, 1999), WO 90/05719 (published May 31, 1990), WO 99/52910 (published October 21, 1999), WO 99/52889 (published October 21, 1999), WO 99/29667 (published June 17, 1999), PCT International Application No. PCT/IB98/01113 (filed July 21, 1998), European Patent Application No. 99302232.1 (filed March 25, 1999), Great Britain patent application number 9912961.1 (filed June 3, 1999), U.S. Provisional Application No. 60/148,464 (filed August 12, 1999), U.S. Patent 5,863,949 (issued January 26, 1999), U.S. Patent 5,861,510 (issued January 19, 1999), and European Patent Publication 780,386 (published June 25, 1997), all of which are incorporated herein in their entireties by reference. Prefened MMP inhibitors are those that do not demonstrate arthralgia. More prefened, are those that selectively inhibit MMP-2 and/or MMP- 9 relative to the other matrix-metalloproteinases (i.e. MMP-1, MMP-3, MMP-4, MMP-5, MMP-6, MMP-7, MMP-8, MMP-10, MMP-11, MMP-12, and MMP-13). Some specific examples of MMP inhibitors useful in the present invention are AG- 3340, RO 32-3555, RS 13-0830, and the compounds recited in the following list: 3-[[4-(4-fluoro-phenoxy)-benzenesulfonyl]-(l-hydroxycarbamoyl-cyclopentyl)- amino] -propionic acid; 3-exo-3-[4-(4-fluoro-phenoxy)-benzenesulfonylamino]-8- oxa-bicyclo [3.2. l]octane-3 -carboxylic acid hydroxyamide; (2R, 3R) l-[4-(2- chloro-4-fluoro-benzyloxy)-benzenesulfonyl]-3-hydroxy-3-methyl-piperidine-2- carboxylic acid hydroxyamide; 4-[4-(4-fluoro-phenoxy)-benzenesulfonylamino]- tetrahydro-pyran-4-carboxylic acid hydroxyamide; 3-[[4-(4-fluoro-phenoxy)- benzenesulfonyl]-(l -hydroxycarbamoyl-cyclobutyl)-amino]-propionic acid; 4-[4- (4-chloro-phenoxy)-benzenesulfonylamino]-tetrahydro-ρyran-4-carboxylic acid hydroxyamide; (R) 3-[4-(4-chloro-phenoxy)-benzenesulfonylamino]-tetrahydro- pyran-3 -carboxylic acid hydroxyamide; (2R, 3R) l-[4-(4-fluoro-2-methyl- benzyloxy)-benzenesulfonyl]-3-hydroxy-3-methyl-piperidine-2-c---'boxylic acid hydroxyamide; 3-[[4-(4-fluoro-phenoxy)-benzenesulfonyl]-(l-hydroxycarbamoyl- l-methyl-ethyl)-amino]-propionic acid; 3-[[4-(4-fluoro-phenoxy)- benzenesulfonyl]-(4-hydroxycarbamoyl-tetrahydro-pyran-4-yl)-amino]-propionic acid; 3-exo-3-[4-(4-chloro-phenoxy)-benzenesulfonylamino]-8-oxa- bi cyclo[3.2. l]octane-3 -carboxylic acid hydroxyamide; 3-endo-3-[4-(4-fluoro- phenoxy)-benzenesulfonylamino]-8-oxa-bicyclo[3.2.1]octane-3-carboxylic acid hydroxyamide; and (R) 3-[4-(4-fluoro-phenoxy)-benzenesulfonylamino]- tetrahydro-furan-3-carboxylic acid hydroxyamide; and pharmaceutically acceptable salts and solvates of said compounds.
[0218] A compound of the invention can also be used with signal transduction inhibitors, such as agents that can inhibit EGF-R (epidermal growth factor receptor) responses, such as EGF-R antibodies, EGF antibodies, and molecules that are EGF-R inhibitors; NEGF (vascular endothelial growth factor) inhibitors, such as NEGF receptors and molecules that can inhibit NEGF; and erbB2 receptor inhibitors, such as organic molecules or antibodies that bind to the erbB2 receptor, for example, HERCEPTIΝTM (Genentech, Inc.). EGF-R inhibitors are described in, for example in WO 95/19970 (published July 27, 1995), WO 98/14451 (published April 9, 1998), WO 98/02434 (published January 22, 1998), and United States Patent 5,747,498 (issued May 5, 1998), and such substances can be used in the present invention as described herein. EGFR-inhibiting agents include, but are not limited to, the monoclonal antibodies C225 and anti-EGFR 22Mab (ImClone Systems Incorporated), ABX-EGF (Abgenix/Cell Genesys), EMD-7200 (Merck KgaA), EMD-5590 (Merck KgaA), MDX-447/H-477 (Medarex Inc. and Merck KgaA), and the compounds ZD-1834, ZD-1838 and ZD-1839 (AstraZeneca), PKI-
166 (Νovartis), PKI-166/CGP-75166 (Νovartis), PTK 787 (Νovartis), CP 701 (Cephalon), leflunomide (Pharmacia/Sugen), CI-1033 (Warner Lambert Parke Davis), CI-1033/PD 183,805 (Warner Lambert Parke Davis), CL-387,785 (Wyeth- Ayerst), BBR-1611 (Boehringer Mannheim GmbH/Roche), Νaamidine A (Bristol Myers Squibb), RC-3940-II (Pharmacia), BIBX-1382 (Boehringer fngelheim), OLX-103 (Merck & Co.), NRCTC-310 (Nentech Research), EGF fusion toxin (Seragen Inc.), DAB-389 (Seragen Lilgand), ZM-252808 (Imperial Cancer Research Fund), RG-50864 (IΝSERM), LFM-A12 (Parker Hughes 'Cancer Center), WHI-P97 (Parker Hughes Cancer Center), GW-282974 (Glaxo), KT-8391 (Kyowa Hakko) and EGF-R Naccine (York Medical/Centro de Immunologia Molecular
(CIM)). These and other EGF-R-inhibiting agents can be used in the present invention. [0219] VEGF inhibitors, for example SU-5416 and SU-6668 (Sugen Inc.), SH- 268 (Schering), and NX-1838 (NeXstar) can also be combined with the compound of the present invention. VEGF inhibitors are described in, for example in WO 99/24440 (published May 20, 1999), PCT International Application PCT/IB99/00797 (filed May 3, 1999), in WO 95/21613 (published August 17, 1995), WO 99/61422 (published December 2, 1999), United States Patent 5,834,504 (issued November 10, 1998), WO 98/50356 (published November 12, 1998), United States Patent 5,883,113 (issued March 16, 1999), United States Patent 5,886,020 (issued March 23, 1999), United States Patent 5,792,783 (issued August 11, 1998), WO 99/10349 (published March 4, 1999), WO 97/32856
(published September 12, 1997), WO 97/22596 (published June 26, 1997), WO 98/54093 (published December 3, 1998), WO 98/02438 (published January 22, 1998), WO 99/16755 (published April 8, 1999), and WO 98/02437 (published January 22, 1998), all of which are incorporated herein in their entireties by reference. Other examples of some specific VEGF inhibitors useful in the present invention are IM862 (Cytran Inc.); anti-VEGF monoclonal antibody of Genentech, Inc.; and angiozyme, a synthetic ribozyme from Ribozyme and Chiron. These and other VEGF inhibitors can be used in the present invention as described herein. ErbB2 receptor inhibitors, such as GW-282974 (Glaxo Wellcome pic), and the monoclonal antibodies AR-209 (Aronex Pharmaceuticals Inc.) and 2B-1 (Chiron), can furthermore be combined with the compound of the invention, for example those indicated in WO 98/02434 (published January 22, 1998), WO 99/35146 (published July 15, 1999), WO 99/35132 (published July 15, 1999), WO 98/02437 (published January 22, 1998), WO 97/13760 (published April 17, 1997), WO 95/19970 (published July 27, 1995), United States Patent 5,587,458 (issued
December 24, 1996), and United States Patent 5,877,305 (issued March 2, 1999), which are all hereby incorporated herein in their entireties by reference. ErbB2 receptor inhibitors useful in the present invention are also described in United States Provisional Application No. 60/117,341, filed January 27, 1999, and in United States Provisional Application No. 60/117,346, filed January 27, 1999, both of which are incorporated in their entireties herein by reference. The erbB2 receptor inhibitor compounds and substance described in the aforementioned PCT applications, U.S. patents, and U.S. provisional applications, as well as other compounds and substances that inhibit the erbB2 receptor, can be used with the compound of the present invention in accordance with the present invention. [0220] Anti-survival agents include anti-IGF-ER antibodies and anti-integrin agents, such as anti-integrin antibodies.
Diagnostic Methods of Use
[0221] In another aspect, the invention provides diagnostic methods. The anti- CD40 antibodies can be used to detect CD40 in a biological sample in vitro or in vivo. In one embodiment, the invention provides a method for diagnosing the presence or location of an CD40-expressing tumor in a subject in need thereof, comprising the steps of injecting the antibody into the subject, determining the expression of CD40 in the subject by localizing where the antibody has bound, comparing the expression in the subject with that of a normal reference subject or standard, and diagnosing the presence or location of the tumor. [0222] The anti-CD40 antibodies can be used in a conventional immunoassay, including, without limitation, an ELISA, an RIA, FACS, tissue immunohistochemistry, Western blot or immunoprecipitation. The anti-CD40 antibodies of the invention can be used to detect CD40 from humans. In another embodiment, the anti-CD40 antibodies can be used to detect CD40 from Old World primates such as cynomolgus and rhesus monkeys, chimpanzees and apes. The invention provides a method for detecting CD40 in a biological sample comprising contacting a biological sample with an anti-CD40 antibody of the invention and detecting the bound antibody. In one embodiment, the anti-CD40 antibody is directly labeled with a detectable label. In another embodiment, the anti-CD40 antibody (the first antibody) is unlabeled and a second antibody or other molecule that can bind the anti-CD40 antibody is labeled. As is well known to one of skill in the art, a second antibody is chosen that is able to specifically bind the particular species and class of the first antibody. For example, if the anti-CD40 antibody is a human IgG, then the secondary antibody could be an anti-human- IgG. Other molecules that can bind to antibodies include, without limitation,
Protein A and Protein G, both of which are available commercially, e.g., from Pierce Chemical Co. [0223] Suitable labels for the antibody or secondary antibody have been disclosed supra, and include various enzymes, prosthetic groups, fluorescent materials, luminescent materials and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, /3-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; and examples of suitable radioactive material include1251, 1311, 35S or 3H.
[0224] In other embodiments, CD40 can be assayed in a biological sample by a competition immunoassay utilizing CD40 standards labeled with a detectable substance and an unlabeled anti-CD40 antibody. In this assay, the biological sample, the labeled CD40 standards and the anti-CD40 antibody are combined and the amount of labeled CD40 standard bound to the unlabeled antibody is determined. The amount of CD40 in the biological sample is inversely proportional to the amount of labeled CD40 standard bound to the anti-CD40 antibody. [0225] One can use the immunoassays disclosed above for a number of purposes. For example, the anti-CD40 antibodies can be used to detect CD40 in cells in cell culture. In a prefened embodiment, the anti-CD40 antibodies are used to determine the amount of CD40 on the surface of cells that have been treated with various compounds. This method can be used to identify compounds that are useful to activate or inhibit CD40. According to this method, one sample of cells is treated with a test compound for a period of time while another sample is left untreated. If the total level of CD40 is to be measured, the cells are lysed and the total CD40 level is measured using one of the immunoassays described above. The total level of CD40 in the treated versus the untreated cells is compared to determine the effect of the test compound. [0226] A prefened immunoassay for measuring total CD40 levels is an ELISA or
Western blot. If the cell surface level of CD40 is to be measured, the cells are not lysed, and the cell surface levels of CD40 are measured using one of the immunoassays described above. A prefened immunoassay for determining cell surface levels of CD40 includes the steps of labeling the cell surface proteins with a detectable label, such as biotin or 125I, immunoprecipitating the CD40 with an anti-CD40 antibody and then detecting the labeled CD40. Another prefened immunoassay for determining the localization of CD40, e.g., cell surface levels, is by using immunohistochemistry. Methods such as ELISA, RIA, Western blot, immunohistochemistry, cell surface labeling of integral membrane proteins and immunoprecipitation are well known in the art. See, e.g., Harlow and Lane, supra. In addition, the immunoassays can be scaled up for high throughput screening in order to test a large number of compounds for either activation or inhibition of CD40.
[0227] The anti-CD40 antibodies of the invention can also be used to determine the levels of CD40 in a tissue or in cells derived from the tissue. In some embodiments, the tissue is a diseased tissue. In some embodiments, the tissue is a tumor or a biopsy thereof, hi some embodiments of the method, a tissue or a biopsy thereof is excised from a patient. The tissue or biopsy is then used in an immunoassay to determine, e.g., total CD40 levels, cell surface levels of CD40 or localization of CD40 by the methods discussed above. [0228] The above-described diagnostic method can be used to determine whether a tumor expresses high levels of CD40, which could be indicative that the tumor is a target for treatment with anti-CD40 antibody. Further, the same method can also be used to monitor the effect of the treatment with anti-CD40 antibody by detecting cell death in the tumor. The diagnostic method can also be used to determine whether a tissue or cell expresses insufficient levels of CD40 or activated CD40, and thus is a candidate for treatment with activating anti-CD40 antibodies, CD40L and or other therapeutic agents for increasing CD40 levels or activity.
[0229] The antibodies of the present invention can also be used in vivo to identify tissues and organs that express CD40. In some embodiments, the anti-CD40 antibodies are used to identify CD40-expressing tumors. One advantage of using the human anti-CD40 antibodies of the present invention is that they may safely be used in vivo without eliciting an immune response to the antibody upon administration, unlike antibodies of non-human origin or with humanized antibodies.
[0230] The method comprises the steps of administering a detectably labeled an anti-CD40 antibody or a composition comprising them to a patient in need of such a diagnostic test and subjecting the patient to imaging analysis to determine the location of the CD40-expressing tissues. Imaging analysis is well known in the medical art, and includes, without limitation, x-ray analysis, magnetic resonance imaging (MRI) or computed tomography (CE). The antibody can be labeled with any agent suitable for in vivo imaging, for example a contrast agent, such as barium, which can be used for x-ray analysis, or a magnetic contrast agent, such as a gadolinium chelate, which can be used for MRI or CE. Other labeling agents include, without limitation, radioisotopes, such as 99Tc. In another embodiment, the anti-CD40 antibody will be unlabeled and will be imaged by administering a second antibody or other molecule that is detectable and that can bind the anti- CD40 antibody. In embodiment, a biopsy is obtained from the patient to determine whether the tissue of interest expresses CD40.
Therapeutic Methods of Use
[0231] In another aspect, invention provides therapeutic methods of using an anti-CD40 antibody of the invention. [0232] A human agonist anti-CD40 antibody of the invention can be administered to a human or to a non-human mammal that expresses a cross- reacting CD40. The antibody can be administered to such a non-human mammal (i.e., a primate, cynomolgus or rhesus monkey) for veterinary purposes or as an animal model of human disease. Such animal models are useful for evaluating the therapeutic efficacy of antibodies of this invention.
[0233] In some embodiments, the anti-CD40 antibody is administered to a subject who suffers from primary and/or combined immunodeficiencies, including CD40- dependent immunodeficiency with Hyper-IgM syndrome, Common Variable Immunodeficiency, Braton's Agammaglobulinemia, IgG subclass deficiencies, and X-linked SCID (common gamma chain mutations). In some embodiments, the anti-CD40 antibody is administered to treat a subject who is immunosuppressed, for example due to chemotherapy, or has an immune- debilitating disease, including any acquired immune deficiency disease, such as HIV. In some embodiments, the anti-CD40 antibody is administered to enhance the immunity of an elderly subject. In some embodiments, the anti-CD40 antibody is administered to treat a subject who has a bacterial, viral, fungal or parasitic infection. In some embodiments, a human agonist anti-CD40 antibody of the invention may be administered prophylactically to a subject who, because of age, illness or general poor health is susceptible to infection to prevent or to reduce the number or severity of infections. [0234] In some embodiments, the anti-CD40 antibody is administered to a subject who has a hyperproliferative disorder.
[0235] In some embodiments, the anti-CD40 antibody is administered to treat a subject who has a tumor, i some embodiments, the tumor is CD40 positive. In some embodiments, the tumor is a CD40 negative. The tumor can be a solid tumor or a non-solid tumor such as lymphoma. In some embodiments, an anti-CD40 antibody is administered to a patient who has a tumor that is cancerous. In some embodiments, the antibody inhibits cancer cell proliferation, inhibits or prevents an increase in tumor weight or volume, and/or causes a decrease in tumor weight or volume.
[0236] Patients that can be treated with anti-CD40 antibodies or antibody portions of the invention include, but are not limited to, patients that have been diagnosed as having brain cancer, lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head and neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, gastric cancer, colorectal cancer, colon cancer, breast cancer, gynecologic tumors (e.g., uterine sarcomas, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina or carcinoma of the vulva), cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system (e.g., cancer of the thyroid, parathyroid or adrenal glands), sarcomas of soft tissues, leukemia, myeloma, multiple myeloma, cancer of the urethra, cancer of the penis, prostate cancer, chronic or acute leukemia, solid tumors of childhood, Hodgkin's disease, lymphocytic lymphomas, non-Hodgkin lymphoma, cancer of the bladder, liver cancer, renal cancer, cancer of the kidney or ureter (e.g., renal cell carcinoma, carcinoma of the renal pelvis), or neoplasms of the central nervous system (e.g., primary CNS lymphoma, spinal axis tumors, brain stem "gliόmas or pituitary adenomas), glioma or fibrosarcoma. [0237] The antibody may be administered from three times daily to once every six months, and preferably may be admimstered via an oral, mucosal, buccal, intranasal, inhalable, intravenous, subcutaneous, intramuscular, parenteral, intratumor, transdermal or topical route. The antibody can also be administered continuously via a minipump. The antibody generally will be administered for as long as the tumor is present provided that the antibody causes the tumor or cancer to stop growing or to decrease in weight or volume. The dosage of antibody generally will be in the range of 0.025 to 50 mg/kg, more preferably 0.1 to 50 mg/kg, more preferably 0.1-20 mg/kg, 0.1-10 mg/kg, 0.1-5 mg/kg or even more preferable 0.1-2 mg/kg.. The antibody can also be administered prophylactically. [0238] hi some embodiments, the anti-CD40 antibody is administered as part of a therapeutic regimen that includes one or more additional antineoplastic drags or molecules to a patient who has a hyperproliferative disorder, such as cancer or a tumor. Exemplary antitumor agents include, but are not limited to, mitotic inhibitors, alkylating agents, anti-metabolites, intercalating agents, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biological response modifiers, anti-hormones, kinase inhibitors, matrix metalloprotease inhibitors, genetic therapeutics and anti-androgens. In more prefened embodiments, the anti-CD40 antibody is administered with an antineoplastic agent, such as adriamycin or taxol In some prefened embodiments, the anti-CD40 therapy is performed along with radiotherapy, chemotherapy, photodynamic therapy, surgery or other immunotherapy. In some embodiments, the anti-CD40 antibody is administered with one or more additional antibodies. For example, the anti-CD40 antibody can be administered with antibodies that are known to inhibit tumor or cancer cell proliferation. Such antibodies include, but are -not limited to, an antibody that inhibits CTLA4, erbB2 receptor, EGF-R, IGF-1R, CD20 or VEGF.
[0239] In some embodiments, the anti-CD40 antibody is labeled with a radiolabel, an immunotoxin or a toxin, or is a fusion protein comprising a toxic peptide. The anti-CD40 antibody or anti-CD40 antibody fusion protein directs the radiolabel, immunotoxin, toxin or toxic peptide to the tumor or cancer cell, hi a prefened embodiment, the radiolabel, immunotoxin, toxin or toxic peptide is internalized by the tumor or cancer cell after the anti-CD40 antibody binds to the CD40 on the surface of the cell.
[0240] In another aspect, the anti-CD40 antibody can be used therapeutically to induce apoptosis of specific cells in a patient. In many cases, the cells targeted for apoptosis are cancerous or tumor cells. Thus, the invention provides a method of inducing apoptosis by administering an anti-CD40 antibody to a patient in need thereof.
[0241] In another aspect, the invention provides a method of administering an activating anti-CD40 antibody to a patient to increase CD40 activity. An anti- CD40 antibody is administered with one or more other factors that increase CD40 activity. Such factors include CD40L, and/or analogues of CD40L that activate CD40.
[0242] In some embodiments, the anti-CD40 antibody is administered with one or more additional immune enhancing agents, including, without limitation IFN- βl, IL-2, IL-8, IL-12, IL-15, IL-18, IL-23, IFN-γ, and GM-CSF. [0243] In some embodiments, a human agonist anti-CD40 antibody of the invention is used as an adjuvant to enhance the efficacy of a vaccine. When used in this way, the anti-CD-40 antibody activates CD40 on antigen presenting cells, including B cells, dendritic cells and monocytes as well as enhancing the production of immunomodulatory molecules, such as cytokines and chemokines. The immunostimulatory effect of the antibody enhances the immune response of the vaccinated subject to the vaccine antigen.
[0244] In another aspect, the invention provides a method for generating a dendritic cell vaccine for cancer or for dendritic cell immunotherapy. According to the method dendritic cells from a cancer patient axe cultured for 1-5 -days with tumor lysate or homogenate, tumor cells killed by inadiation or other means, or tumor specific antigens (e.g., peptides, idiotypes) and 1-10 μg ml of an anti-CD40 antibody. The tumor antigen-pulsed dendritic cells are re-injected into the patient to stimulate anti-tumor immune responses, particularly anti-tumor CTL responses. Monocyte-derived dendritic cells for use in the method can be obtained from a peripheral blood sample by culture in IL-4 and GM-CSF. Dendritic cells also can be derived from the bone manow of a patient by magnetic purification or sorting of CD34 positive cells, followed by culture in IL-4 and GM-CSF.
Gene Therapy
[0245] The nucleic acid molecules of the instant invention can be administered to a patient in need thereof via gene therapy. The therapy may be either in vivo or ex vivo. In a prefened embodiment, nucleic acid molecules encoding both a heavy chain and a light chain are administered to a patient. In a more prefened embodiment, the nucleic acid molecules are administered such that they are stably integrated into chromosomes of B cells because these cells are specialized for producing antibodies. In a prefened embodiment, precursor B cells are transfected or infected ex vivo and re-transplanted into a patient in need thereof. In another embodiment, precursor B cells or other cells are infected in vivo using a virus known to infect the cell type of interest. Typical vectors used for gene therapy include liposomes, plasmids and viral vectors. Exemplary viral vectors are retroviruses, adenoviruses and adeno-associated viruses. After infection either in vivo or ex vivo, levels of antibody expression can be monitored by taking a sample from the treated patient and using any immunoassay known in the art or discussed herein.
[0246] In a prefened embodiment, the gene therapy method comprises the steps of administering an isolated nucleic acid molecule encoding the heavy chain or an antigen-binding portion thereof of an anti-CD40 antibody and expressing the nucleic acid molecule. In another embodiment, the gene therapy method comprises the steps of administering an isolated nucleic acid molecule encoding the light chain or an antigen-binding portion thereof of an anti-CD40 antibody and expressing the nucleic acid molecule. In a more prefened method, the gene therapy method comprises the steps of administering of an isolated nucleic acid molecule encoding the heavy chain or an antigen-binding portion thereof and an isolated nucleic acid molecule encoding the light chain or the antigen-binding portion thereof of an anti-CD40 antibody of the invention and expressing the nucleic acid molecules. The gene therapy method may also comprise the step of administering another anti-cancer agent, such as taxol or adriamycin. [0247] In order that this invention may be better understood, the following examples are set forth. These examples are for purposes of illustration only and are not to be construed as limiting the scope of the invention in any manner.
EXAMPLE I
Generation of Hybridomas Producing Anti-CD40 Antibody
[0248] Antibodies of the invention were prepared, selected, and assayed as follows:
Immunization and hybridoma generation
[0249] We immunized eight to ten week old XenoMice intraperitoneally or in their hind footpads with either a CD40-IgG fusion protein (10 μg/dose/mouse) or with 300.19-CD40 cells which is a transfected cell line that express human CD40 on its plasma membrane (10 x 10 cells/dose/mouse). We repeated this dose five to seven times over a three to eight week period. Four days before fusion, we gave the mice a final injection of the extracellular domain of human CD40 in PBS. We fused the spleen and lymph node lymphocytes from immunized mice with the non- secretory myeloma P3-X63-Ag8.653 cell line, and subjected the fused cells to HAT selection as previously described (Galfre and Milstein, Methods Enzymol. 73:3-46, 1981). We recovered a panel of hybridomas all secreting CD40 specific human IgG2 c antibodies. We selected eleven hybridomas for further study and designated them 3.1.1, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.29.1 and 24.2.1. [0250] We deposited hybridomas 3.1.1, 7.1.2, 10.8.3, 15.1.1 and 21.4.1 in the American Type Culture Collection (ATCC) in accordance with the Budapest
Treaty, 10801 University Boulevard, Manassas, VA 20110-2209, on August 6, 2001. We deposited hybridomas 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1, 23.29.1 and 24.2.1 in the ATCC on July 16, 2002. The hybridomas have been assigned the following deposit numbers: Hybridoma Deposit No.
3.1.1 (LN 15848) PTA-3600 7.1.2 (LN 15849) PTA-3601
10.8.3 (LN 15850) PTA-3602
15.1.1 (LN 15851) PTA-3603
21.4.1 (LN 15853) PTA-3605 21.2.1 (LN 15874) PTA-4549
22.1.1 (LN 15875) PTA-4550
23.5.1 (LN 15855) PTA-4548
23.25.1 (LN 15876) PTA-4551
23.28.1 (LN 15877) PTA-4552 23.29.1 (LN 15878) PTA-4553
24.2.1 (LN 15879) PTA-4554
EXAMPLE II
Sequences of Anti-CD40- Antibodies Prepared in Accordance with the Invention
[0251 ] To analyze the structure of antibodies produced in accordance with the invention, we cloned nucleic acids encoding heavy and light chain fragments from hybridomas producing anti-CD40 monoclonal antibodies. Cloning and sequencing was accomplished as follows.
[0252] We isolated Poly(A)+ mRNA from approximately 2 X 105 hybridoma cells derived from XenoMouse™ mice immunized with human CD40 as described in Example I using a Fast-Track kit (Invitrogen). We followed by PCR the generation of random primed cDNA. We used human VH or human V c family specific variable region primers (Marks et al, "Oligonucleotide primers for polymerase chain reaction amplification of human immunoglobulin variable genes and design of family-specific oligonucleotide probes." Eur. J. Immunol. 21.-985-991 (1991)) or a universal human VH primer, MG-30,
CAGGTGCAGCTGGAGCAGTCIGG (SEQ ID NO: 118), in conjunction with primers specific for the human Cj2 constant region , MG-40d, 5'-GCTGAGGGAGTAGAGTCCTGAGGA-3' (SEQ ID NO: 119) or CK constant region (hcP2; as previously described in Green et al, 1994). We obtained nucleic acid molecules encoding human heavy and kappa light chain transcripts from the anti-CD40 producing hybridomas by direct sequencing of PCR products generated from poly(A ) RNA using the primers described above. We also cloned PCR products into pCRII using a TA cloning kit (Invitrogen) and sequenced both strands using Prism dye-terminator sequencing kits and an ABI 377 sequencing machine. We analyzed all sequences by alignments to the "V BASE sequence directory" (Tomlinson et al, MRC Centre for Protein Engineering, Cambridge, UK) using Mac Vector and Geneworks software programs. [0253] Further, we subjected monoclonal antibodies 3.1.1, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.28.1, 23.29.1 and 24.2.1 to full length DNA cloning and sequencing. For such sequencing, we isolated RNA from approximately 4 X 106 hybridoma cells using QIAGEN RNeasy RNA isolation kit (QIAGEN). We reverse transcribed the mRNA using oligo-dT(l 8) and the Advantage RT/PCR kit (Clonetech). We used V Base to design forward amplification primers that included restriction sites, optimal Kozak sequence, the ATG start site and part of the signal sequence of the heavy chain. Table 1 lists the forward amplification primers used to sequence the antibody clones.
TABLE 1
Figure imgf000079_0001
Figure imgf000080_0001
We used the same method to design a primer to include the 3' coding sequences, the stop codon of the IgG2 constant region, (5 -TTCTCTGATCAGAATTCC TATCATTTACCCGGAGACAGGGAGAG-3') (SEQ ID NO: 125) and restriction sites.
[0254] We also used the same method to design a primer around the ATG start site of the kappa chain: (5'-CTTCAAGCTTACCCGGGCCACCATGAGGCTCC CTGCTCAGC-3') (SEQ ID NO: 126). An optimal Kozak sequence (CCGCCACC) was added 5' to the ATG start site. This primer was used to PCR clone the light chains of following antibody clones: 3.1.1, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1 and 23.29.1. We used a second forward primer 5'-TCTTC AAGCTTGCCCGGGCCCGCCACCATGGAAACCCCAGCGCAG-3' (SEQ ID NO. 134) to clone the light chains of clones 23.28.1 and 24.2.1. We also used the same method to design a primer around the stop codon of the kappa constant region (5'-TTCTTTGATCAGAATTCTCACTAACACTCTCCCCTGTTGAAGC- 3')(SEQ ID NO: 127). We used the primer pairs to amplify the cDNAs using Advantage High Fidelity PCR Kit (Clonetech). We obtained the sequence of the PCR product by direct sequencing using standard techniques (e.g., primer walking) using dye-terminator sequencing kits and an ABI sequencing machine. We cloned the PCR product into a mammalian expression vector and we sequenced clones to confirm somatic mutations. For each clone, we verified the sequence on both strands in at least three reactions.
Gene Utilization Analysis
[0255] Table 2 sets forth the gene utilization evidenced by selected hybridoma clones of antibodies in accordance with the invention: TABLE 2
Heavy and Light Chain Gene Utilization
Figure imgf000081_0001
Sequence And Mutation Analysis
[0256] As will be appreciated, gene utilization analysis provides only a limited overview of antibody stracture. As the B-cells in XenoMouse™ animals stochastically generate V-D-J heavy or V-J kappa light chain transcripts, there are a number of secondary processes that occur, including, without limitation, somatic hypermutation, deletions, Ν-additions, and CDR3 extensions. See, for example, Mendez et al, Nature Genetics 15:146-156 (1997) and International Patent Publication WO 98/24893. Accordingly, to further examine antibody stracture, we generated predicted amino acid sequences of the antibodies from the cDΝAs obtained from the clones. Table A provides the sequence identifiers for each of the nucleotide and predicted amino acid sequences of the sequenced antibodies. [0257] Tables 3-7 provide the nucleotide and predicted amino acid sequences of the heavy and kappa light chains of antibodies 3.1.1 (Table 3), 7.1.2 (Table 4), 10.8.3 (Table 5), 15.1.1 (Table 6) and 21.4.1 (Table 7).
[0258] Tables 8-13 provide the nucleotide and predicted amino acid sequences of the variable domain of the heavy chain and kappa light chain of antibodies 21.2.1 (Table 8), 22.1.1 (Table 9), 23.5.1 (Table 10), 23.28.1 (Table 11), 23.29.1 (Table 12) and 24.2.1 (Table l3).
[0259] The DNA sequence from the full-length sequencing of monoclonal antibody 23.28.1 differs from DNA sequences obtained from sequencing the NH region of the initial PCR product by one base pair (C to G), resulting in a change of residue 16 of the natural heavy chain from D to E.
[0260] Tables 14-19 provide the nucleotide and predicted amino acid sequences of the heavy and kappa light chains of antibodies 21.2.1 (Table 14), 22.1.1 (Table 15), 23.5.1 (Table 16), 23.28.1 (Table 17), 23.29.1 (Table 18) and 24.2.1 (Table 19). In the Tables, the signal peptide sequence (or the bases encoding the same) are underlined.
[0261] We generated two mutated antibodies, 22.1.1 and 23.28.1. The heavy chain of antibody 22.1.1 was mutated to change a cysteine residue at position 109 to an alanine residue. We designated the mutated clone 22.1.1H-C019A. The light chain of antibody 23.28.1 at position 92 was mutated also to change a cysteine residue to an alanine residue. We designated the mutated clone 23.28.1L-C92A. [0262] Mutagenesis of specific residues was carried out by designing primers and using the QuickChange Site Directed Mutagenesis Kit from Stratagene, according to the manufacturer's instructions. Mutations were confirmed by automated sequencing, and mutagenized inserts were subcloned into expression vectors.
[0263] Table 20 provides the nucleotide and amino acid sequences of the mutated heavy chain of antibody 22.1.1H-C109A. Table 21 provides the nucleotide and amino acid sequences of the mutated light chain of antibody 23.28.1. The mutated DΝA codons are shown in italics. The mutated amino acid residue is in bold. Table 3: DNA and protein sequences of antibody 3.1.1
DESCRIPTION: SEQUENCE (signal sequence underlined):
Heavy Chain ATGGAGTTTGGGCTGAGCTGGGTTTTCCTCGTTGC DNA Sequence TCTTTTAAGAGGTGTCCAGTGTCAGGTGCAGCTG
GTGGAGTCTGGGGGAGGCGTGGTCCAGCCTGGG
AGGTCCCTGAGACTCTCCTGTGCAGCCTCTGGAT
TCACCTTCAGTAGTTATGGCATGCACTGGGTCCG
CCAGGCTCCAGGCAAGGGGCTGGAGTGGGTGGC
AGTTATATCAAAGGATGGAGGTAATAAATACCAT
GCAGACTCCGTGAAGGGCCGATTCACCATCTCCA
GAGACAATTCCAAGAATGCGCTGTATCTGCAAAT
GAATAGCCTGAGAGTTGAAGACACGGCTGTGTAT
TACTGTGTGAGAAGAGGGCATCAGCTGGTTCTGG
GATACTACTACTACAACGGTCTGGACGTCTGGGG
CCAAGGGACCACGGTCACCGTCTCCTCAGCCTCC
ACCAAGGGCCCATCGGTCTTCCCCCTGGCGCCCT
GCTCCAGGAGCACCTCCGAGAGCACAGCGGCCCT
GGGCTGCCTGGTCAAGGACTACTTCCCCGAACCG
GTGACGGTGTCGTGGAACTCAGGCGCTCTGACCA
GCGGCGTGCACACCTTCCCAGCTGTCCTACAGTC
CTCAGGACTCTACTCCCTCAGCAGCGTGGTGACC
GTGCCCTCCAGCAACTTCGGCACCCAGACCTACA
CCTGCAACGTAGATCACAAGCCCAGCAACACCAA
GGTGGACAAGACAGTTGAGCGCAAATGTTGTGTC
GAGTGCCCACCGTGCCCAGCACCACCTGTGGCAG
GACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAA
GGACACCCTCATGATCTCCCGGACCCCTGAGGTC
ACGTGCGTGGTGGTGGACGTGAGCCACGAAGAC
CCCGAGGTCCAGTTCAACTGGTACGTGGACGGCG
TGGAGGTGCATAATGCCAAGACAAAGCCACGGG
AGGAGCAGTTCAACAGCACGTTCCGTGTGGTCAG
CGTCCTCACCGTTGTGCACCAGGACTGGCTGAAC
GGCAAGGAGTACAAGTGCAAGGTCTCCAACAAA
GGCCTCCCAGCCCCCATCGAGAAAACCATCTCCA
AAACCAAAGGGCAGCCCCGAGAACCACAGGTGT
ACACCCTGCCCCCATCCCGGGAGGAGATGACCAA
GAACCAGGTCAGCCTGACCTGCCTGGTCAAAGGC
TTCTACCCCAGCGACATCGCCGTGGAGTGGGAGA
GCAATGGGCAGCCGGAGAACAACTACAAGACCA
CACCTCCCATGCTGGACTCCGACGGCTQCTTCTTC
CTCTACAGCAAGCTCACCGTGGACAAGAGCAGGT
GGCAGCAGGGGAACGTCTTCTCATGCTCCGTGAT
GCATGAGGCTCTGCACAACCACTACACGCAGAAG
AGCCTCTCCCTGTCTCCGGGTAAATGA DESCRIPTION: SEQUENCE (signal sequence underlined):
Heavy Chain MEFGLSWVFLVALLRGVOCQVOLVESGGGVVQPG Protein Sequence RSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVA
VISKDGGNKYHADSVKGRFTISRDNSKNALYLQMN
SLRVEDTAVYYCVRRGHQLVLGYYYYNGLDVWG
QGTTVTVSSASTKGPSVFPLAPCSRSTSESTAALGCL
VKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLY
SLSSVVTVPSSNFGTQTYTCNVDHKPSNTKVDKTV
ERKCCVECPPCPAPPVAGPSVFLFPPKPKDTLMISRT
PEVTCVVVDVSHEDPEVQ-3TSFWYVDGVEVHNAKTK
PREEQFNSTFRVVSVLTVVHQDWLNGKEYKCKVS
NKGLPAPIEKTISKTKGQPREPQVYTLPPSREEMTK
NQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTP
PMLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHE
ALHNHYTQKSLSLSPGK
Light Chain DNA ATGAGGCTCCCTGCTCAGCTCCTGGGGCTGCTAA Sequence TGCTCTGGGTCTCTGGATCCAGTGGGGATATTGT
GCTGACTCAGTCTCCACTCTCCCTGCCCGTCACCC
CTGGAGAGCCGGCCTCCATCTCCTGCAGGTCTAG
TCAGAGCCTCTTGTATAGTAATGGATACAACTTTT
TGGATTGGTACCTGCAGAAGCCAGGGCAGTCTCC
ACAGCTCCTGATCTATTTGGGTTCTAATCGGGCCT
CCGGGGTCCCTGACAGGTTCAGTGGCAGTGGATC
AGGCACAGATTTTACACTGAAAATCAGCAGATTG
GAGGCTGAGGATGTTGGGGTTTATTACTGCATGC
AAGCTCTACAAACTCCTCGGACGTTCGGCCAAGG
GACCAAGGTGGAAATCAAACGAACTGTGGCTGC
ACCATCTGTCTTCATCTTCCCGCCATCTGATGAGC
AGTTGAAATCTGGAACTGCCTCTGTTGTGTGCCT
GCTGAATAACTTCTATCCCAGAGAGGCCAAAGTA
CAGTGGAAGGTGGATAACGCCCTCCAATCGGGTA
ACTCCCAGGAGAGTGTCACAGAGCAGGACAGCA
AGGACAGCACCTACAGCCTCAGCAGCACCCTGAC
GCTGAGCAAAGCAGACTACGAGAAACACAAAGT
CTACGCCTGCGAAGTCACCCATCAGGGCCTGAGC
TCGCCCGTCACAAAGAGCTTCAACAGGGGAGAGT
GTTAG
Light Chain MRLPAOLLGLLMLWVSGSSGD-VLTOSPLSLPVTPG Protein Sequence EPASISCRSSQSLLYSNGYNFLDWYLQKPGQSPQLLI
YLGSNRASGNPDRFSGSGSGTDFTLKISRLEAEDVG
VYYCMQALQTPRTFGQGTKVE-KRTVAAPSVFIFPP
SDEQLKSGTASNVCLLΝΝFYPREAKVQWKVDΝAL
QSGΝSQESVTEQDSKDSTYSLSSTLTLSKADYEKHK
VYACEVTHQGLSSPVTKSFΝRGEC
Figure imgf000085_0001
Figure imgf000086_0001
Table 4: DNA and protein sequences of antibody 7.1.2
DESCRIPTION: SEQUENCE (signal sequence underlined):
Heavy Chain ATGGAGTTTGGGCTGAGCTGGGTTTTCCTCGTTGC DNA Sequence TCTTTTAAGAGGTGTCCAGTGTCAGGTGCAGCTG
GTGGAGTCTGGGGGAGGCGTGGTCCAGCCTGGG
AGGTCCCTGAGACTCTCCTGTGCAGCCTCTGGAT
TCACCTTCAGTAGCTATGGCATGCACTGGGTCCG
CCAGGCTCCAGGCAAGGGGCTGGAGTGGGTGGC
AGTTATATCAAATGATGGAGATAATAAATACCAT
GCAGACTCCGTGTGGGGCCGATTCACCATCTCCA
GAGACAATTCCAGGAGCACGCTTTATCTGCAAAT
GAACAGCCTGAGAGCTGAGGACACGGCTGTATAT
TACTGTGCGAGAAGAGGCATGGGGTCTAGTGGG
AGCCGTGGGGATTACTACTACTACTACGGTTTGG
ACGTCTGGGGCCAAGGGACCACGGTCACCGTCTC
CTCAGCCTCCACCAAGGGCCCATCGGTCTTCCCC
CTGGCGCCCTGCTCCAGGAGCACCTCCGAGAGCA
CAGCGGCCCTGGGCTGCCTGGTCAAGGACTACTT
CCCCGAACCGGTGACGGTGTCGTGGAACTCAGGC
GCTCTGACCAGCGGCGTGCACACCTTCCCAGCTG
TCCTACAGTCCTCAGGACTCTACTCCCTCAGCAG
CGTGGTGACCGTGCCCTCCAGCAACTTCGGCACC
CAGACCTACACCTGCAACGTAGATCACAAGCCCA
GCAACACCAAGGTGGACAAGACAGTTGAGCGCA
AATGTTGTGTCGAGTGCCCACCGTGCCCAGCACC
ACCTGTGGCAGGACCGTCAGTCTTCCTCTTCCCCC
CAAAACCCAAGGACACCCTCATGATCTCCCGGAC
CCCTGAGGTCACGTGCGTGGTGGTGGACGTGAGC
CACGAAGACCCCGAGGTCCAGTTCAACTGGTACG
TGGACGGCGTGGAGGTGCATAATGCCAAGACAA
AGCCACGGGAGGAGCAGTTCAACAGCACGTTCC
GTGTGGTCAGCGTCCTCACCGTTGTGCACCAGGA
CTGGCTGAACGGCAAGGAGTACAAGTGCAAGGT
CTCCAACAAAGGCCTCCCAGCCCCCATCGAGAAA
ACCATCTCCAAAACCAAAGGGCAGCCCCGAGAA
CCACAGGTGTACACCCTGCCCCCATCCCGGGAGG
AGATGACCAAGAACCAGGTCAGCCTGACCTGCCT
GGTCAAAGGCTTCTACCCCAGCGACATCGCCGTG
GAGTGGGAGAGCAATGGGCAGCCGGAGAACAAC
TACAAGACCACACCTCCCATGCTGGACTCCGACG
GCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGAC
AAGAGCAGGTGGCAGCAGGGGAACGTCTTCTCAT
GCTCCGTGATGCATGAGGCTCTGCACAACCACTA
CACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA
TGA
Figure imgf000088_0001
Figure imgf000089_0001
Table 5: DNA and protein sequences of antibody 10.8.3
DESCRIPTION: SEQUENCE (signal sequence underlined):
Heavy Chain ATGAAACACCTGTGGTTCTTCCTCCTGCTGGTGGC DNA Sequence AGCTCCCAGATGGGTCCTGTCCCAGGTGCAGCTG
CAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCGG
AGACCCTGTCCCTCACCTGCACTGTCTCTGGTGGC
TCCATCAGTAGTTACTACTGGATCTGGATCCGGC
AGCCCGCCGGGAAGGGACTGGAATGGATTGGGC
GTGTCTATACCAGTGGGAGCACCAACTACAACCC
CTCCCTCAAGAGTCGAGTCACCATGTCAGTAGAC
ACGTCCAAGAACCAGTTCTCCCTGAAGCTGAGCT
CTGTGACCGCCGCGGACACGGCCGTGTATTACTG
TGCGAGAGATGGTCTTTACAGGGGGTACGGTATG
GACGTCTGGGGCCAAGGGACCACGGTCACCGTCT
CCTCAGCCTCCACCAAGGGCCCATCGGTCTTCCC
CCTGGCGCCCTGCTCCAGGAGCACCTCCGAGAGC
ACAGCGGCCCTGGGCTGCCTGGTCAAGGACTACT
TCCCCGAACCGGTGACGGTGTCGTGGAACTCAGG
CGCTCTGACCAGCGGCGTGCACACCTTCCCAGCT
GTCCTACAGTCCTCAGGACTCTACTCCCTCAGCA
GCGTGGTGACCGTGCCCTCCAGCAACTTCGGCAC
CCAGACCTACACCTGCAACGTAGATCACAAGCCC
AGCAACACCAAGGTGGACAAGACAGTTGAGCGC
AAATGTTGTGTCGAGTGCCCACCGTGCCCAGCAC
CACCTGTGGCAGGACCGTCAGTCTTCCTCTTCCCC
CCAAAACCCAAGGACACCCTCATGATCTCCCGGA
CCCCTGAGGTCACGTGCGTGGTGGTGGACGTGAG
CCACGAAGACCCCGAGGTCCAGTTCAACTGGTAC
GTGGACGGCGTGGAGGTGCATAATGCCAAGACA
AAGCCACGGGAGGAGCAGTTCAACAGCACGTTC
CGTGTGGTCAGCGTCCTCACCGTTGTGCACCAGG
ACTGGCTGAACGGCAAGGAGTACAAGTGCAAGG
TCTCCAACAAAGGCCTCCCAGCCCCCATCGAGAA
AACCATCTCCAAAACCAAAGGGCAGCCCCGAGA
ACCACAGGTGTACACCCTGCCCCCATCCCGGGAG
GAGATGACCAAGAACCAGGTCAGCCTGACCTGCC
TGGTCAAAGGCTTCTACCCCAGCGACATCGCCGT
GGAGTGGGAGAGCAATGGGCAGCCGGAGAACAA
CTACAAGACCACACCTCCCATGCTGGACTCCGAC
GGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGG
ACAAGAGCAGGTGGCAGCAGGGGAACGTCTTCT
CATGCTCCGTGATGCATGAGGCTCTGCACAACCA
CTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGT
AAATGA
Figure imgf000091_0001
Figure imgf000092_0001
Table 6: DNA and protein sequences of antibody 15.1.1
DESCRIPTION: SEQUENCE (signal sequence underlined):
Heavy Chain ATGAAACATCTGTGGTTCTTCCTTCTCCTGGTGGC DNA Sequence AGCTCCCAGATGGGTCCTGTCCCAGGTGCAGCTG
CAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCGG
AGACCCTGTCCCTCACCTGCACTGTCTCTGGTGGC
TCCATCAGAAGTTACTACTGGACCTGGATCCGGC
AGCCCCCAGGGAAGGGACTGGAGTGGATTGGAT
ATATCTATTACAGTGGGAGCACCAACTACAATCC
CTCCCTCAAGAGTCGAGTCACCATATCAGTAGAC
ATGTCCAAGAACCAGTTCTCCCTGAAGCTGAGTT
CTGTGACCGCTGCGGACACGGCCGTTTATTACTG
TGCGAGAAAGGGTGACTACGGTGGTAATTTTAAC
TACTTTCACCAGTGGGGCCAGGGAACCCTGGTCA
CCGTCTCCTCAGCCTCCACCAAGGGCCCATCGGT
CTTCCCCCTGGCGCCCTGCTCCAGGAGCACCTCC
GAGAGCACAGCGGCCCTGGGCTGCCTGGTCAAG
GACTACTTCCCCGAACCGGTGACGGTGTCGTGGA
ACTCAGGCGCTCTGACCAGCGGCGTGCACACCTT
CCCAGCTGTCCTACAGTCCTCAGGACTCTACTCCC
TCAGCAGCGTGGTGACCGTGCCCTCCAGCAACTT
CGGCACCCAGACCTACACCTGCAACGTAGATCAC
AAGCCCAGCAACACCAAGGTGGACAAGACAGTT
GAGCGCAAATGTTGTGTCGAGTGCCCACCGTGCC
CAGCACCACCTGTGGCAGGACCGTCAGTCTTCCT
CTTCCCCCCAAAACCCAAGGACACCCTCATGATC
TCCCGGACCCCTGAGGTCACGTGCGTGGTGGTGG
ACGTGAGTCACGAAGACCCCGAGGTCCAGTTCAA
CTGGTACGTGGACGGCGTGGAGGTGCATAATGCC
AAGACAAAGCCACGGGAGGAGCAGTTCAACAGC
ACGTTCCGTGTGGTCAGCGTCCTCACCGTTGTGC
ACCAGGACTGGCTGAACGGCAAGGAGTACAAGT
GCAAGGTCTCCAACAAAGGCCTCCCAGCCCCCAT
CGAGAAAACCATCTCCAAAACCAAAGGGCAGCC
CCGAGAACCACAGGTGTACACCCTGCCCCCATCC
CGGGAGGAGATGACCAAGAACCAGGTCAGCCTG
ACCTGCCTGGTCAAAGGCTTCTACCCCAGCGACA
TCGCCGTGGAGTGGGAGAGCAATGGGCAGCCGG
AGAACAACTACAAGACCACACCTCCCATGCTGGA
CTCCGACGGCTCCTTCTTCCTCTACAGCAAGCTCA
CCGTGGACAAGAGCAGGTGGCAGCAGGGGAACG
TCTTCTCATGCTCCGTGATGCATGAGGCTCTGCAC
AACCACTACACGCAGAAGAGCCTCTCCCTGTCTC
CGGGTAAATGA DESCRIPTION: SEQUENCE (signal sequence underlined):
Heavy Chain MKHLWFFLLLVAAPRWNLSOVOLOESGPGLVKPSE Protein Sequence TLSLTCTVSGGSIRSYYWTWIRQPPGKGLEWIGY-Y
YSGSTΝYΝPSLKSRVTISVDMSKΝQFSLKLSSVTAA
DTAVYYCARKGDYGGΝFΝYFHQWGQGTLVTVSS
ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPV
TVSWΝSGALTSGVHTFPAVLQSSGLYSLSSWTVPS
SΝFGTQTYTCΝVDHKPSΝTKVDKTVERKCCVECPP
CPAPPVAGPSVFLFPPKPKDTLMISRTPEVTCVWD
NSHEDPENQFΝWYNDGNENHΝAKTKPREEQFΝSTF
RNNSNLTNNHQDWLΝGKEYKCKNSΝKGLPAPIEK
TISKTKGQPREPQVYTLPPSREEMTKΝQVSLTCLVK
GFYPSDIAVEWESΝGQPEΝΝYKTTPPMLDSDGSFFL
YSKLTVDKSRWQQGΝVFSCSVMHEALHΝHYTQKS
LSLSPGK
Light Chain DΝA ATGAGGCTCCCTGCTCAGCTCCTGGGGCTGCTAA Sequence TGCTCTGGGTCTCTGGATCCAGTGGGGATATTGT
GATGACTCAGTCTCCACTCTCCCTGCCCGTCACCC
CTGGAGAGCCGGCCTCCATCTCCTGCAGGTCTAG
TCAGAGCCTCCTACATACTAATGGATACAACTAT
TTCGATTGGTACCTGCAGAAGCCAGGGCAGTCTC
CACAACTCCTGATCTATTTGGGTTCTAATCGGGCC
TCCGGGGTCCCTGACAGGTTCAGTGGCAGTGGAT
CAGGCACAGATTTTACACTGAAAATCAGCAGAGT
GGAGGCTGAGGATGTTGGGGTTTATTACTGCATG
CAAGCTCTACAAACTCCGTACAGTTTTGGCCAGG
GGACCAAGCTGGAGATCAAACGAACTGTGGCTG
CACCATCTGTCTTCATCTTCCCGCCATCTGATGAG
CAGTTGAAATCTGGAACTGCCTCTGTTGTGTGCCT
GCTGAATAACTTCTATCCCAGAGAGGCCAAAGTA
CAGTGGAAGGTGGATAACGCCCTCCAATCGGGTA
ACTCCCAGGAGAGTGTCACAGAGCAGGACAGCA
AGGACAGCACCTACAGCCTCAGCAGCACCCTGAC
GCTGAGCAAAGCAGACTACGAGAAACACAAAGT
CTACGCCTGCGAAGTCACCCATCAGGGCCTGAGC
TCGCCCGTCACAAAGAGCTTCAACAGGGGAGAGT
GTTAG
Light Chain MRLPAOLLGLLMLWVSGSSGDINMTOSPLSLPVTP Protein Sequence GEPASISCRSSQSLLHTΝGYΝYFDWYLQKPGQSPQL
LIYLGSΝRASGVPDRFSGSGSGTDFTLKISRVEAEDV
GVYYCMQALQTPYSFGQGTKLEIKRTVAAPSVFIFP
PSDEQLKSGTASVVCLLΝΝFYPREAKVQWKVDΝA
LQSGΝSQESNTEQDSKDSTYSLSSTLTLSKADYEKH
KVYACEVTHQGLSSPVTKSFΝRGEC
Figure imgf000095_0001
Table 7: DNA and protein sequences of antibody 21.4.1
DESCRIPTION: SEQUENCE (signal sequence underlined):
Heavy Chain ATGGACTGGACCTGGAGGATCCTCTTCTTGGTGG DNA Sequence CAGCAGCCACAGGAGCCCACTCCCAGGTGCAGCT
GGTGCAGTCTGGGGCTGAGGTGAAGAAGCCTGG
GGCCTCAGTGAAGGTCTCCTGCAAGGCTTCTGGA
TACACCTTCACCGGCTACTATATGCACTGGGTGC
GACAGGCCCCTGGACAAGGGCTTGAGTGGATGG
GATGGATCAACCCTGACAGTGGTGGCACAAACTA
TGCACAGAAGTTTCAGGGCAGGGTCACCATGACC
AGGGACACGTCCATCAGCACAGCCTACATGGAGC
TGAACAGGCTGAGATCTGACGACACGGCCGTGTA
TTACTGTGCGAGAGATCAGCCCCTAGGATATTGT
ACTAATGGTGTATGCTCCTACTTTGACTACTGGG
GCCAGGGAACCCTGGTCACCGTCTCCTCAGCCTC
CACCAAGGGCCCATCGGTCTTCCCCCTGGCGCCC
TGCTCCAGGAGCACCTCCGAGAGCACAGCGGCCC
TGGGCTGCCTGGTCAAGGACTACTTCCCCGAACC
GGTGACGGTGTCGTGGAACTCAGGCGCTCTGACC
AGCGGCGTGCACACCTTCCCAGCTGTCCTACAGT
CCTCAGGACTCTACTCCCTCAGCAGCGTGGTGAC
CGTGCCCTCCAGCAACTTCGGCACCCAGACCTAC
ACCTGCAACGTAGATCACAAGCCCAGCAACACCA
AGGTGGACAAGACAGTTGAGCGCAAATGTTGTGT
CGAGTGCCCACCGTGCCCAGCACCACCTGTGGCA
GGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCA
AGGACACCCTCATGATCTCCCGGACCCCTGAGGT
CACGTGCGTGGTGGTGGACGTGAGCCACGAAGA
CCCCGAGGTCCAGTTCAACTGGTACGTGGACGGC
GTGGAGGTGCATAATGCCAAGACAAAGCCACGG
GAGGAGCAGTTCAACAGCACGTTCCGTGTGGTCA
GCGTCCTCACCGTTGTGCACCAGGACTGGCTGAA
CGGCAAGGAGTACAAGTGCAAGGTCTCCAACAA
AGGCCTCCCAGCCCCCATCGAGAAAACCATCTCC
AAAACCAAAGGGCAGCCCCGAGAACCACAGGTG
TACACCCTGCCCCCATCCCGGGAGGAGATGACCA
AGAACCAGGTCAGCCTGACCTGCCTGGTCAAAGG
CTTCTACCCCAGCGACATCGCCGTGGAGTGGGAG
AGCAATGGGCAGCCGGAGAACAACTACAAGACC
ACACCTCCCATGCTGGACTCCGACGGCTCCTTCTT
CCTCTACAGCAAGCTCACCGTGGACAAGAGCAGG
TGGCAGCAGGGGAACGTCTTCTCATGCTCCGTGA
TGCATGAGGCTCTGCACAACCACTACACGCAGAA
GAGCCTCTCCCTGTCTCCGGGTAAATGA
Figure imgf000097_0001
Figure imgf000098_0001
Table 8: DNA and protein sequences of mature variable domains of 21.2.1 antibody
Figure imgf000099_0001
Table 9: DNA and protein sequences of mature variable domains of 22.1.1 antibody
Figure imgf000100_0001
Table 10: DNA and protein sequences of mature variable domains of 23.5.1 antibody
Figure imgf000101_0001
Table 11 : DNA and protein sequences of mature variable domains of 23.28.1 antibody
Figure imgf000102_0001
Figure imgf000103_0001
Table 12: DNA and protein sequences of mature variable domains of 23.29.1 antibody
Figure imgf000103_0002
Table 13: DNA and protein sequences of mature variable domains of 24.2.1 antibody
Figure imgf000104_0001
Table 14: DNA and protein sequences of antibody 21.2.1
DESCRIPTION: SEQUENCE (signal sequence underlined):
Heavy Chain ATGGAGTTTGGGCTGAGCTGGGTTTTCCTCGTTGC DNA TCTTTTAAGAGGTGTCCAGTGTCAGGTGCAGCTG
GTGGAGTCTGGGGGAGGCGTGGTCCAGCCTGGG
AGGTCCCTGAGACTCTCCTGTGCAGCCTCTGGAT
TCACCTTCAGTAGCTATGTCATGCACTGGGTCCG
CCAGGCTCCAGGCAAGGGGCTGGAGTGGGTGGC
AGTTATGTCATATGATGGAAGTAGTAAATACTAT
GCAAACTCCGTGAAGGGCCGATTCACCATCTCCA
GAGACAATTCCAAGAACACGCTGTATCTGCAAAT
AAACAGCCTGAGAGCTGAGGACACGGCTGTGTAT
TACTGTGCGAGAGATGGGGGTAAAGCAGTGCCTG
GTCCTGACTACTGGGGCCAGGGAATCCTGGTCAC
CGTCTCCTCAGCCTCCACCAAGGGCCCATCGGTC
TTCCCCCTGGCGCCCTGCTCCAGGAGCACCTCCG
AGAGCACAGCGGCCCTGGGCTGCCTGGTCAAGG
ACTACTTCCCCGAACCGGTGACGGTGTCGTGGAA
CTCAGGCGCTCTGACCAGCGGCGTGCACACCTTC
CCAGCTGTCCTACAGTCCTCAGGACTCTACTCCCT
CAGCAGCGTGGTGACCGTGCCCTCCAGCAACTTC
GGCACCCAGACCTACACCTGCAACGTAGATCACA
AGCCCAGCAACACCAAGGTGGACAAGACAGTTG
AGCGCAAATGTTGTGTCGAGTGCCCACCGTGCCC
AGCACCACCTGTGGCAGGACCGTCAGTCTTCCTC
TTCCCCCCAAAACCCAAGGACACCCTCATGATCT
CCCGGACCCCTGAGGTCACGTGCGTGGTGGTGGA
CGTGAGCCACGAAGACCCCGAGGTCCAGTTCAAC
TGGTACGTGGACGGCGTGGAGGTGCATAATGCCA
AGACAAAGCCACGGGAGGAGCAGTTCAACAGCA
CGTTCCGTGTGGTCAGCGTCCTCACCGTTGTGCAC
CAGGACTGGCTGAACGGCAAGGAGTACAAGTGC
AAGGTCTCCAACAAAGGCCTCCCAGCCCCCATCG
AGAAAACCATCTCCAAAACCAAAGGGCAGCCCC
GAGAACCACAGGTGTACACCCTGCCCCCATCCCG
GGAGGAGATGACCAAGAACCAGGTCAGCCTGAC
CTGCCTGGTCAAAGGCTTCTACCCCAGCGACATC
GCCGTGGAGTGGGAGAGCAATGGGCAGCCGGAG
AACAACTACAAGACCACACCTCCCATGCTGGACT
CCGACGGCTCCTTCTTCCTCTACAGCAA'GCTCACC
GTGGACAAGAGCAGGTGGCAGCAGGGGAACGTC
TTCTCATGCTCCGTGATGCATGAGGCTCTGCACA
ACCACTACACGCAGAAGAGCCTCTCCCTGTCTCC
GGGTAAATGA
Figure imgf000106_0001
Table 15: DNA and protein sequences of antibody 22.1.1
DESCRIPTION: SEQUENCE (signal sequence underlined):
Heavy Chain ATGGAGTTTGGGCTGAGCTGGGTTTTCCTCGTTGC DNA TCTTTTAAGAGGTGTCCAGTGTCAGGTGCAACTG
GTGGAGTCTGGGGGAGGCGTGGTCCAGCCTGGG
AGGTCCCTGAGACTCTCCTGTGCAGCCTCTGGAT
TCACCTTCAGTCGCTATGGCATGCACTGGGTCCG
CCAGGCTCCAGGCAAGGGGCTGGAGTGGGTGGC
AGTTATATCATCTGATGGAGGTAATAAATACTAT
GCAGACTCCGTGAAGGGCCGATTCACCATCTCCA
GAGACAATTCCAAGAACACGCTGTATCTGCAAAT
GAACAGCCTGAGAGCTGAGGACACGGCTGTGTAT
TACTGTACGAGAAGAGGGACTGGAAAGACTTACT
ACCACTACTGTGGTATGGACGTCTGGGGCCAAGG
GACCACGGTCACCGTCTCCTCAGCCTCCACCAAG
GGCCCATCGGTCTTCCCCCTGGCGCCCTGCTCCA
GGAGCACCTCCGAGAGCACAGCGGCCCTGGGCT
GCCTGGTCAAGGACTACTTCCCCGAACCGGTGAC
GGTGTCGTGGAACTCAGGCGCTCTGACCAGCGGC
GTGCACACCTTCCCAGCTGTCCTACAGTCCTCAG
GACTCTACTCCCTCAGCAGCGTGGTGACCGTGCC
CTCCAGCAACTTCGGCACCCAGACCTACACCTGC
AACGTAGATCACAAGCCCAGCAACACCAAGGTG
GACAAGACAGTTGAGCGCAAATGTTGTGTCGAGT
GCCCACCGTGCCCAGCACCACCTGTGGCAGGACC
GTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGAC
ACCCTCATGATCTCCCGGACCCCTGAGGTCACGT
GCGTGGTGGTGGACGTGAGCCACGAAGACCCCG
AGGTCCAGTTCAACTGGTACGTGGACGGCGTGGA
GGTGCATAATGCCAAGACAAAGCCACGGGAGGA
GCAGTTCAACAGCACGTTCCGTGTGGTCAGCGTC
CTCACCGTTGTGCACCAGGACTGGCTGAACGGCA
AGGAGTACAAGTGCAAGGTCTCCAACAAAGGCC
TCCCAGCCCCCATCGAGAAAACCATCTCCAAAAC
CAAAGGGCAGCCCCGAGAACCACAGGTGTACAC
CCTGCCCCCATCCCGGGAGGAGATGACCAAGAAC
CAGGTCAGCCTGACCTGCCTGGTCAAAGGCTTCT
ACCCCAGCGACATCGCCGTGGAGTGGGAGAGCA
ATGGGCAGCCGGAGAACAACTACAAGACCACAC
CTCCCATGCTGGACTCCGACGGCTCCTTCTTCCTC
TACAGCAAGCTCACCGTGGACAAGAGCAGGTGG
CAGCAGGGGAACGTCTTCTCATGCTCCGTGATGC
ATGAGGCTCTGCACAACCACTACACGCAGAAGA
GCCTCTCCCTGTCTCCGGGTAAATGA DESCRIPTION: SEQUENCE (signal sequence underlined):
Heavy Chain MEFGLSWNFLVALLRGVOCOVOLVESGGGVVOPG Protein RSLRLSCAASGFTFSRYGMHWNRQAPGKGLEWNA
NISSDGGΝKYYADSNKGRFTISRDΝSKΝTLYLQMΝ
SLRAEDTANYYCTRRGTGKTYYHYCGMDNWGQG
TTVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVK
DYFPEPVTVSWΝSGALTSGVHTFPANLQSSGLYSLS
SVVTVPSSΝFGTQTYTCΝNDHKPSΝTKVDKTVERK
CCVECPPCPAPPVAGPSVFLFPPKPKDTLMISRTPEV
TCVVVDVSHEDPEVQFΗWYVDGVEVHΝAKTKPRE
EQFΝSTFRVVSVLTVVHQDWLΝGKEYKCKVSΝKG
LPAPIEKTISKTKGQPREPQVYTLPPSREEMTKΝQVS
LTCLVKGFYPSDIAVEWESΝGQPEΝΝYKTTPPMLD
SDGSFFLYSKLTVDKSRWQQGΝVFSCSVMHEALHΝ
HYTQKSLSLSPGK
Light Chain DΝA ATGAGGCTCCCTGCTCAGCTCCTGGGGCTGCTAA
TGCTCTGGGTCTCTGGATCCAGTGGGGATATTGT
GATGACTCAGTCTCCACTCTCCCTGCCCGTCACCC
CTGGAGAGCCGGCCTCCATCTCCTGCAGGTCTAG
TCAGAGCCTCCTGTATAGTAATGGATATAACTAT
TTGGATTGGTACCTGCAGAAGCCAGGGCAGTCTC
CACACCTCCTGATCTATTTGGGTTCTAATCGGGCC
TCCGGGGTCCCTGACAGGTTCAGTGGCAGTGGTT
CAGGCACTGATTTTACACTGAAAATCAGCAGAGT
GGAGGCTGAGGATGTTGGGGTTTATTACTGCATG
CAAGCTCTACAAACTCCTCGGACGTTCGGCCAAG
GGACCAAGGTGGAAATCAAACGAACTGTGGCTG
CACCATCTGTCTTCATCTTCCCGCCATCTGATGAG
CAGTTGAAATCTGGAACTGCCTCTGTTGTGTGCCT
GCTGAATAACTTCTATCCCAGAGAGGCCAAAGTA
CAGTGGAAGGTGGATAACGCCCTCCAATCGGGTA
ACTCCCAGGAGAGTGTCACAGAGCAGGACAGCA
AGGACAGCACCTACAGCCTCAGCAGCACCYTGAC
GCTGAGCAAAGCAGACTACGAGAAACACAAAGT
CTACGCCTGCGAAGTCACCCATCAGGGCCTGAGC
TCGCCCGTCACAAAGAGCTTCAACAGGGGAGAGT
GTTAG
Light Chain MRLPAOLLGLLMLWVSGSSGDIVMTOSPLSLPVTP Protein GEPASISCRSSQSLLYSΝGYΝYLDWYLQKPGQSPHL
LIYLGSΝRASGNPDRFSGSGSGTDFTLKISRNEAEDN
GNYYCMQALQTPRTFGQGTKNEIKRTNAAPSNFIFP
PSDEQLKSGTASNNCLLΝΝFYPREAKNQWKNDΝA
LQSGΝSQESVTEQDSKDSTYSLSSTLTLSKADYEKH
KVYACEVTHQGLSSPVTKSFΝRGEC Table 16: DNA and protein sequences of antibody 23.5.1
DESCRIPTION: SEQUENCE (signal sequence underlined):
Heavy Chain ATGGAGTTTGGGCTGAGCTGGGTTTTCCTCGTTGC DNA TCTTTTAAGAGGTGTCCAGTGTCAGGTGCAGCTG
GTGGAGTCTGGGGGAGGCGTGGTCCAGCCTGGG
AGGTCCCTGAGACTCTCCTGTGTAGCCTCTGGATT
CACCTTCAGTAACTATGGCATGCACTGGGTCCGC
CAGGCTCCAGGCAAGGGGCTGGAGTGGGTGGCA
ATTATATCATATGATGGAAGTAATAAATACTATG
CAGACTCCGTGAAGGGCCGATTCACCATCTCCAG
AGACAATTCCAAGAACACGCTGTATGTGCAAATG
AACAGCCTGAGAGCTGAGGACACGGCTGTGTATT
ACTGTGCGAGACGCGGTCACTACGGGAGGGATTA
CTACTCCTACTACGGTTTGGACGTCTGGGGCCAA
GGGACCACGGTCACCGTCTCCTCAGCCTCCACCA
AGGGCCCATCGGTCTTCCCCCTGGCGCCCTGCTC
CAGGAGCACCTCCGAGAGCACAGCGGCCCTGGG
CTGCCTGGTCAAGGACTACTTCCCCGAACCGGTG
ACGGTGTCGTGGAACTCAGGCGCTCTGACCAGCG
GCGTGCACACCTTCCCAGCTGTCCTACAGTCCTC
AGGACTCTACTCCCTCAGCAGCGTGGTGACCGTG
CCCTCCAGCAACTTCGGCACCCAGACCTACACCT
GCAACGTAGATCACAAGCCCAGCAACACCAAGG
TGGACAAGACAGTTGAGCGCAAATGTTGTGTCGA
GTGCCCACCGTGCCCAGCACCACCTGTGGCAGGA
CCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGG
ACACCCTCATGATCTCCCGGACCCCTGAGGTCAC
GTGCGTGGTGGTGGACGTGAGCCACGAAGACCCC
GAGGTCCAGTTCAACTGGTACGTGGACGGCGTGG
AGGTGCATAATGCCAAGACAAAGCCACGGGAGG
AGCAGTTCAACAGCACGTTCCGTGTGGTCAGCGT
CCTCACCGTTGTGCACCAGGACTGGCTGAACGGC
AAGGAGTACAAGTGCAAGGTCTCCAACAAAGGC
CTCCCAGCCCCCATCGAGAAAACCATCTCCAAAA
CCAAAGGGCAGCCCCGAGAACCACAGGTGTACA
CCCTGCCCCCATCCCGGGAGGAGATGACCAAGAA
CCAGGTCAGCCTGACCTGCCTGGTCAAAGGCTTC
TACCCCAGCGACATCGCCGTGGAGTGGGAGAGC
AATGGGCAGCCGGAGAACAACTACAAGACCACA
CCTCCCATGCTGGACTCCGACGGCTCCTTCTTCCT
CTACAGCAAGCTCACCGTGGACAAGAGCAGGTG
GCAGCAGGGGAACGTCTTCTCATGCTCCGTGATG
CATGAGGCTCTGCACAACCACTACACGCAGAAGA
GCCTCTCCCTGTCTCCGGGTAAATGA
Figure imgf000110_0001
Table 17: DNA and protein sequences of antibody 23.28.1
DESCRIPTION: SEQUENCE (signal sequence underlined):
Heavy Chain ATGAAACATCTGTGGTTCTTCCTTCTCCTGGTGGC DNA AGCTCCCAGATGGGTCCTGTCCCAGGTGCAGCTG
CAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCGG
AGACCCTGTCCCTCACCTGCACTGTCTCTGGTGGC
TCCATCAGAGGTTACTACTGGAGCTGGATCCGGC
AGCCCCCTGGGAAGGGACTGGAGTGGATTGGGT
ATATCTATTACAGTGGGAGCACCAACTACAACCC
CTCCCTCAAGAGTCGAGTCACCATATCAGTAGAC
ACGTCCAAGAACCAGTTCTCCCTGAAGCTGAACT
CTGTGACCGCTGCGGACACGGCCGTGTATTATTG
TGCGAGAAAGGGGGGCCTCTACGGTGACTACGG
CTGGTTCGCCCCCTGGGGCCAGGGAACCCTGGTC
ACCGTCTCCTCAGCCTCCACCAAGGGCCCATCGG
TCTTCCCCCTGGCGCCCTGCTCCAGGAGCACCTCC
GAGAGCACAGCGGCCCTGGGCTGCCTGGTCAAG
GACTACTTCCCCGAACCGGTGACGGTGTCGTGGA
ACTCAGGCGCTCTGACCAGCGGCGTGCACACCTT
CCCAGCTGTCCTACAGTCCTCAGGACTCTACTCCC
TCAGCAGCGTGGTGACCGTGCCCTCCAGCAACTT
CGGCACCCAGACCTACACCTGCAACGTAGATCAC
AAGCCCAGCAACACCAAGGTGGACAAGACAGTT
GAGCGCAAATGTTGTGTCGAGTGCCCACCGTGCC
CAGCACCACCTGTGGCAGGACCGTCAGTCTTCCT
CTTCCCCCCAAAACCCAAGGACACCCTCATGATC
TCCCGGACCCCTGAGGTCACGTGCGTGGTGGTGG
ACGTGAGCCACGAAGACCCCGAGGTCCAGTTCAA
CTGGTACGTGGACGGCGTGGAGGTGCATAATGCC
AAGACAAAGCCACGGGAGGAGCAGTTCAACAGC
ACGTTCCGTGTGGTCAGCGTCCTCACCGTTGTGC
ACCAGGACTGGCTGAACGGCAAGGAGTACAAGT
GCAAGGTCTCCAACAAAGGCCTCCCAGCCCCCAT
CGAGAAAACCATCTCCAAAACCAAAGGGCAGCC
CCGAGAACCACAGGTGTACACCCTGCCCCCATCC
CGGGAGGAGATGACCAAGAACCAGGTCAGCCTG
ACCTGCCTGGTCAAAGGCTTCTACCCCAGCGACA
TCGCCGTGGAGTGGGAGAGCAATGGGCAGCCGG
AGAACAACTACAAGACCACACCTCCCATGCTGGA
CTCCGACGGCTCCTTCTTCCTCTACAGCAAGCTCA
CCGTGGACAAGAGCAGGTGGCAGCAGGGGAACG
TCTTCTCATGCTCCGTGATGCATGAGGCTCTGCAC
AACCACTACACGCAGAAGAGCCTCTCCCTGTCTC
CGGGTAAATGA - Ill
Figure imgf000112_0001
Table 18: DNA and protein sequences of antibody 23.29.1
DESCRIPTION: SEQUENCE (signal sequence underlined):
Heavy Chain ATGGAGTTTGGGCTGAGCTGGGTTTTCCTCGTTGC DNA TCTTTTAAGAGGTGTCCAGTGTCAGGTGCAACTG
GTGGAGTCTGGGGGAGGCGTGGTCCAGCCTGGG
AGGTCCCTGAGACTCTCCTGTGCAGCCTCTGGAT
TCACCTTCAGTAGCTATGCCATGCACTGGGTCCG
CCAGGCTCCAGGCAAGGGGCTGGAGTGGGTGGC
AGTTATATCATATGATGGAAGTAATAAATACTAT
GCAGACTCCGTGAAGGGCCGATTCACCATCTACA
GAGACAATTCCAAGAACACGCTGTATCTGCAAAT
GAACAGCCTGAGAGCTGAGGACACGGCTGTGTAT
TACTGTGCGAGACGCGGTCACTACGGGAATAATT
ACTACTCCTATTACGGTTTGGACGTCTGGGGCCA
AGGGACCACGGTCACCGTCTCCTCAGCCTCCACC
AAGGGCCCATCGGTCTTCCCCCTGGCGCCCTGCT
CCAGGAGCACCTCCGAGAGCACAGCGGCCCTGG
GCTGCCTGGTCAAGGACTACTTCCCCGAACCGGT
GACGGTGTCGTGGAACTCAGGCGCTCTGACCAGC
GGCGTGCACACCTTCCCAGCTGTCCTACAGTCCT
CAGGACTCTACTCCCTCAGCAGCGTGGTGACCGT
GCCCTCCAGCAACTTCGGCACCCAGACCTACACC
TGCAACGTAGATCACAAGCCCAGCAACACCAAG
GTGGACAAGACAGTTGAGCGCAAATGTTGTGTCG
AGTGCCCACCGTGCCCAGCACCACCTGTGGCAGG
ACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAG
GACACCCTCATGATCTCCCGGACCCCTGAGGTCA
CGTGCGTGGTGGTGGACGTGAGCCACGAAGACCC
CGAGGTCCAGTTCAACTGGTACGTGGACGGCGTG
GAGGTGCATAATGCCAAGACAAAGCCACGGGAG
GAGCAGTTCAACAGCACGTTCCGTGTGGTCAGCG
TCCTCACCGTTGTGCACCAGGACTGGCTGAACGG
CAAGGAGTACAAGTGCAAGGTCTCCAACAAAGG
CCTCCCAGCCCCCATCGAGAAAACCATCTCCAAA
ACCAAAGGGCAGCCCCGAGAACCACAGGTGTAC
ACCCTGCCCCCATCCCGGGAGGAGATGACCAAGA
ACCAGGTCAGCCTGACCTGCCTGGTCAAAGGCTT
CTACCCCAGCGACATCGCCGTGGAGTGGGAGAGC
AATGGGCAGCCGGAGAACAACTACAAGACCACA
CCTCCCATGCTGGACTCCGACGGCTCCTTCTTCCT
CTACAGCAAGCTCACCGTGGACAAGAGCAGGTG
GCAGCAGGGGAACGTCTTCTCATGCTCCGTGATG
CATGAGGCTCTGCACAACCACTACACGCAGAAGA
GCCTCTCCCTGTCTCCGGGTAAATGA
Figure imgf000114_0001
Figure imgf000115_0001
Table 19: DNA and protein sequences of antibody 24.2.1
DESCRIPTION: SEQUENCE (signal sequence underlined):
Heavy Chain ATGAAACATCTGTGGTTCTTCCTTCTCCTGGTGGC DNA AGCTCCCAGATGGGTCCTGTCCCAGGTGCAGCTG
CAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCGG
AGACCCTGTCCCTCACCTGCACTGTCTCTGGTGGC
TCCATCAGAGGTTACTACTGGAGCTGGATCCGGC
AGCCCCCAGGGAAGGGACTGGAGTGGATTGGGT
ATATCTATTACAGTGGGAGCACCAACTACAACCC
CTCCCTCAAGAGTCGAGTCACCATATCAGTAGAC
ACGTCCAAGAACCAGTTCTCCCTGAAGCTGAGTT
CTGTGACCGCTGCGGACACGGCCGTGTATTACTG
TGCGAGAAGGGGGGGCCTCTACGGTGACTACGG
CTGGTTCGCCCCCTGGGGCCAGGGAACCCTGGTC
ACCGTCTCCTCAGCCTCCACCAAGGGCCCATCGG
TCTTCCCCCTGGCGCCCTGCTCCAGGAGCACCTCC
GAGAGCACAGCGGCCCTGGGCTGCCTGGTCAAG
GACTACTTCCCCGAACCGGTGACGGTGTCGTGGA
ACTCAGGCGCTCTGACCAGCGGCGTGCACACCTT
CCCAGCTGTCCTACAGTCCTCAGGACTCTACTCCC
TCAGCAGCGTGGTGACCGTGCCCTCCAGCAACTT
CGGCACCCAGACCTACACCTGCAACGTAGATCAC
AAGCCCAGCAACACCAAGGTGGACAAGACAGTT
GAGCGCAAATGTTGTGTCGAGTGCCCACCGTGCC
CAGCACCACCTGTGGCAGGACCGTCAGTCTTCCT
CTTCCCCCCAAAACCCAAGGACACCCTCATGATC
TCCCGGACCCCTGAGGTCACGTGCGTGGTGGTGG
ACGTGAGCCACGAAGACCCCGAGGTCCAGTTCAA
CTGGTACGTGGACGGCGTGGAGGTGCATAATGCC
AAGACAAAGCCACGGGAGGAGCAGTTCAACAGC
ACGTTCCGTGTGGTCAGCGTCCTCACCGTTGTGC
ACCAGGACTGGCTGAACGGCAAGGAGTACAAGT
GCAAGGTCTCCAACAAAGGCCTCCCAGCCCCCAT
CGAGAAAACCATCTCCAAAACCAAAGGGCAGCC
CCGAGAACCACAGGTGTACACCCTGCCCCCATCC
CGGGAGGAGATGACCAAGAACCAGGTCAGCCTG
ACCTGCCTGGTCAAAGGCTTCTACCCCAGCGACA
TCGCCGTGGAGTGGGAGAGCAATGGGCAGCCGG
AGAACAACTACAAGACCACACCTCCCATGCTGGA
CTCCGACGGCTCCTTCTTCCTCTACAGCAAGCTCA
CCGTGGACAAGAGCAGGTGGCAGCAGGGGAACG
TCTTCTCATGCTCCGTGATGCATGAGGCTCTGCAC
AACCACTACACGCAGAAGAGCCTCTCCCTGTCTC
CGGGTAAATGA DESCRIPTION: SEQUENCE (signal sequence underlined):
Heavy Chain MKHLWFFLLLVAAPRWVLSOVOLOESGPGLVKPSE Protein TLSLTCTVSGGS-RGYYWSWIRQPPGKGLEWIGY-Y
YSGSTNYNPSLKSRVTISVDTSKNQFSLKLSSVTAA
DTAVYYCARRGGLYGDYGWFAPWGQGTLVTVSS
ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPN
TNSWΝSGALTSGVHTFPAVLQSSGLYSLSSVVTVPS
SΝFGTQTYTCΝVDHKPSΝTKVDKTNERKCCVECPP
CPAPPNAGPSVFLFPPKPKDTLMISRTPEVTCWVD
VSHEDPEVQFΝWYVDGVEVHΝAKTKPREEQFΝSTF
RVVSVLTVVHQDWLΝGKEYKCKVSΝKGLPAPIEK
TISKTKGQPREPQVYTLPPSREEMTKΝQVSLTCLVK
GFYPSDIAVEWESΝGQPEΝΝYKTTPPMLDSDGSFFL
YSKLTVDKSRWQQGΝVFSCSVMHEALHΝHYTQKS
LSLSPGK
Light Chain DΝA ATGGAAACCCCAGCGCAGCTTCTCTTCCTCCTGCT
ACTCTGGCTCCCAGATACCACCGGAGAAATTGTG
TTGACGCAGTCTCCAGGCACCCTGTCTTTGTCTCC
AGGGGAAAGAGCCACCCTCTCCTGCAGGGCCAGT
CAGAGTGTTAGCAGCACCTACTTAGCCTGGTACC
AGCAGAAACCTGGCCAGGCTCCCAGGCTCCTCAT
CTATGGTGCATCCAGCAGGGCCACTGGCATCCCA
GACAGGTTCAGTGGCAGTGGGTCTGGGACAGACT
TCACTCTCACCATCAGCAGACTGGAGCCTGAAGA
TTTTGCAGTGTATTACTGTCAGCAGTATAGTAGCT
TATTCACTTTCGGCCCTGGGACCAAAGTGGATAT
CAAACGAACTGTGGCTGCACCATCTGTCTTCATC
TTCCCGCCATCTGATGAGCAGTTGAAATCTGGAA
CTGCCTCTGTTGTGTGCCTGCTGAATAACTTCTAT
CCCAGAGAGGCCAAAGTACAGTGGAAGGTGGAT
AACGCCCTCCAATCGGGTAACTCCCAGGAGAGTG
TCACAGAGCAGGACAGCAAGGACAGCACCTACA
GCCTCAGCAGCACCCTGACGCTGAGCAAAGCAG
ACTACGAGAAACACAAAGTCTACGCCTGCGAAGT
CACCCATCAGGGCCTGAGCTCGCCCGTCACAAAG
AGCTTCAACAGGGGAGAGTGTTAG
Light Chain METPAQLLFLLLLWLPDTTGE-VLTOSPGTLSLSPGE Protein RATLSCRASQSVSSTYLAWYQQKPGQAPRLLIYGA
SSRATGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYC
QQYSSLFTFGPGTKNDIKRTNAAPSNFIFPPSDEQLK
SGTASNVCLLΝΝFYPREAKVQWKVDΝALQSGΝSQ
ESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACE
VTHQGLSSPVTKSFΝRGEC Table 20: DNA and protein sequences of the mature variable domains of antibody 22.1.1H-C109A
Figure imgf000118_0001
Table 21: DNA and protein sequences of the mature variable domains of antibody 23.28.1L-C92A
Figure imgf000118_0002
EXAMPLE III
Analysis of Heavy and Light Chain Amino Acid Substitutions
[0264] Figures 1D-1H and 2D-2H provide sequence alignments between the predicted heavy chain variable domain amino acid sequences of monoclonal antibodies 3.1.1, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.28.1, 23.28.1H-D16E, 23.29.1 and 24.2.1 antibodies and the germline amino acid sequences of their respective genes. Most of the heavy chain CDR3 regions contain amino acid insertions. [0265] The DLR1 gene used in the VH domain of antibody 21.4.1 codes for two cysteine (Cys) residues. Mass spectrometry analysis and homology modeling demonstrated that the two Cys residues are disulfide-linked, and that this disulfide link does not disrupt the stracture of the antibody.
[0266] Figures 1A-1C and 2A-2C provide sequence alignments between the predicted light chain variable amino acid sequences of monoclonal antibodies 3.1.1, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.28.1, 23.28.1L-C92A, 23.29.1 and 24.2.1 clones and the germline amino acid sequences of their respective genes. The light chains of these antibodies are derived from three different N c genes. Seven of the eleven antibodies use the A3/A19 VK gene, six of which have two mutations in the CDRl region. Further, five of the seven antibodies that use the A3/A19 V/c gene, also use the J/cl gene; in all of these antibodies the first amino acid derived from the J/cl gene is consistently changed from a W to an R.
[0267] It will be appreciated that many of the above-identified amino acid substitutions or insertions exist in close proximity to or within a CDR. Such substitutions would appear to bear some effect upon the binding of the antibody to the CD40 molecule. Further, such substitutions could have significant effect upon the affinity of the antibodies.
EXAMPLE IN Species Crossreactivity of the Antibodies of the Invention [0268] We performed FACS analyses to determine the binding and affinity of the antibodies of the invention for CD40 from various species, particularly certain old world monkeys. We incubated aliquots of human and monkey whole blood for 1 hour on ice with increasing concentrations of anti-CD40 antibodies of the invention exemplified herein or with an anti-keyhole limpet hemocyanin (KLH) antibody as a negative control We then incubated the samples for 30 minutes on
_ 5 ice with anti-human IgG2-conjugated RPE (phycoerythrin). We measured binding by flow cytometry of CD19/CD20 positive B cells and analyzed the histograms of fluorescence intensity (F12-H) versus cell number (Counts) using CellQuest software. We estimated binding (KD) for each antibody from graphs of mean fluorescence intensity versus antibody concentration. We controlled for depletion
10 of the antibody by measuring binding over a range of cell concentrations.
[0269] We tested antibodies 3.1.1, 7.1.2, 10.8.3, 15.1.1 and 21.4.1 for binding to human, rhesus and cynomolgus B cells. We also tested antibodies 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1, 23.29.1 and 24.2.1 for binding to human and cynomolgus B cells.
15 [0270] We observed that the maximum signal and the concentration for half maximum binding to monkey cells, was within a factor of two to the conesponding parameters for human B cells. No binding was observed in similar experiments with mouse, rat, rabbit and dog blood.
EXAMPLE V
20 Selectivity of Antibodies for CD40
[0271] We conducted another in vitro assay to determine the selectivity of antibodies of the invention with respect to CD40.
CD40 Selectivity ELISA: Materials and Methods
[0272] We coated a 96-well FluroNUNC plate (Nunc Cat No. 475515) with four 25 antigens: CD40/Ig, CD44/Ig, RANK/Ig, 4-lBB/Ig, TNFR-1/Ig and TNFR-2/Ig
(antigens generated in-house), overnight at +4°C at 1 μg/ml of lOOμl/well in 0.1M sodium bicarbonate buffer, pH 9.6. We then washed the plate with PBST (PBS + 0.1% Tween-20) three times and blocked the plate with PBST+0.5%BSA at 150 μl/well. We incubated the plate at room temperature for 1 hour and then washed
30 with PBST three times. Next, we diluted the anti-CD40 antibodies generated in
Example I in block at 1 μg/ml and added the diluted antibodies to the plate. We incubated the plate at room temperature for 1 hour then washed with PBST three times. We then treated the wells that contained the antibodies generated in Example I with 100 ml/well anti-human IgG2-HRP (Southern Biotech Cat No.9070-05) at a 1 :4000 dilution in block. Also, we treated one row with anti- human IgG (Jackson Cat No. 209-035-088) diluted to 1 :5000 in block and added at 100 μl/well to normalize for plate coating. We also treated one row with anti- human CD40-HRP (Pharmingen Cat No. 345815/Custom HRP conjugated) at 0.05 μg/ml diluted in block as a positive control. We incubated the plate at room temperature for 1 hour and then washed with PBST three times. We added TMB substrate (K & P Labs) at 100 μl/well and incubated the plate for 5 to 10 minutes. We then read the plate using a Spectra-Max™ plate reader. The results showed that the antibodies have a selectivity for CD40 that is at least 100 times greater than their selectivity for RANK, 4-1BB, TNFR-1 and TNFR-2 in that the CD4 — specific signal (CD40 signal minus background) is at least 100X greater than the conesponding signal for the other molecules.
EXAMPLE VI
Epitope Classification Studies
[0273] Having demonstrated that the antibodies of the invention are selective for CD40, we performed competition binding analysis using BIAcore and FACS.
BIAcore Competition Studies
[0274] We conducted BIAcore competition studies to determine whether the human anti-CD40 antibodies of the invention bind to the same or distinct sites on the CD40 molecule. [0275] In these experiments we used a BIAcore 2000 instrument, following the manufacturer's protocols. Protein-A was immobilized on the sensor chip surfaces of the BIAcore. A saturating concentration of CD40-Ig which comprises the extracellular domain of CD40 was bound to the sensorchip. We then bound a first human agonist anti-CD40 antibody of the invention, a commercial anti-CD40 antibody or CD40L to the sensorchip-bound CD40 under saturating conditions. We then measured the ability of a second human agonist anti-CD40 antibody of the invention to compete with the first antibody, commercial antibody or CD40L for binding to CD40. This technique enabled us to assign the antibodies to different binding groups. Binding to CD40 indicated recognition of an independent epitope. Lack of binding may indicate recognition of the same epitope or overlapping epitopes.
FACS Studies
[0276] We conducted FACS studies to determine whether the human anti-CD40 antibodies of the invention bind to the same or distinct sites on the CD40 molecule, and to determine whether they bind to the same or distinct site on the CD40 molecule as commercially available anti-CD40 antibodies EA5 (Alexis Cat. No. ANC-300-050), LOB7/6 (Serotec MCA/590PE) and 5C3 (Pharmingen # 555458 (unlabeled) and 555460 (PE labeled for FACS).
[0277] We counter-stained dendritic cells treated with anti-CD40 antibodies of the invention with PE labeled EA5 or PE labeled LOB7/6 antibody on ice for 30 minutes. After a wash, cell staining was analyzed on a B-D caliber cytometer. Reduced binding of the commercial antibodies was interpreted as an indication that the test antibody bound to the same or overlapping epitope. [0278] Competition binding analysis by BIAcore and FACS showed that the epitopes recognized by mAb 21.4.1 antibodies overlaps with the epitope recognized by the EA5 antibody, did not overlap with the epitope recognized by the commercially available LOB7/6 antibody and does not overlap with the binding site for CD40L. . The epitopes recognized by the remaining antibodies do overlap with the binding site for CD40L.
[0279] Table 22 summarizes the results of these epitope classification studies.
TABLE 22
BIAcore Competition Epitope Classification of Certain Anti-CD40 Antibodies Of The Invention
Figure imgf000123_0001
EXAMPLE Nil
Upregulation of Surface Molecules by Anti-CD40 Antibodies [0280] We conducted a whole blood assay to determine whether the human anti- CD40 antibodies of the invention upregulate the expression of surface molecules on B cells.
[0281 ] Human or primate whole blood was diluted 1 : 1 with RPMI medium and incubated 24 hours with various concentrations of CD40 agonist antibodies or controls. Cells were stained for 30 minutes (on ice, in the dark) for HLA-DR, ICAM, B7-1, B7-2, CD19/CD20, CD40, CD23 and CD71, using commercially available, fluorochrome labeled antibody reagents. The cells were then analyzed on a FACS-Caliber (Becton-Dickinson). B-cells were identified by gating on CD 19 or CD20 positive cells, and activation markers determined for this gate. [0282] The maximum fold increase of median fluorescence (at ≤l μg/ml antibody), and mean EC50 obtained using one of the anti-CD40 antibodies of the claimed invention (21.4.1) are shown in Table 23.
TABLE 23
Upregulation of B-Cell Surface Molecules by an Anti-CD40 Antibody of the Invention
Figure imgf000124_0001
[0283] We also conducted experiments to determine whether the human anti-
CD40 antibodies of the invention upregulate the expression of surface molecules of monocyte-derived dendritic cell.
Preparation of the monocyte derived dendritic cells
[0284] Peripheral blood was collected from normal human volunteers. Mononuclear cells were isolated using Sigma Accuspin tubes (St. Louis, MO), washed with RPMI media (Gibco BRL, Rockville, MD) and placed into tissue culture flasks at 5 x 106/ml in complete RPMI medium (containing 100 U/ml penicillin/streptomycin, 10 mM HEPES buffer, 2 mM glutamine, 0.1 mM non- essential amino acids; all from Gibco BRL); and 10% fetal calf serum (Hyclone, Logan, Utah). After a 3 hours of incubation at 37°C (5% CO ), non-adherent cells were removed and the T cells were isolated using selection columns (R&D systems, Minneapolis, MN). The adherent cells were washed with RPMI medium and incubated for 7 days in complete RPMI medium supplemented with 10 ng/ml IL-4 (R&D systems) and 100 ng/ml GM-CSF (R&D systems). The non-adherent cells were then isolated, washed, and utilized as monocyte derived dendritic cells (mDCs) for all experiments. The remaining adherent cells were removed using trypsin / EDTA and utilized in experiments employing adherent monocytes. [0285] To determine whether the anti-CD40 antibodies of the invention upregulate the expression of cell surface markers, the monocyte derived dendritic cells were cultured with various concentrations of agonist antibodies for 48-72 hours, followed by staining (30 minutes, on ice, in the dark) for HLA-DR, ICAM, B7-1, B7-2, CD40 and CD83, using commercially available fluorochrom labeled antibody reagents. The cells were then analyzed on a FACS-Caliber (Becton- Dickinson).
[0286] The maximum fold increase of median fluorescence (at <1 μg/ml antibody), and mean EC50 obtained using one of the anti-CD40 antibodies of the claimed invention (21.4.1) are shown in Table 24.
TABLE 24
Upregulation of Dendritic Cell Surface Molecules by an Anti-CD40 Antibody of the Invention
Figure imgf000125_0001
[0287] We conducted similar experiments with B cells and mDCs using various anti-CD40 antibodies of the invention and additional markers. We measured the expression of B cell surface molecules (MHC-II, ICAM, B7-1, B7-2 and CD23) as described above but using 1 μg/ml of the anti-CD40 antibody. The results of this experiment are presented in Table 25. We measured the expression of dendritic cell surface molecules (MHC-II, ICAM, B7-1, B7-2 and CD83) after 72 hours as indicated above but using 1 μg/ml of the anti-CD40 antibody. The results of this experiment are presented in Table 26. Tables 25-26 show the fold increase in median intensity +/- standard deviation.
TABLE 25
Upregulation of B-Cell Surface Molecules by Anti-CD40 Antibodies Of The Invention
Figure imgf000126_0001
TABLE 26
Upregulation of Dendritic Cell Surface Molecules by Anti-CD40 Antibodies Of The Invention
Figure imgf000126_0002
Figure imgf000127_0001
[0288] Table 27 compares the upregulation of cell surface molecules in dendritic cells over B cells in terms of the ratio of the mean-fold increase on dendritic cells over the mean-fold increase on B cells.
TABLE 27 Upregulation of Cell Surface Molecules On Dendritic Cells Over B Cells
Figure imgf000127_0002
EXAMPLE NIII Enhancement of Cytokine Secretion [0289] We conducted a monocyte derived dendritic cell assay to determine whether the human anti-CD40 antibodies of the invention enhance the secretion of IL-12p40, IL-12p70 and IL-8.
[0290] The monocyte derived dendritic cells and the adherent moήocytes were prepared as described above. Cells were cultured in the presence of an anti-CD40 antibody of the invention (21.4.1 ) or with a anti-keyhole limpet hemocyanin (KLH) antibody as a negative control. The cytokines were measured in the supematants at 24 hours by ELISA (R&D systems). In some studies (see Table 28), the monocyte derived dendritic cells treated with the antibody also were co- stimulated with either 100 ng/ml LPS (Sigma), 1000 U/ml IFNγ (R&D systems) or 25 ng/ml IL-1/3 R&D systems.
[0291] The anti-CD40 antibody enhanced IL-12ρ40, IL-12p70 and IL-8 production in both monocyte derived dendritic cells and adherent monocytes. The presence of LPS further enhanced the production of IL-12p40 and IL-12p70. Only minimal levels of cytokines were detected in the supematants of dendritic cells incubated with the isotype control antibody, anti-KLH. Representative results are presented in Table 28 and in Figures 3 and 4. Table 28 summarizes the principle cytokines produced by dendritic cells or adherent monocytes by 1 μg/ml of an anti- CD40 antibody of the invention (21.4.1) +/- 100 ng/ml LPS. As shown in Figure 3, the anti-CD40 antibody enhanced IL-12p40 production by human dendritic cells. Figure 4 illustrates enhanced IL-12p70 production by human dendritic cells in the presence of antibody and 100 ng/ml LPS.
TABLE 28
Enhancement of IL-12p40, IL-12p70 and IL-8 Secretion by an Anti-CD40 Antibody of the Invention
Figure imgf000128_0001
ND = not determined [0292] Similar experiments were performed using multiple anti-CD40 antibodies of the invention. The monocyte derived dendritic cells were prepared as described above and cultured in the presence of various concentrations of the anti-CD40 antibodies and were co-stimulated with 100 ng/ml LPS (Sigma). The IL-12p70 in the supernatant was measured at 24 hours by ELISA (R&D systems) and the for each antibody EC5Q was determined. The results of the experiments are presented in Table 29.
TABLE 29
Enhancement of IL-12p70 Secretion In Dendritic Cells
DC IL-12p70
Antibody EC<;0 Max Clone μg/ml pg/ml
21.4.1 0.3 1796-7004
22.1.1 0.1 720-1040
23.25.1 0.2 540-960
23.5.1 0.1 676-1112
24.2.1 0.2 754-3680
3.1.1 0.2 668-960
23.28.1 0.2 1332-1404
23.29.1 0.1 852-900
21.2.1 0.03 656-872
[0293] We also tested the ability of the anti-CD40 antibodies of the invention to enhance the secretion of EFN-gamma from T cells in an allogenic T cell dendritic cell assay. To perform this assay, T cells and monocytes were isolated from the peripheral blood of healthy volunteers. Monocytes were differentiated into dendntic cells using the above-described methods. 1 x 10 T cells obtained from an individual were cultured with 1 x 105 dendritic cells obtained from a different individual in the presence of an anti-CD40 antibody of the invention or a control antibody. After 4 days of culture, the supematants were assayed for IFN-gamma secretion by ELISA. The results of this assay are shown in Table 30. TABLE 30
Enhancement of IFN-gamma Secretion by Anti-CD40 Antibodies Of The Invention
Allo DC/T LFNγ
Antibody EC_5j- Max Clone μg/ml pg/ml
2L4.1 0.3 212
22.1.1 0.3 110-180
23.25.1 0.3 180-232
23.5.1 0.2 150-240
24.2.1 0.2 111-194
3.1.1 0.1 100-195
23.28.1 0.2 120-190
23.29.1 0.3 134-150
21.2.1 0.03 230-256
EXAMPLE LX
Induction of Inflammatory Cytokines by the Anti-CD40 Antibodies of the Invention
[0294] Antibodies 10.8.3, 15.1.1, 21.4.1 and 3.1.1 were tested in a whole-blood cytokine release assay described by Wing et al, Tlierapeutic. Immunol. 2:183-90 (1995) to determine if inflammatory cytokines are induced by the antibodies at 1, 10 and 100 μg/ml concentration. No significant release of TNF-α, IL-1/3, IFN-7, or IL-6 was observed with these antibodies at the indicated concentrations in blood from 10 normal donors.
EXAMPLE X
Enhancement of Immunogenicity of Cell Line Jy by Anti-CD40 Antibodies
[0295] CD40 positive JIYOYE cells (ATCC CCL 87) ("Jy cells") were cultured and maintained in RPMI medium. JIYOYE cells were incubated for 24 hours with an anti-CD40 antibody of the invention (21.4.1), or with an isotype matched antibody (anti-KLH), in complete RPMI medium. Cells were then washed and treated with 25 mg mitomycin C (Sigma) / 7 ml media for 60 min. These cells were then incubated with isolated human T cells at a 1 : 100 ratio for 6 days at 37°C (5% CO2). T cells were then collected, washed, and the level of CTL activity determined against fresh chromium 51 (New England Nuclear, Boston, MA) labeled JIYOYE cells. Specific CTL activity was calculated as % specific cytolysis=(cytolysis Jy (cpm) - spontaneous cytolysis (cpm))/(total cytolysis (cpm) - spontaneous cytolysis (cpm)).
[0296] As Figure 5 illustrates, an anti-CD40 antibody of the invention (21.4.1) significantly enhanced the immunogenecity against Jy cells treated with the antibody.
EXAMPLE XI
Animal Tumor Model
[0297] To further investigate the anti-tumor activity of the anti-CD40 antibodies made in accordance with the invention, we designed a SCID-beige mouse model to test the in vivo effect of the antibody on tumor growth.
[0298] We obtained SCID-beige mice from Charles River and we allowed the mice to acclimate one week prior to use. We injected tumor cells (Daudi cells (ATCC CCL 213), CD40(-) K562 cells (ATCC CCL 243) and CD40(+) Raji cells (ATCC CCL 86), BT474 breast cancer cells (ATCC HTB 20) or PC-3 prostate cells (ATCC CRL 1435)) subcutaneously at a concentration of 1 x 107 cells/animal In some cases, we injected T cells (5 x 105) and dendritic cells (1 x 105) from the same human donor along with the tumor cells. We also injected an anti-CD40 antibody of the invention, or an isotype matched control (anti-KLH), intraperitoneally, immediately prior to tumor injection (one injection only). We then measured tumor growth. Specific experiments are described below. [0299] In one experiment, we injected an anti-CD40 antibody of the invention (21.4.1), or an isotype matched control (anti-KLH), intraperitoneally, at a dose of 10 mg/kg immediately prior to tumor injection (one injection only). The tumor cells (Daudi cells) were injected subcutaneously at a concentration of 1 x 107 cells/animal We measured tumor growth with calipers at days 17, 19, 20, 21, 25, 26, 27 and 28 after implantation in the presence of human T cells and dendritic cells. As shown in Figure 6, the anti-CD40 antibody inhibited tumor growth by about [60]%.
[0300] In another experiment, we injected an anti-CD40 antibody of the invention (21.4.1), or an isotype matched control (anti-KLH), intraperitoneally, at a dose of 0.1 mg/kg, 1 mg/kg or 10 mg/kg immediately prior to tumor injection (one injection only). The tumor cells (K562 cells) were injected subcutaneously at a concentration of 1 x 107 cells/animal In this experiment we injected T cells (5 x 105) and dendritic cells (1 x 105) from the same human donor along with the tumor cells. We measured tumor growth with calipers at days 17, 19, 20, 21, 25, 26, 27 and 28 after implantation. As shown in Figure 7, the anti-CD40 antibody inhibited tumor growth by 60-85%.
[0301] In another experiment, we injected an anti-CD40 antibody of the invention (21.4.1, 23.29.1 or 3.1.1), or an isotype matched confrol (anti-KLH), intraperitoneally, immediately prior to tumor injection (one injection only). The isotype matched control antibody and antibody 21.4.1 were injected at a dose of 1 mg/ml. Antibodies 23.29.1 and 3.1.1 were injected at a dose of 1, 0.1, 0.01, 0.001 or 0.0001 mg/kg. The tumor cells (K562 cells) were injected subcutaneously at a concentration of 1 x 107 cells/animal In this experiment we injected T cells (5 x 105) and dendritic cells (1 x 105) from the same human donor along with the tumor cells. We then measured tumor growth with calipers at day 28 after implantation. The results of this experiment are shown in Figures 8 and 9. Each point in the figures represents a measurement from an individual animal. [0302] In another experiment, we injected an anti-CD40 antibody of the invention (21.4.1 ), or an isotype matched control (anti-KLH), intraperitoneally, immediately prior to tumor injection (one injection only). The antibodies were injected at a dose of 1, 0.1, 0.01, 0.001 or 0.0001 mg/kg. The tumor cells (Raji cells) were injected subcutaneously at a concentration of 1 x 107 cells/animal hi some animals, we injected T cells (5 x 105) and dendritic cells (1 x 105) from the same human donor along with the tumor cells. We then measured tumor growth with calipers at day 28 after implantation. The results of this experiment are shown in Figure 10. Each point in the figure represents a measurement from an individual animal.
[0303] hi yet another experiment, we injected an anti-CD40 antibody of the invention (21.4.1, 23.28.1, 3.1.1 or 23.5.1), or an isotype matched confrol (anti- KLH), intraperitoneally, immediately prior to tumor injection (one injection only). The antibodies were injected at a dose of 1 or 0.1 mg/kg. The tumor cells (Raji cells) were injected subcutaneously at a concentration of 1 x 107 cells/animal We then measured tumor growth with calipers at day 28 after implantation. The results of this experiment are shown in Figure 11. Each point in the figure represents a measurement from an individual animal.
[0304] In yet another experiment, we injected an anti-CD40 antibody of the invention (21.4.1, 23.29.1, or 3.1.1), or an isotype matched control (anti-KLH), intraperitoneally, immediately prior to tumor injection (one injection only). The antibodies were injected at a dose of 1 mg/kg. The tumor cells (BT474 breast cancer cells) were injected subcutaneously at a concentration of 1 x 107 cells/animal We injected T cells (5 x 105) and dendritic cells (1 x 105) from the same human donor along with the tumor cells. We then measured tumor growth with calipers at day 39 after implantation. As shown in Figure 12, all of the antibodies inhibited breast cancer tumor growth. Each point in the figure represents a measurement from an individual animal.
[0305] In yet another experiment, we injected an anti-CD40 antibody of the invention (3.1.1), or an isotype matched control (anti-KLH), infraperitoneally, immediately prior to tumor injection (one injection only). The antibodies were injected at a dose of 1 mg/kg. The tumor cells (PC-3 prostate tumor cells) were injected subcutaneously at a concentration of 1 x 107 cells/animal We then measured tumor growth with calipers at day 41 after implantation. As shown in Figure 13, the anti-CD40 antibody inhibited prostate tumor growth by about 60%. Each point in the figure represents a measurement from an individual animal. EXAMPLE XII
Survival of SCID-Beige Mice Injected with Daudi Tumor Cells And Treated With The Anti-CD40 Antibodies Of The Invention
[0306] In another experiment, we injected an anti-CD40 antibody of the invention, or an isotype matched (one injection) confrol, intraperitoneally, immediately prior to tumor injection. The antibodies were injected at a dose of 1 or 0.1 mg/kg. The tumor cells (Daudi cells) were injected intravenously at a dose of 5 x 106 cells/animal We then momtor animal survival. As shown in Figure 14, all of the anti-CD40 antibodies tested prolonged the survival of mice injected tumors by at least six days.
[0307] Table 31 lists the ED50 of the anti-CD40 antibodies in the different solid tumor models described in Example XI. Table 31 summarizes the in vivo antitumor activity of some of the anti-CD40 antibodies of the invention in SCID mice. In addition, the table lists the ED50 of the anti-CD40 antibodies in the Daudi systemic tumor model described above in Example XII.
TABLE 31
ED50 Of Anti-CD40 Antibodies Of The Invention Using Different In Nivo Tumor Models in SCID mice
Antibody CD40(-) CD40(+) Raji CD40(+) Raji CD40(+)
K562 & T/DC Daudi
& T/DC sub- subsubcutaneous cutaneous intra-venous cutaneous (mg/kg) (mg/kg) (mg/kg)
(mg/kg)
21.4.1 0.005 0.0008 0.016 0.1
22.1.1 0.01 ΝD > 1.0 0.1
23.25.1 ≥ l.O ΝD > 1.0 ΝD
23.5.1 > 1.0 ΝD ≥ l.O ΝD
24.2.1 > 1.0 ΝD > 1.0 ΝD
3.1.1 0.02 ΝD ≥ O.l ≤ O.l
23.28.1 > 1.0 ΝD ≥ l.O 0.1
23.29.1 0.009 ΝD > 1.0 ≤ O.l
21.2.1 ≤ l .O ΝD ΝD ΝD
ΝD= Not Done EXAMPLE Xiπ
Determination of Affinity Constants (KD) of Fully Human Anti-CD40 Antibodies by BIAcore
[0308] We performed affinity measures of purified antibodies by surface plasmon resonance using the BIAcore 3000 instrument, following the manufacturer's protocols.
[0309] The Biosensor biospecific interaction analysis instrument (BIAcore) uses surface plasmon resonance to measure molecular interactions on a CM5 sensor chip. Changes in the refractive indices between two media, glass and carboxymethylated dextran, caused by the interaction of molecules to the dextran side of the sensor chip, is measured and reported as changes in arbitrary reflectance units (RU) as detailed in the manufacturer's application notes. [0310] The carboxymethylated dextran surface of a flow cell on a sensor chip was activated by derivatization with 0.05 M N-hydroxysuccinimide mediated by
0.2 M N-ethyl-N' -(dimethylaminopropyl) carbodiimide for 7 min. CD40-Ig fusion protein (described in Example I) at a concentration of 5 μg/ml, in lOmM Na acetate, pH 3.5, was manually injected into the flow cell at a rate of 5 μl/min and covalently immobilized to the flow cell surface with the desired amount of RU's. Deactivation of unreacted N-hydroxysuccinimide esters was performed using 1 M ethanolamine hydrochloride, pH 8.5. Following immobilization, the flow cells are cleaned of any unreacted or poorly bound material with 5 regeneration injections of 5 μl of 50 mM NaOH until a stable baseline is achieved. Flow cell 2, a high density surface, measured approximately 300 RU's following surface preparation and flow cell 3, a low density surface, measured approximately 150 RU's. For flow cell 1, the activated blank surface, 35 μl of 10 mM Na acetate buffer was injected during immobilization in place of antigen. Flow cell 4 contained approximately 450 RU's of immobilized CTLA4-Ig, an inelevant antigen control. [0311] A dilution series of each antibody was prepared in the concentration range of 100 μg/ml to 0.1 μg/ml by half logs. The flow rate was set at 5 μl/min and 25 μl of each concentration point sample was injected over the sensor chip with a regeneration injection of 5 μl of 50 mM NaOH between each concentration of antibody injected. The data was analyzed using BIAevaluation 3.0 software. [0312] In reverse orientation kinetic experiments, the antibody 21.4.1 was immobilized to the sensor chip surface using the protocol described above. Anti- KLH was used as a control antibody surface. The antigen, CD40-Ig fusion protein, was injected in the concentration range of 100 μg/ml to 0.1 μg/ml [0313] Table 32 lists affinity measurements for representative anti-CD40 antibodies ofthe present invention:
TABLE 32
Affinity Measurements For Anti-CD40 Antibodies Of The Invention
Figure imgf000136_0001
EXAMPLE XIV
Epitope Mapping of Anti-CD40 Antibodies
[0314] The binding assays were done using Protein A purified CD40-human IgGl Fc fusion antigen. The human CD40-IgGl Fc fusion protein was cloned at Pfizer. The human CD40 IgGl fusion protein was expressed in a mammalian cell line and purified over Protein A column. The purity of the fusion antigen was assessed by SDS/PAGE.
[0315] CD40 has a structure of a typical type I transmembrane protein. The mature molecule is composed of 277 amino acids. The extracellular domain of CD40 consists of four TNFR-hke cysteine rich domains. See, e.g., Neismith and Sprang, TIBS 23:74-79 (1998); van Kooten and Banchereau, J. Leukocyte Biol. 67:2-17 (2000); Stamenkovic et al, EMBOJ. 8:1403-1410(1989).
Binding of Anti-CD40 Antibodies to Reduced and Non-Reduced Human CD40:
[0316] Because the extracellular domain of CD40 consists of four cysteine rich domains, disraption ofthe intramolecular bonds, by reducing agent, can change antibody reactivity. To determine whether disraption ofthe intramolecular bonds, by reducing agent, changed the reactivity of selected anti-CD40 antibodies ofthe invention, purified CD40-hIgG was loaded on SDS/PAGE (4-20% gel) under non- reducing (NR), or reducing (R), conditions. SDS PAGE was performed by the method of Laemmli, using a mini-gel system. Separated proteins were fransfened on to nitrocellulose membrane. Membranes were blocked using PBS containing 5% (w/v) non fat dried milk for at least 1 hour before developing, and probed for 1 hr with each antibody. Anti-CD40 antibodies were detected using HRP-conjugated goat anti-human immunoglobulins (1:8,000 dilution; Catalog No. A-8667 from Sigma). Membranes were developed by using enhanced Chemiluminescence (ECL®; Amersham Bioscience) according to the manufacturer's instructions. [0317] The Western Blot was then probed with four anti-CD40 antibodies of the invention: 21.4.1, 23.25.1, 23.29.1 and 24.2.1 (1 μg/ml,) followed by HRP conjugated goat anti-human IgG (1 :8000 dilution). The results of this experiment are show in Figure 15. The results indicate that antibodies 21.4.1, 23.25.1, 23.29.1 and 24.2.1 bind non-reduced but do not bind reduced CD40, the antibodies, thus, recognize a conformational epitope.
Binding of Anti-CD40 Antibodies to Human CD40 Domain Deleted Proteins:
[0318] The extracellular region of CD4O includes four TNFR-like repeat domains (refened to as D1-D4). See, e.g., Neismith and Sprang, TIBS 23:74-79 (1998); van Kooten and Banchereau, J Leukocyte Biol. 67:2-17 (2000); Stamenkovic et al, EMBOJ. 8:1403-1410(1989). Figure 16 shows the amino acid sequences ofthe mouse and human CD40 domains D1-D4. To investigate the contribution of different regions ofthe CD40 molecule in the presentation ofthe epitope, a number of domain deleted mutants were constructed. [0319] To make the human CD40 deletion constructs, the entire extracellular domain of human CD40 (amino acids 1-193) was amplified from human B cells (CD19+) cDNA (Multiple tissue cDNA panels, Catalog No. K1428-1, from Clontech) by PCR using sequence specific primers, and a 6XHis-tag was added at the C-terminal. A human CD40 5' primer 5'-GCAAGCTTCACCAATGGT TCGTCTGCCTCTGCAGTG-3' (SEQ ID NO: 135) was used with different combination of 3' primers for cloning of full length and truncated CD40 molecules. The 3' primer for cloning the full-length extracellular domain of human CD40 was: 5'-TCAGTGATGGTGATGGTGATGTCTCAGCCGAT CCTGGGGACCA-3' (SEQ ID NO: 136). The 3' primer used to clone the D1-D3 domains of human CD40 was: 5'-TCAGTGATGGTGATGGTGATGTGGGCA GGGCTCGCGATGGTAT-3' (SEQ ID NO: 137) The 3' primer used to clone the D1-D2 domains of CD40 was: 5'-TCAGTGATGGTGATGGTGATGA CAGGTGCAGATGGTGTCTGTT-3' (SEQ ID NO: 138). After these constructs of truncated CD40 cDNA were generated, they were expressed in the 293F cell line using the pCR3.1 vector (Invitrogen). The CD40-6XHis fusion proteins were purified by elution from a nickel column.
[0320] The amino acid sequences of these four deletion mutants are shown in Table 33.
TABLE 33 CD40 His-Tag Fusion Proteins
Figure imgf000138_0001
Figure imgf000139_0001
[0321] To express these human CD40 deletion constructs, the constructs were cloned into the pCR3.1 vector (Invitrogen) and expression was assessed in various stable and transiently transfected 293F cell lines. The supematants from transiently transfected 293F cells were analyzed for binding to antibodies 21.4.1, 23.25.1, 23.29.1 and 24.2.1 by ELISA and Western Blot. [0322] ELISA assays were preformed using supernatant from 293F cells transfected with different CD40 constructs. ELISA plates were coated goat anti- human CD40 polyclonal antibodies (R&D catalog No. AF 632) or goat anti-mouse CD40 polyclonal antibodies (R&D catalog No. AF 440) diluted to 1 μg/ml in ELISA plate coating buffer. Expression of CD40 constructs in 293F cells was confirmed by detection with biotinylated goat anti-human CD40 (R&D catalog No. BAF 632), goat anti-mouse CD40 (R&D catalog No. BAF 440), or HRP- conjugated anti-His (C terminal) antibody (Invitrogen, Catalog No. 46-0707). Binding of anti-CD40 human antibodies were detected with HRP conjugated goat anti-human IgG (FC specific Caltag HI 0507), diluted 1 :2,000. The results, as shown in Table 34, indicate that most if not all ofthe epitope recognized by mAbs 21.4.1, 23.28.1 and 23.29.1 is located in the D1-D2 region of CD40 while the epitope for mAb 24.2.1 is located at least partly in domain D3-D4. A human CD40-rabbit Fc fusion protein was used a control to confirm the specificity ofthe antibody binding.
TABLE 34 ELISA: Antibody Binding To CD40 Deletion Mutants
Figure imgf000139_0002
Figure imgf000140_0001
[0323] The CD40 deletion constructs also were analyzed by Western Blot analysis. The results are shown in Table 35. The ELISA results show that the binding site of antibodies 21.4.1, 23.25.1, 23.29.1 and 24.2.1 involves domains Dl- D3. The results also show that the binding site for antibodies 21.4.1, 23.25.1 and 23.29.1 involve domains D1-D2, and that the binding site of antibody 24.2.1 involves domain D3.
TABLE 35 Western Blot: Antibody Binding To CD40 Deletion Mutant
Figure imgf000140_0002
Binding of Anti-CD40 Antibodies to Mouse CD40:
[0324] We set out to determine the ability of antibodies 21.4.1 , 23.25.1 , 23.29.1 and 24.2.1 to bind mouse CD40.
[0325] For this experiment, mouse CD40 was amplified from mouse B cells cDNA. Mouse CD40(Dl-D3)-6xHis fusion protein was cloned into pCR3.1, which utilizes the CMV promoter, to drive transcription. The 5' primer used to clone the extracellular domain ofthe mouse CD40 was: 5'- TGCAAGCTTCACCATGGTGTCTTTGCCTCGGCTGTG-3'. The 3' primer used to clone the Dl - D3 domains of mouse CD40 was: 5'- GTCCTCGAGTCAGTGATGGTGATGGTGATGTGGGCAGGGATGACAGAC- 3'. Mouse and human cDNA constructs were transfected into 293F cells transiently. The expression of recombinant CD40 was detected by ELISA using polyclonal antibodies against mouse and human CD40, anti-His antibodies, and anti-CD40 antibodies 21.4.1, 23.25.1, 23.29.1 and 24.2.1. The results of these experiments are shown in Table 36. This experiment shows that all antibodies are specific to human CD40 and do not cross react with mouse CD40.
TABLE 36
Cross-Reactivity of Mouse and Human CD40
Figure imgf000141_0001
Binding of Anti-CD40 Antibodies to of Human/Mouse Chimeric CD40:
[0326] Because antibodies 21.4.1, 23.25.1, 23.29.1 and 24.2.1 do not bind mouse
CD40, we constructed human/mouse chimeric CD40 proteins to more definitively map the epitopes of those antibodies.
[0327] For the construction of in-frame fusions ofthe human and murine CD40 chimeric proteins, we used unique restriction sites at the borders of CD40 domains at identical positions in the cDNA of both human and mouse CD40. Various cDNA constructs of CD40 were generated using the EcoRI restriction site at the end of domain 1 (nucleotide 244, amino acid 64) and the Banl restriction site at the end of domain 2 (nucleotide 330, amino acid 94) (Figure 17).
[0328] Various CD40 domains were amplified by PCR and ligated. This approach allowed the replacement of various domains ofthe mouse CD40 by the homologous domains from the human CD40. The constructs obtained are shown in Figure 18.
[0329] We then determined whether antibodies 21.4.1, 23.25.1, 23.29.1 and 24.2.1 were able to bind the mouse/human chimeric CD40 proteins by ELISA. The results of this experiment are shown in Table 37. As shown in Table 37, mAbs 21.4.1 and 23.25.1 recognize and epitope that is located partly in Dl and partly in D2; mAb 23.29.1 recognizes an epitope located mostly if not completely in D2; and mAb 24.2.1 recognizes an epitope located in D2 and D3.
TABLE 37
Antibody Binding to Chimeric CD40 Proteins
Figure imgf000142_0001
[0330] AU publications and patent applications cited in this specification are herein incorporated by reference as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference. Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be readily apparent to those of ordinary skill in the art in light ofthe teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope ofthe invention.

Claims

What is Claimed is:
1. A chimeric or human monoclonal antibody or antigen-binding portion thereof that specifically binds to and activates human CD40, wherein said antibody or antigen-binding portion thereof comprises: a) a heavy chain comprising amino acid sequences of a heavy chain CDRl, a heavy chain CDR2 and a heavy chain CDR3 from a heavy chain variable region; wherein said amino acid sequences ofthe heavy chain CDRl and the heavy chain CDR2 are independently selected from a CDRl and a CDR2 of a heavy chain variable region, respectively, wherein the sequence of said heavy chain variable region comprises no more than 18 amino acid changes from the amino acid sequence encoded by a germline VH 3-30+, 4-59, 1-02, 4.35 or 3-30.3 gene; wherein said amino acid sequence ofthe heavy chain CDR3 is from a CDR3 of a heavy chain variable region, wherein said heavy chain variable region is selected from the group consisting of i) a heavy chain variable region of an antibody selected from the group consisting of 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-V88A-V97A, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.29.1 and 24.2.1; ii) a heavy chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOS: 2, 10, 18, 26, 34, 42, 50, 58, 66, 74 and 82, or a heavy chain variable region comprising an amino acid sequence from the heavy chain amino acid sequence selected from the group consisting of SEQ ID NOS: 90, 92, 96 and 98, or said amino acid sequence lacking a signal sequence; and iii) a heavy chain variable region encoded by a nucleic acid sequence selected from the group consisting of SEQ ID NOS: 1, 9, 17, 25, 33, 41, 49, 57, 65, 73 and 81, or a heavy chain variable region comprising an amino acid sequence encoded by the heavy chain nucleic acid sequence selected from the group consisting of 89, 91, 95 and 97, or said encoded heavy chain variable region lacking a signal sequence; wherein the amino acid sequence of said heavy chain CDR3 may have up to two conservative amino acid substitutes and/or two non-conservative amino acid insertions, deletions or substitutions therefrom; or b) a light chain comprising amino acid sequences of a light chain CDRl, a light chain CDR2 and a light chain CDR3 from a light chain variable region, wherein said amino acid sequences ofthe light chain CDRl and the light chain CDR3 are independently selected from CDRl and a CDR3 of a light chain variable region, respectively, wherein the light chain variable region comprises no more than ten amino acid changes from the amino acid sequence encoded by a germline V c A3/A19, L5 or A27 gene; and wherein said amino acid sequence ofthe light chain CDR2 is from a light chain variable region, wherein said light chain variable region is selected from the group consisting of i) a light chain variable region of an antibody selected from the group consisting of 3.1.1, 3.1.1L-L4M-L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.29.1 and 24.2.1; ii) a light chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOS: 4, 12, 20, 28, 36, 44, 52, 60, 68, 76 and 84, or a light chain variable region comprising an amino acid sequence from the light chain amino acid sequence selected from the group consisting of SEQ ID NOS: 94 and 100, or said amino acid sequence lacking a signal sequence; and iii) a light chain variable region encoded by a nucleic acid sequence selected from the group consisting of SEQ ID NOS: 3, 11, 19, 27, 35, 43, 51, 59, 67, 75 and 83, or a light chain variable region comprising an amino acid sequence encoded by the light chain nucleic sequence selected from the group consisting of SEQ ID NOS: 93 and 99, or said encoded light chain variable region lacking a signal sequence; wherein the amino acid sequence of said light chain CDR2 may have up to two conservative amino acid substitutes and/or two non-conservative amino acid insertions, deletions or substitutions therefrom.
2. The antibody or antigen-binding portion thereof according to claim 1, wherein
(a) the amino acid sequences of said heavy chain CDRl and said heavy chain CDR2 each have up to four conservative amino acid substitutions and two non-conservative amino acid insertions, deletions or substitutions from the amino acid sequences encoded by the germline VH 3-30+, 4-59, 1-02, 4.35 or 3- 30.3 gene; or
(b) the amino acid sequences of said light chain CDRl and said light chain CDR3 each have up to three conservative amino acid substitutes and two non-conservative amino acid insertions, deletions or substitutions from the gennline V/c A3/A19, L5 or A27 gene.
3. The antibody or antigen-binding portion thereof according to claim 1, wherein
(a) the amino acid sequences of said heavy chain CDRl and said heavy chain CDR2 are each independently selected from a CDRl and a CDR2 of a heavy chain variable region, wherein said heavy chain variable region is selected from the group consisting of: i) a heavy chain variable region of an antibody selected from the group consisting of 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-V88A-V97A, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.29.1 and 24.2.1; ii) a heavy chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOS: 2, 10, 18, 26, 34, 42, 50, 58, 66, 74, 82, 90, 92, 96 and 98, or said amino acid sequence lacking a signal sequence; and iii) a heavy chain variable region encoded by a nucleic acid sequence selected from the group consisting of SEQ ID NOS: 1, 9, 17, 25, 33, 41, 49, 57, 65, 73, 81, 89, 91, 95 and 97, or said encoded heavy chain variable region lacking a signal sequence; wherein the amino acid sequences of said heavy chain CDRl and said heavy chain CDR2 each may have up to two conservative amino acid substitutes and/or two non-conservative amino acid insertions, deletions or substitutions therefrom; or
(b) the amino acid sequences of said light chain CDRl and said light chain CDR3 are each independently selected from CDRl and a CDR3 from a light chain variable region, wherein said light chain variable region is selected from the group consisting of: i) a light chain variable region of an antibody selected from the group consisting of 3.1.1, 3.1.1L-L4M-L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.29.1 and 24.2.1; ii) a light chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOS: 4, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 94 and 100, or said amino acid sequence lacking a signal sequence; and iii) a light chain variable region encoded by a nucleic acid sequence selected from the group consisting of SEQ ID NOS: 3, 11, 19, 27, 35, 43, 51, 59, 67, 75, 83, 93 and 99, or said encoded light chain variable region lacking a signal sequence; wherein the amino acid sequences of said light chain CDRl and said light chain CDR3 each may have up to two conservative amino acid substitutions and/or two non-conservative amino acid insertions, deletions or substitutions therefrom.
4. The antibody or antigen-binding portion thereof according to claim 3, wherein
(a) the amino acid sequences of said heavy chain CDRl, said heavy chain CDR2 and said heavy chain CDR3 are each independently selected from a heavy chain variable region, wherein said heavy chain variable region is selected from the group consisting of: i) a heavy chain variable region of an antibody selected from the group consisting of 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-V88A-V97A,
7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.29.1 and 24.2.1; ii) a heavy chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOS: 2, 10, 18, 26, 34, 42,
50, 58, 66, 74, 82, 90, 92, 96 and 98, or said amino acid sequence lacking a signal sequence; and iii) a heavy chain variable region encoded by a nucleic acid sequence selected from the group consisting of SEQ ID NOS: 1, 9, 17, 25, 33, 41, 49, 57, 65, 73, 81, 89, 91, 95 and 97, or said encoded heavy chain variable region lacking a signal sequence; or
(b) the amino acid sequences of said light chain CDRl and said light chain CDR3 are each independently selected from CDRl and a CDR3 from a light chain variable region, wherein said light chain variable region is selected from the group consisting of: i) a light chain variable region of an antibody selected from the group consisting of 3.1.1, 3.1.1L-L4M-L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.29.1 and 24.2.1; ii) a light chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOS: 4, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 94 and 100, or said amino acid sequence lacking a signal sequence; and iii) a light chain variable region encoded by a nucleic acid sequence selected from the group consisting of SEQ ID NOS: 3, 11, 19, 27, 35, 43,
51, 59, 67, 75, 83, 93 and 99, or said encoded light chain variable region lacking a signal sequence.
5. The antibody or antigen-binding portion thereof according to claim 1, wherein
(a) said heavy chain comprises the amino acid sequence of a heavy chain variable region selected from the group consisting of i) a heavy chain variable region of an antibody selected from the group consisting of 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-V88A-V97A,
7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.29.1 and 24.2.1; ii) a heavy chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOS: 2, 10, 18, 26, 34, 42,
50, 58, 66, 74, 82, 90, 92, 96 and 98, or said amino acid sequence lacking a signal sequence; and iii) a heavy chain variable region encoded by a nucleic acid sequence selected from the group consisting of SEQ ID NOS: 1, 9, 17, 25, 33, 41, 49, 57, 65, 73, 81, 89, 91, 95 and 97, or said encoded heavy chain variable region lacking a signal sequence; wherein the amino acid sequences of said heavy chain variable region may have up to six conservative amino acid substitutions and/or four non- conservative amino acid insertions, deletions or substitutions therefrom; or
(b) said light chain comprises the amino acid sequence of a light chain variable region selected from the group consisting of i) a light chain variable region of an antibody selected from the group consisting of 3.1.1, 3.1.1L-L4M-L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.29.1 and 24.2.1; ii) a light chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOS: 4, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 94 and 100, or said amino acid sequence lacking a signal sequence; and iii) a light chain variable region encoded by a nucleic acid sequence selected from the group consisting of SEQ ID NOS: 3, 11, 19, 27, 35, 43,
51, 59, 67, 75, 83, 93 and 99, or said encoded light chain variable region lacking a signal sequence; wherein the amino acid sequences of said light chain variable region may have up to six conservative amino acid substitutes and/or four non- conservative amino acid insertions, deletions or substitutions therefrom.
6. The antibody or antigen-binding portion thereof according to claim 5, wherein (a) said heavy chain comprises the amino acid sequence of a heavy chain variable region selected from the group consisting of i) a heavy chain variable region of an antibody selected from the group consisting of 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-V88A-V97A, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H-D16E, 23.29.1 and 24.2.1; ii) a heavy chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOS: 2, 10, 18, 26, 34, 42,
50, 58, 66, 74, 82, 90, 92, 96 and 98, or said amino acid sequence lacking a signal sequence; and iii) a heavy chain variable region encoded by a nucleic acid sequence selected from the group consisting of SEQ JX) NOS: 1, 9, 17, 25, 33, 41, 49, 57, 65, 73, 81, 89, 91, 95 and 97, or said encoded heavy chain variable region lacking a signal sequence; or
(b) said light chain comprises the amino acid sequence of a light chain variable region selected from the group consisting of i) a light chain variable region of an antibody selected from the group consisting of 3.1.1, 3.1.1L-L4M-L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.29.1 and 24.2.1; ii) a light chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOS: 4, 12, 20, 28, 36, 44, 52, 60, 68, 76, 84, 94 and 100, or said amino acid sequence lacking a signal sequence; and iii) a light chain variable region encoded by a nucleic acid sequence selected from the group consisting of SEQ ID NOS: 3, 11, 19, 27, 35, 43,
51, 59, 67, 75, 83, 93 and 99, or said encoded light chain variable region lacking a signal sequence.
7. The antibody or antigen-binding portion thereof according to claim 1, wherein
(a) said heavy chain comprises an amino acid sequence selected from the group consisting of i) a heavy chain of an antibody selected from the group consisting of 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-V88A-V97A, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H- D16E, 23.29.1 and 24.2.1; ii) a heavy chain comprising an amino acid sequence selected from the group consisting of SEQ ID NOS: 6, 14, 22, 30, 38, 46, 54, 62,
70, 78 and 86, or said amino acid sequence lacking a signal sequence; and iii) a heavy chain encoded by a nucleic acid sequence selected from the group consisting of SEQ ID NOS: 5, 13, 21, 29, 37, 45, 53, 61, 69, 77 and 85, or said encoded heavy chain lacking a signal sequence; wherein said amino acid sequence may have up to six conservative amino acid substitutes and/or four non-conservative amino acid insertions, deletions or substitutions therefrom; or
(b) said light chain comprises the amino acid sequence selected from the group consisting of i) a light chain of an antibody selected from the group consisting of 3.1.1, 3.1.1L-L4M-L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K and 24.2.1; ii) a light chain comprising an amino acid sequence selected from the group consisting of SEQ ID NOS: 8, 16, 24, 32, 40, 48, 56, 64, 72, 80, 88 and 102, or said amino acid sequence lacking a signal sequence; and iii) a light chain encoded by a nucleic acid sequence selected from the group consisting of SEQ ID NOS: 7, 15, 23, 31, 39, 47, 55, 63,
71, 79, 87 and 101, or said encoded light chain lacking a signal sequence; wherein said amino acid sequence may have up to six conservative amino acid substitutes and/or four non-conservative amino acid insertions, deletions or substitutions therefrom in each ofthe CDR regions ofthe light chain.
8. The antibody or antigen-binding portion thereof according to claim 7, wherein
(a) said heavy chain comprises an amino acid sequence selected from the group consisting of i) a heavy chain of an antibody selected from the group consisting of 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-V88A-V97A, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1H- D16E, 23.29.1 and 24.2.1; ii) a heavy chain comprising an amino acid sequence selected from the group consisting of SEQ ID NOS: 6, 14, 22, 30, 38, 46, 54, 62,
70, 78, 86, 90, 92, 96 and 98, or said amino acid sequence lacking a signal sequence; and iii) a heavy chain encoded by a nucleic acid sequence selected from the group consisting of SEQ ID NOS: 5, 13, 21, 29, 37, 45, 53, 61, 69, 77, 85, 89, 91, 95 and 97, or said encoded heavy chain lacking a signal sequence; or
(b) said light chain comprises the amino acid sequence selected from the group consisting of i) a light chain of an antibody selected from the group consisting of 3.1.1, 3.1.1L-L4M-L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K and 24.2.1; ii) a light chain comprising an amino acid sequence selected from the group consisting of SEQ ID NOS: 8, 16, 24, 32, 40, 48, 56, 64, 72, 80, 88 and 102, or said amino acid sequence lacking a signal sequence; and iii) a light chain encoded by a nucleic acid sequence selected from the group consisting of SEQ J-D NOS: 7, 15, 23, 31, 39, 47, 55, 63,
71, 79, 87 and 101, or said encoded light chain lacking a signal sequence.
9. The antibody or antigen-binding portion thereof according to any one of claims 1-8, wherein said antibody or antigen-binding portion thereof comprises a heavy chain according to (a) and a light chain according to (b).
10. The antibody or antigen-binding portion thereof according to claim 1, wherein the antibody or portion thereof comprises a heavy chain and a light chain, and wherein the amino acid sequences ofthe heavy chain and light chain are selected from the group consisting of: a) the amino acid sequence ofthe heavy chain and the amino acid sequence ofthe light chain of 3.1.1; b) the amino acid sequence ofthe heavy chain and the amino acid sequence ofthe light chain of 7.1.2; c) the amino acid sequence ofthe heavy chain and the amino acid sequence ofthe light chain of 10.8.3; d) the amino acid sequence ofthe heavy chain and the amino acid sequence ofthe light chain of 15.1.1; e) the amino acid sequence ofthe heavy chain and the amino acid sequence ofthe light chain of 21.4.1; f) the amino acid sequence ofthe heavy chain and the amino acid sequence ofthe light chain of 21.2.1; g) the amino acid sequence ofthe heavy chain and the amino acid sequence ofthe light chain of 22.2.1; h) the amino acid sequence ofthe heavy chain and the amino acid sequence ofthe light chain of 22.1.1H-C109A; i) the amino acid sequence ofthe heavy chain and the amino acid sequence ofthe light chain of 23.5.1; j) the amino acid sequence ofthe heavy chain and the amino acid sequence ofthe light chain of 23.25.1; k) the amino acid sequence ofthe heavy chain and the amino acid sequence ofthe light chain of 23.28.1;
1) the amino acid sequence ofthe heavy chain and the amino acid sequence ofthe light chain of 23.28.1L-C92A; m) the amino acid sequence ofthe heavy chain and the amino acid sequence ofthe light chain of 23.28.1H-D16E; n) the amino acid sequence ofthe heavy chain and the amino acid sequence ofthe light chain of 23.29.1; o) the amino acid sequence ofthe heavy chain and the amino acid sequence ofthe light chain of 24.2.1; p) the amino acid sequence ofthe heavy chain and the amino acid sequence ofthe light chain of 3.1.1H-A78T; q) the amino acid sequence ofthe heavy chain and the amino acid sequence ofthe light chain of 3.1.1H-A78T-V88A-V97A; r) the amino acid sequence ofthe heavy chain and the amino acid sequence ofthe light chain of 3.1.1L-L4M-L83V; and s) the amino acid sequence ofthe heavy chain and the amino acid sequence ofthe light chain of 23.29.1L-R174K; wherein the amino acid sequences ofthe heavy and light chains may each have up to six conservative amino acid substitutions and/or four non- conservative amino acid insertions, deletions or substitutions therefrom .
11. The antibody or antigen-binding portion thereof according to claim 1 , wherein the antibody or portion thereof comprises a heavy chain and a light chain, and wherein the amino acid sequences ofthe heavy chain variable region of said heavy chain and the light chain variable region of said light chain are selected from the group consisting of: a) the amino acid sequence selected from the group consisting of SEQ ID NOS: 2, 90 and 92, and the amino acid sequence selected from the group consisting of SEQ ID NOS: 4 and 94; b) the amino acid sequence of SEQ ID NO: 10 and the amino acid sequence of SEQ ID NO: 12; c) the amino acid sequence of SEQ ID NO: 18 and the amino acid sequence of SEQ ID NO: 20; d) the amino acid sequence of SEQ ID NO: 26 and the amino acid sequence of SEQ ID NO: 28; e) the amino acid sequence of SEQ ID NO: 34 and the amino acid sequence of SEQ ID NO: 36; f) the amino acid sequence of SEQ ID NO: 42 and the amino acid sequence of SEQ ID NO: 44; g) the amino acid sequence selected from the group consisting of SEQ ID NOS: 50 and 96 and the amino acid sequence of SEQ ID NO: 52; h) the amino acid sequence of SEQ ID NO: 58 and the amino acid sequence of SEQ ID NO: 60; i) the amino acid sequence selected from the group consisting of SEQ ID NOS: 66 and 98, and the amino acid sequence selected from the group consisting of SEQ ID NOS: 68 and 100; and j) the amino acid sequence of SEQ ID NO: 74 and the amino acid sequence of SEQ ID NO: 78; wherein said amino acid sequences optionally lack a signal sequence and wherein the amino acid sequences ofthe heavy and light chains may each have up to six conservative amino acid substitutions and/or four non-conservative amino acid insertions, deletions or substitutions therefrom.
12. The antibody or antigen-binding portion thereof according to either of claims 10 or 11, wherein the antibody or portion thereof does not have any conservative amino acid substitutions or non-conservative amino acid insertions, deletions or substitutions therefrom.
13. The antibody according to claim 1, wherein the antibody is selected from the group consisting of 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-V88A-V97A, 3.1.1L-L4M, L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.28.1H-D16E, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K and 24.2.1.
14. The antibody or antigen-binding portion thereof according to any one of claims 1-13, wherein the antibody or portion thereof has at least one property selected from the group consisting of: a) does not bind to mouse, rat, dog and/or rabbit B cells; b) binds to human, cynomolgus and/or rhesus B cells; c) has a selectivity for CD40 that is at least 100 times greater than its selectivity for receptor activator of nuclear factor-kappa B (RANK), 4- IBB (CD 137), tumor necrosis factor receptor- 1 (TNFR-1) and tumor necrosis factor receptor-2 (TNFR-2); d) binds to CD40 with a KD of 4 x 10"10 M or less; e) has an off rate for CD40 of Koff of 2 x 10"4 or smaller; f) inhibits tumor growth in vivo in the presence of human T cells and/or human dendritic cells; g) inhibits the growth of CD40-positive tumors in the absence of human immune cells; h) increases expression of ICAM, MHC-II, B7-2, CD71, CD23 and/or CD71 on the surface of human B-cells; i) increases secretion of IL-12p40, IL-12p70 and/or IL-8 by human dendritic cells; j) increases expression of ICAM, MHC-II, B7-2 and/or CD83 on the surface of human dendritic cells; k) increases expression of interferon-gamma by human T cells during allogenic stimulation;
1) binds human CD40 in presence of human CD40L; m) binds to an epitope of human CD40 contained in domain 1 or domain 2 ofthe extracellular domain of CD40; and n) binds to an epitope of human CD40 contained in domain 2 or domain 3 ofthe extracellular domain of CD40.
15. An antibody or antigen-binding portion thereof that binds specifically to and activates human CD40, wherein the antibody or portion thereof has at least one property selected from the group consisting of: a) cross-competes for binding to CD40 with an antibody selected from the group consisting of 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-V88A-V97A, 3.1.1L-L4M, L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.28.1H-D16E, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K and 24.2.1; b) binds to the same epitope of CD40 as an antibody selected from the group consisting of 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-V88A-V97A, 3.1.1L- L4M, L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.28.1H-D16E, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K and 24.2.1; c) binds to CD40 with substantially the same KD as an antibody selected from the group consisting of 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-V88A- V97A, 3.1.1L-L4M, L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H- C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.28.1H-D16E, 23.28.1L- C92A, 23.29.1, 23.29.1L-R174K and 24.2.1; and d) binds to CD40 with substantially the same off rate as an antibody selected from the group consisting of 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T- V88A-V97A, 3.1.1L-L4M, L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.28.1H-D16E, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K and 24.2.1.
16. The antibody or antigen-binding portion thereof according to any one of claims 1-15 that is a) an immunoglobulin G (IgG), an IgM, an IgE, an IgA or an IgD molecule, or is derived therefrom; or b) an Fab fragment, an F(ab')2 fragment, an Fv fragment, a single chain antibody, a humanized antibody, a chimeric antibody or a bispecific antibody.
17. A pharmaceutical composition comprising the antibody or portion thereof according to any one of claims 1-16 and a pharmaceutically acceptable carrier.
18. A method of treating cancer in a human with an antibody or antigen- binding portion thereof that specifically binds to and activates human CD40, comprising the step of administering to the human an amount ofthe antibody effective to treat said cancer.
19. A method of treating a patient in need thereof with an anti-CD40 antibody or antigen-binding portion thereof, comprising the step of administering to the patient an effective amount ofthe antibody according to any one of claims 1- 16 or the pharmaceutical composition of claim 17.
20. A method of enhancing an immune response in a human in need thereof, comprising the step of administering to the patient an effective amount of the antibody or antigen-binding portion thereof according to any one of claims 1- 16 or the pharmaceutical composition of claim 17.
21. An isolated cell line that produces the antibody or said antigen binding portion thereof according to any one of claims 1-16 or the heavy chain or light chain of said antibody or said portion thereof.
22. The cell line according to claim 21 that produces an antibody selected from the group consisting of 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-V88A-V97A, 3.1.1L-L4M, L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.28.1H-D16E, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K and 24.2.1, or wherein the antibody has the same amino acid sequences thereof.
23. An isolated nucleic acid molecule that comprises a nucleic acid sequence that encodes the heavy chain or an antigen-binding portion thereof or the light chain or an antigen-binding portion thereof of an antibody or antigen-binding portion thereof according to any one of claims 1-16.
24. The isolated nucleic acid molecule according to claim 23, wherein the nucleic acid molecule comprises a nucleic acid sequence selected from the group consisting of: a) a nucleic acid sequence encoding the amino acid sequence ofthe heavy chain or the antigen-binding portion thereof of the antibody selected from the group consisting of 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-V88A-V97A, 3.1.1L- L4M, L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.28.1H-D16E, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K and 24.2.1; b) a nucleic acid sequence encoding the amino acid sequence ofthe light chain or the antigen-binding portion thereof of the antibody selected from the group consisting of 3.1.1, 3.1.1H-A78T, 3.1.1H-A78T-V88A-V97A, 3.1.1L-L4M, L83V, 7.1.2, 10.8.3, 15.1.1, 21.4.1, 21.2.1, 22.1.1, 22.1.1H-C109A, 23.5.1, 23.25.1, 23.28.1, 23.28.1L-C92A, 23.28.1H-D16E, 23.28.1L-C92A, 23.29.1, 23.29.1L-R174K and 24.2.1; c) a nucleic acid sequence encoding the amino acid sequence of selected from the group consisting of SEQ TD NOS: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100 and 102, or said amino acid sequence lacking a signal sequence; and d) a nucleic acid sequence selected from the group consisting of SEQ ID NOS: 1, 3, 5, ,7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 81, 83, 85, 87, 89, 91, 93, 95, 97, 99 and 101.
25. A vector comprising the nucleic acid molecule according to either of claims 23 or 24, wherein the vector optionally comprises an expression control sequence operably linked to the nucleic acid molecule.
26. A host cell comprising the vector according to claim 25 or the nucleic acid molecule according to either of claims 23 or 24.
27. A method ofmaking an anti-CD40 antibody or antigen-binding portion thereof, comprising culturing the host cell according to claim 26 or the cell line according to claim 21 under suitable conditions and recovering said antibody or antigen-binding portion.
28. A non-human transgenic animal or transgenic plant comprising the nucleic acid according to either of claims 23 or 24, wherein the non-human transgenic animal or transgenic plant expresses said nucleic acid.
29. A method ofmaking an antibody or antigen-binding portion thereof that specifically binds to human CD40, comprising the step of isolating the antibody from the non-human transgenic animal or transgenic plant according to claim 28.
30. A method of treating a subject in need thereof with an antibody or antigen-binding portion thereof that specifically binds to human CD40, comprising the steps of
(a) administering an effective amount of an isolated nucleic acid molecule encoding the heavy chain or the antigen-binding portion thereof, an isolated nucleic acid molecule encoding the light chain or the antigen-binding portion thereof, or both the nucleic acid molecules encoding the light chain and the heavy chain or antigen-binding portions thereof; and
(b) expressing the nucleic acid molecule.
31. A chimeric or human monoclonal antibody or antigen-binding portion thereof that specifically binds to and activates human CD40, wherein the antibody or portion thereof comprises a heavy chain and a light chain, and wherein the amino acid sequences ofthe heavy chain and the light chain are selected from the group consisting of: a) the amino acid sequence of SEQ ID NO: 6, and the amino acid sequence of SEQ ID NO: 8; b) the amino acid sequence of SEQ ID NO: 14, and the amino acid sequence of SEQ ID NO: 16; c) the amino acid sequence of SEQ ID NO: 22, and the amino acid sequence of SEQ ID NO: 24; d) the amino acid sequence of SEQ ID NO: 30, and the amino acid sequence of SEQ ID NO: 32; and e) the amino acid sequence of SEQ ID NO: 46, and the amino acid sequence of SEQ ID NO: 48.
PCT/US2002/036107 2001-11-09 2002-11-08 Antibodies to cd40 WO2003040170A2 (en)

Priority Applications (24)

Application Number Priority Date Filing Date Title
NZ533180A NZ533180A (en) 2001-11-09 2002-11-08 A monoclonal antibody or antigen binding portion that specifically binds to and activates human CD40
APAP/P/2004/003034A AP1918A (en) 2001-11-09 2002-11-08 Antibodies to CD40
EA200400654A EA011449B1 (en) 2001-11-09 2002-11-08 Human monoclonal antibodies to cd40 and methods for use thereof
AU2002356926A AU2002356926B2 (en) 2001-11-09 2002-11-08 Antibodies to CD40
JP2003542215A JP4616555B2 (en) 2001-11-09 2002-11-08 Antibody against CD40
CA2466128A CA2466128C (en) 2001-11-09 2002-11-08 Antibodies to cd40
BRPI0214137A BRPI0214137B8 (en) 2001-11-09 2002-11-08 human monoclonal antibody or antigen-binding portion thereof that specifically binds to and activates human cd40, isolated hybridoma cell line, pharmaceutical composition comprising the same, use thereof in the manufacture of a drug and method of manufacturing said antibodies or antigen-binding portion of it
DK02802898.3T DK1476185T3 (en) 2001-11-09 2002-11-08 Antibodies to CD40
SI200231036T SI1476185T1 (en) 2001-11-09 2002-11-08 Antibodies to cd40
ES02802898T ES2432968T3 (en) 2001-11-09 2002-11-08 Antibodies against CD40
HU0402247A HU229829B1 (en) 2001-11-09 2002-11-08 Antibodies to cd40
CN028245709A CN1582165B (en) 2001-11-09 2002-11-08 Antibodies to CD40
IL16182302A IL161823A0 (en) 2001-11-09 2002-11-08 Antibodies to cd40 and pharmaceutical compositionscontaining the same
MEP-2008-814A ME00503B (en) 2001-11-09 2002-11-08 Antibodies to cd40
EP02802898.3A EP1476185B1 (en) 2001-11-09 2002-11-08 Antibodies to cd40
MXPA04004467A MXPA04004467A (en) 2001-11-09 2002-11-08 Antibodies to cd40.
YU39204A RS52484B (en) 2001-11-09 2002-11-08 Antibodies to cd40
TNP2004000078A TNSN04078A1 (en) 2001-11-09 2004-05-05 ANTIBODIES TO CD40
IL161823A IL161823A (en) 2001-11-09 2004-05-06 Antibodies to cd40 and pharmaceutical compositions containing the same
IS7253A IS2940B (en) 2001-11-09 2004-05-06 Antibodies against CD40
ZA2004/04207A ZA200404207B (en) 2001-11-09 2004-05-28 Antibodies to cd40
HRP20040525AA HRP20040525B1 (en) 2001-11-09 2004-06-08 Antibodies to cd40
NO20042388A NO334339B1 (en) 2001-11-09 2004-06-08 Antibody to CD40
HK05102440.7A HK1069122A1 (en) 2001-11-09 2005-03-22 Antibodies to cd40 cd40

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US34898001P 2001-11-09 2001-11-09
US60/348,980 2001-11-09

Publications (3)

Publication Number Publication Date
WO2003040170A2 WO2003040170A2 (en) 2003-05-15
WO2003040170A3 WO2003040170A3 (en) 2003-10-09
WO2003040170A9 true WO2003040170A9 (en) 2004-06-10

Family

ID=23370387

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2002/036107 WO2003040170A2 (en) 2001-11-09 2002-11-08 Antibodies to cd40

Country Status (47)

Country Link
US (9) US7288251B2 (en)
EP (2) EP2343086A3 (en)
JP (3) JP4616555B2 (en)
KR (1) KR100830086B1 (en)
CN (2) CN103450360B (en)
AP (1) AP1918A (en)
AR (1) AR039067A1 (en)
AU (1) AU2002356926B2 (en)
BR (1) BRPI0214137B8 (en)
CA (1) CA2466128C (en)
CL (1) CL2009001779A1 (en)
CR (2) CR7343A (en)
CU (1) CU23745A3 (en)
CY (1) CY1115675T1 (en)
DK (1) DK1476185T3 (en)
DO (1) DOP2002000507A (en)
EA (1) EA011449B1 (en)
EC (1) ECSP045093A (en)
EG (1) EG25801A (en)
ES (1) ES2432968T3 (en)
GE (1) GEP20074222B (en)
GT (1) GT200200228A (en)
HK (1) HK1069122A1 (en)
HN (1) HN2002000318A (en)
HR (1) HRP20040525B1 (en)
HU (1) HU229829B1 (en)
IL (2) IL161823A0 (en)
IS (1) IS2940B (en)
MA (1) MA27139A1 (en)
ME (1) ME00503B (en)
MX (1) MXPA04004467A (en)
MY (1) MY137641A (en)
NI (1) NI200200128A (en)
NO (1) NO334339B1 (en)
NZ (1) NZ533180A (en)
OA (1) OA12725A (en)
PA (1) PA8557701A1 (en)
PE (1) PE20030846A1 (en)
PL (2) PL214634B1 (en)
PT (1) PT1476185E (en)
RS (1) RS52484B (en)
SI (1) SI1476185T1 (en)
TN (1) TNSN04078A1 (en)
TW (2) TWI331040B (en)
UY (1) UY27537A1 (en)
WO (1) WO2003040170A2 (en)
ZA (1) ZA200404207B (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9107906B1 (en) 2014-10-28 2015-08-18 Adma Biologics, Inc. Compositions and methods for the treatment of immunodeficiency
US9221915B2 (en) 2005-09-07 2015-12-29 Pfizer Inc. Human monoclonal antibodies to activin receptor-like kinase-1
USRE45847E1 (en) 2004-01-09 2016-01-19 Pfizer Inc. Antibodies to MAdCAM

Families Citing this family (236)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7244827B2 (en) * 2000-04-12 2007-07-17 Agensys, Inc. Nucleic acid and corresponding protein entitled 24P4C12 useful in treatment and detection of cancer
US6943235B1 (en) * 1999-04-12 2005-09-13 Agensys, Inc. Transmembrane protein expressed in prostate cancer
US20050255106A1 (en) * 1999-05-22 2005-11-17 Linda Diehl Induction of anti-tumor ctl immunity through in vivo triggering of 4-1bb and/or cd40
US20030059427A1 (en) * 2000-04-28 2003-03-27 Force Walker R. Isolation and characterization of highly active anti-CD40 antibody
WO2002020619A2 (en) 2000-09-07 2002-03-14 Schreiber John R HUMAN ANTIBODIES AGAINST PSEUDOMONAS AERUGINOSA LPS DERIVED FROM TRANSGENIC XENOMOUSE$m(3)
PT1391464E (en) 2001-04-27 2007-11-15 Kirin Pharma Kk Anti-cd40 monoclonal antibody
US7658924B2 (en) * 2001-10-11 2010-02-09 Amgen Inc. Angiopoietin-2 specific binding agents
AR039067A1 (en) * 2001-11-09 2005-02-09 Pfizer Prod Inc ANTIBODIES FOR CD40
US20040185040A1 (en) 2001-11-21 2004-09-23 Celltech R & D Limited Modulating immune responses
WO2003045318A2 (en) * 2001-11-21 2003-06-05 Celltech R & D, Inc. Manipulation of cytokine levels using cd83 gene products
ATE546150T1 (en) * 2002-06-07 2012-03-15 Zymogenetics Inc USE OF IL-21 AND MONOCLONAL ANTIBODIES TO TREAT SOLID TUMORS
US7052694B2 (en) * 2002-07-16 2006-05-30 Mayo Foundation For Medical Education And Research Dendritic cell potentiation
HN2004000285A (en) 2003-08-04 2006-04-27 Pfizer Prod Inc ANTIBODIES DIRECTED TO c-MET
EP1680140B1 (en) * 2003-10-16 2011-04-20 Imclone LLC Fibroblast growth factor receptor-1 inhibitors and methods of treatment thereof
DK1694360T3 (en) * 2003-11-04 2010-10-18 Novartis Vaccines & Diagnostic Use of antagonist anti-CD40 antibodies to treat autoimmune and inflammatory diseases and organ transplant rejection
US8277810B2 (en) * 2003-11-04 2012-10-02 Novartis Vaccines & Diagnostics, Inc. Antagonist anti-CD40 antibodies
CA2548015A1 (en) 2003-12-05 2005-06-23 John R. Schreiber Human anti-pseudomonas-aeruginosa antibodies derived from transgenic xenomouse
US20050136055A1 (en) * 2003-12-22 2005-06-23 Pfizer Inc CD40 antibody formulation and methods
PL1707627T3 (en) 2003-12-25 2013-04-30 Kyowa Hakko Kirin Co Ltd Antagonistic anti-CD40 antibody mutant
NZ550518A (en) 2004-03-23 2009-11-27 Biogen Idec Inc Agents that cross link TNF receptors for treating cancer
US20060099203A1 (en) * 2004-11-05 2006-05-11 Pease Larry R B7-DC binding antibody
JP5902367B2 (en) * 2004-06-30 2016-04-13 メイヨ・ファウンデーション・フォー・メディカル・エデュケーション・アンド・リサーチ antibody
CA2601417C (en) 2004-07-01 2018-10-30 Novo Nordisk A/S Human anti-kir antibodies
US20070286855A1 (en) * 2004-08-03 2007-12-13 Mayo Foundation For Medical Education And Research Improving treatments
US7563443B2 (en) * 2004-09-17 2009-07-21 Domantis Limited Monovalent anti-CD40L antibody polypeptides and compositions thereof
DK1836225T3 (en) 2005-01-06 2012-02-27 Innate Pharma Sas Kir-binding agents and methods for using them
US8716451B2 (en) 2005-01-12 2014-05-06 Kyowa Hakko Kirin Co., Ltd Stabilized human IgG2 and IgG3 antibodies
CA2602375C (en) * 2005-03-23 2018-07-24 Genmab A/S Antibodies against cd38 for treatment of multiple myeloma
EP2444421A1 (en) 2005-04-26 2012-04-25 Pfizer Inc. P-Cadherin antibodies
ES2429564T3 (en) * 2005-05-18 2013-11-15 Novartis Ag Procedures for the diagnosis and treatment of diseases that have an autoimmune and / or inflammatory component
US8337851B2 (en) * 2005-05-18 2012-12-25 Novartis Ag Methods of monitoring the efficacy of anti-CD40 antibodies in treating a subject for a CD40-expressing cancer
GB0512225D0 (en) * 2005-06-16 2005-07-27 Univ Sheffield Immunoglobulin molecules
AU2006269940C1 (en) 2005-07-18 2013-11-07 Seagen Inc. Beta-glucuronide-linker drug conjugates
WO2007103048A2 (en) * 2006-03-01 2007-09-13 Regents Of The University Of Colorado Tlr agonist (flagellin)/cd40 agonist/antigen protein and dna conjugates and use thereof for inducing synergistic enhancement in immunity
AU2007248628B2 (en) 2006-05-03 2013-02-07 The Regents Of The University Of Colorado, A Body Corporate CD40 agonist antibody/type1 interferon synergistic adjuvant combination, conjugates containing and use thereof as a therapeutic to enhance cellular immunity
JP4383525B2 (en) 2006-05-09 2009-12-16 ファイザー・プロダクツ・インク Cycloalkylamino acid derivatives and pharmaceutical compositions thereof
EP1854810A1 (en) * 2006-05-09 2007-11-14 PanGenetics B.V. Deimmunized antagonistic anti-human CD40 monoclonal antibody from the ch5D12 antibody
WO2007131575A1 (en) 2006-05-12 2007-11-22 Fondazione Centro San Raffaele Del Monte Tabor Tolerogenic dendritic cells, method for their production and uses thereof
US20090074711A1 (en) * 2006-09-07 2009-03-19 University Of Southhampton Human therapies using chimeric agonistic anti-human cd40 antibody
WO2008072723A1 (en) * 2006-12-14 2008-06-19 Forerunner Pharma Research Co., Ltd. ANTI-Claudin-3 MONOCLONAL ANTIBODY, AND TREATMENT AND DIAGNOSIS OF CANCER USING THE SAME
PT2059534E (en) 2007-02-23 2012-07-17 Schering Corp Engineered anti-il-23p19 antibodies
JP2010518858A (en) 2007-02-23 2010-06-03 シェーリング コーポレイション Engineered anti-IL-23P19 antibody
AU2008260498B2 (en) 2007-05-30 2012-11-29 Xencor, Inc. Methods and compositions for inhibiting CD32b expressing cells
EP1997830A1 (en) * 2007-06-01 2008-12-03 AIMM Therapeutics B.V. RSV specific binding molecules and means for producing them
US8039597B2 (en) * 2007-09-07 2011-10-18 Agensys, Inc. Antibodies and related molecules that bind to 24P4C12 proteins
AU2008323701B2 (en) 2007-11-07 2015-03-26 Genentech, Inc Methods and compositions for assessing responsiveness of B-cell lymphoma to treatment with anti-CD40 antibodies
EP2594590B1 (en) 2007-12-14 2014-11-12 Bristol-Myers Squibb Company Method of producing binding molecules for the human OX40 receptor
JO2913B1 (en) * 2008-02-20 2015-09-15 امجين إنك, Antibodies directed to angiopoietin-1 and angiopoietin-2 and uses thereof
ATE542813T1 (en) 2008-08-06 2012-02-15 Pfizer 6-SUBSTITUTED 2-HETEROCYCLYLAMINOPYRAZINE COMPOUNDS AS CHK-1 INHIBITORS
JP2012501323A (en) * 2008-08-29 2012-01-19 アカデミシュ ジーケンハウス ライデン ハー.オー.デー.エン. ルムク Delivery of a CD40 agonist to a subject's tumor draining lymph nodes
EP2198878A1 (en) 2008-12-18 2010-06-23 University Of Miami Polypeptide bombesin antagonists
JO3382B1 (en) 2008-12-23 2019-03-13 Amgen Inc Human cgrp receptor binding antibodies
AU2016244220B2 (en) * 2008-12-23 2018-05-17 Amgen Inc. Human CGRP receptor binding proteins
AU2010229192A1 (en) * 2009-03-06 2011-09-29 Agensys, Inc. Antibody drug conjugates (ADC) that bind to 24P4C12 proteins
JP5883653B2 (en) 2009-03-10 2016-03-15 ベイラー リサーチ インスティテュートBaylor Research Institute Antigen-presenting cell targeting antiviral vaccine
CN108373509A (en) * 2009-03-10 2018-08-07 贝勒研究院 The antiviral vaccine of target antigen presenting cells
CN102448989B (en) * 2009-03-10 2015-09-30 贝勒研究院 Anti-CD 40 antibodies and uses thereof
MX2011010938A (en) 2009-04-18 2012-01-12 Genentech Inc Methods for assessing responsiveness of b-cell lymphoma to treatment with anti-cd40 antibodies.
WO2010123012A1 (en) * 2009-04-20 2010-10-28 協和発酵キリン株式会社 Antibody containing igg2 having amino acid mutation introduced therein
TW201138843A (en) 2009-12-18 2011-11-16 Colgate Palmolive Co Biguanide preservation of precipitated calcium carbonate
WO2011083391A2 (en) 2010-01-05 2011-07-14 Pfizer Inc. Biomarkers for anti-igf-ir cancer therapy
ES2807217T3 (en) 2010-03-31 2021-02-22 Boehringer Ingelheim Int Anti-CD40 antibodies
GB201006096D0 (en) * 2010-04-13 2010-05-26 Alligator Bioscience Ab Novel compositions and uses thereof
FR2959994B1 (en) 2010-05-12 2012-08-24 Lfb Biotechnologies NOVEL 12G4 MUTUS HUMANIZED ANTIBODIES AND THEIR FRAGMENTS DIRECTED AGAINST THE HUMAN TYPE II ANTI-MULLERIAN HORMONE RECEPTOR
EP3441404A1 (en) 2010-09-09 2019-02-13 Pfizer Inc 4-1bb binding molecules
AU2011310887A1 (en) * 2010-09-29 2013-05-02 Universite De Liege Combination of an agonistic anti-CD40 monoclonal antibody or a CD40 ligand and inactivated or attenuated bacteria for use in the treatment and/or prevention of mastitis
AR083847A1 (en) 2010-11-15 2013-03-27 Novartis Ag FC VARIANTS (CONSTANT FRAGMENT) SILENCERS OF ANTI-CD40 ANTIBODIES
JP2014500879A (en) * 2010-11-16 2014-01-16 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Factors and methods for treating diseases correlated with BCMA expression
CN103347898B (en) * 2011-01-10 2017-12-05 Ct大西洋有限公司 Including the therapeutic alliance with tumor associated antigen binding antibody
GB201103578D0 (en) 2011-03-02 2011-04-13 Sabrepharm Ltd Dipyridinium derivatives
PT2683406T (en) 2011-03-11 2019-07-08 Beth Israel Deaconess Medical Ct Inc Anti-cd40 antibodies and uses thereof
CA2830972C (en) 2011-04-19 2018-11-20 Pfizer Inc. Combinations of anti-4-1bb antibodies and adcc-inducing antibodies for the treatment of cancer
JP6152090B2 (en) * 2011-04-21 2017-06-21 ザ リージェンツ オブ ザ ユニバーシティ オブ コロラド,ア ボディー コーポレイトTHE REGENTS OF THE UNIVERSITY OF COLORADO,a body corporate Compositions and methods for treating optic neuritis
TWI598363B (en) 2011-04-21 2017-09-11 必治妥美雅史谷比公司 Antibody polypeptides that antagonize cd40
CN106928362B (en) 2011-04-29 2021-10-26 埃派斯进有限公司 anti-CD 40 antibodies and methods of use thereof
EP2710042A2 (en) 2011-05-16 2014-03-26 Fabion Pharmaceuticals, Inc. Multi-specific fab fusion proteins and methods of use
GB201115280D0 (en) * 2011-09-05 2011-10-19 Alligator Bioscience Ab Antibodies, uses and methods
GB201116092D0 (en) 2011-09-16 2011-11-02 Bioceros B V Antibodies and uses thereof
AP2014007588A0 (en) 2011-09-22 2014-04-30 Amgen Inc CD27L antigen binding proteins
CN104203982B (en) 2011-10-28 2018-08-31 特瓦制药澳大利亚私人有限公司 Polypeptide construct and application thereof
WO2013091661A2 (en) * 2011-12-23 2013-06-27 Aarhus Universitet Proteolytic resistant protein affinity tag
RS62454B1 (en) * 2012-01-31 2021-11-30 Regeneron Pharma Anti-asic1 antibodies and uses thereof
JP5798199B2 (en) * 2012-03-08 2015-10-21 国立研究開発法人科学技術振興機構 Anti-cancer agent
AR090263A1 (en) * 2012-03-08 2014-10-29 Hoffmann La Roche COMBINED ANTIBODY THERAPY AGAINST HUMAN CSF-1R AND USES OF THE SAME
RU2737765C2 (en) 2012-05-04 2020-12-02 Пфайзер Инк. Prostate-associated antigens and immunotherapeutic regimens based on vaccines
US20140004131A1 (en) 2012-05-04 2014-01-02 Novartis Ag Antibody formulation
ES2873877T3 (en) 2012-06-18 2021-11-04 Ospedale San Raffaele Srl Compositions and methods to decrease an immune response
CN109265552A (en) * 2012-10-30 2019-01-25 埃派斯进有限公司 Anti-CD 40 antibodies and its application method
UY35148A (en) 2012-11-21 2014-05-30 Amgen Inc HETERODIMERIC IMMUNOGLOBULINS
CA2899960C (en) * 2013-02-01 2022-05-03 Transbio Ltd Anti-cd83 antibodies and use thereof
US9708375B2 (en) 2013-03-15 2017-07-18 Amgen Inc. Inhibitory polypeptides specific to WNT inhibitors
JP2016515544A (en) 2013-03-18 2016-05-30 バイオセルオーエックス プロダクツ ビー.ブイ. Humanized anti-CD134 (OX40) antibody and use thereof
MX370377B (en) 2013-04-29 2019-12-11 Teva Pharmaceuticals Australia Pty Ltd Anti-cd38 antibodies and fusions to attenuated interferon alpha-2b.
US11117975B2 (en) 2013-04-29 2021-09-14 Teva Pharmaceuticals Australia Pty Ltd Anti-CD38 antibodies and fusions to attenuated interferon alpha-2B
WO2014202552A1 (en) 2013-06-17 2014-12-24 Koninklijke Philips N.V. Magnetic resonance imaging subject support
GB201311487D0 (en) * 2013-06-27 2013-08-14 Alligator Bioscience Ab Bispecific molecules
SI3444271T1 (en) 2013-08-08 2022-03-31 Cytune Pharma Il-15 and il-15ralpha sushi domain based modulokines
CA2923145A1 (en) 2013-09-05 2015-03-12 Amgen Inc. Fc-containing molecules exhibiting predictable, consistent, and reproducible glycoform profiles
AR097584A1 (en) 2013-09-12 2016-03-23 Hoffmann La Roche ANTIBODY COMBINATION THERAPY AGAINST HUMAN CSF-1R AND ANTIBODIES AGAINST HUMAN PD-L1
SG10201804030WA (en) 2013-10-25 2018-07-30 Psioxus Therapeutics Ltd Oncolytic adenoviruses armed with heterologous genes
GB201322583D0 (en) * 2013-12-19 2014-02-05 Alligator Bioscience Ab Antibodies
SG11201604594SA (en) * 2013-12-20 2016-07-28 Hoffmann La Roche Combination therapy with an anti-ang2 antibody and a cd40 agonist
US10286058B2 (en) 2014-01-13 2019-05-14 Baylor Research Institute Vaccines against HPV and HPV-related diseases
US10435475B2 (en) 2014-03-07 2019-10-08 Bristol-Myers Squibb Company Method of using antibody polypeptides that antagonize CD40 to treat IBD
CA2943834A1 (en) 2014-03-31 2015-10-08 Genentech, Inc. Combination therapy comprising anti-angiogenesis agents and ox40 binding agonists
BR112016024546A2 (en) * 2014-04-30 2018-01-23 Max-Delbrück-Centrum Für Molekulare Medizin In Der Helmholtz-Gemeinschaft antibody or antibody fragment, antibody or isolated antibody fragment, antibody-drug conjugate, nucleic acid molecule, host cell, and pharmaceutical composition
UA119352C2 (en) 2014-05-01 2019-06-10 Тева Фармасьютикалз Острейліа Пті Лтд Combination of lenalidomide or pomalidomide and cd38 antibody-attenuated interferon-alpha constructs, and the use thereof
WO2015175861A1 (en) 2014-05-16 2015-11-19 Amgen Inc. Assay for detecting th1 and th2 cell populations
GB201413665D0 (en) 2014-07-03 2014-09-17 Transimmune Ag And Yale University Method for obtaining globally activated monocytes
KR102443258B1 (en) 2014-08-12 2022-09-14 엘리게이터 바이오사이언스 에이비 Combination therapies with anti cd40 antibodies
BR112016029334A2 (en) * 2014-08-14 2018-01-09 Hoffmann La Roche pharmaceutical product, use of an antibody, kit, method for treating a cancer patient and methods and uses of new products
EP3070102A1 (en) 2015-03-18 2016-09-21 F. Hoffmann-La Roche AG Combination therapy of antibodies human cd40 activating antibodies and anti human pld-1 antibodies
US10934362B2 (en) 2014-09-15 2021-03-02 Amgen Inc. Bi-specific anti-CGRP receptor/PAC1 receptor antigen binding proteins and uses thereof
NZ731491A (en) 2014-10-23 2021-12-24 Kira Biotech Pty Ltd Cd83 binding proteins and uses thereof
EA036498B1 (en) 2014-10-29 2020-11-17 Сиэтл Дженетикс, Инк. Dosage and administration of non-fucosylated anti-cd40 antibodies
MX2017005481A (en) 2014-10-29 2017-10-26 Teva Pharmaceuticals Australia Pty Ltd Interferon a2b variants.
TWI595006B (en) 2014-12-09 2017-08-11 禮納特神經系統科學公司 Anti-pd-1 antibodies and methods of use thereof
PT3283527T (en) 2015-04-13 2021-03-03 Five Prime Therapeutics Inc Combination therapy for cancer
JOP20200116A1 (en) 2015-04-24 2017-06-16 Amgen Inc Methods for treating or preventing migraine headache
BR122018004815A2 (en) 2015-04-30 2019-09-10 Psioxus Therapeutics Ltd oncolytic adenovirus encoding protein b7
CA2985718A1 (en) 2015-06-24 2016-12-29 F. Hoffmann-La Roche Ag Anti-transferrin receptor antibodies with tailored affinity
CN107771184B (en) * 2015-06-29 2022-11-01 百时美施贵宝公司 Antibodies against CD40
KR20180021833A (en) * 2015-06-29 2018-03-05 더 락커펠러 유니버시티 Antibodies to CD40 with enhanced agonist activity
CN111375066B (en) 2015-07-16 2023-04-25 百欧肯治疗有限公司 Compositions and methods for treating cancer
EP3307322B1 (en) 2015-09-04 2021-02-17 Primatope Therapeutics Inc. Humanized anti-cd40 antibodies and uses thereof
ES2906823T3 (en) 2015-09-30 2022-04-20 Janssen Biotech Inc Agonist antibodies that specifically bind to human CD40 and methods of use
AR106189A1 (en) 2015-10-02 2017-12-20 Hoffmann La Roche BIESPECTIFIC ANTIBODIES AGAINST HUMAN A-b AND THE HUMAN TRANSFERRINE RECEIVER AND METHODS OF USE
MA43023A (en) 2015-10-02 2018-08-08 Hoffmann La Roche HUMAN TRANSFERRIN / ANTI-HUMAN BISPECIFIC CD20 / ANTI-HUMAN RECEPTOR ANTIBODIES AND METHODS FOR USE THEREOF
GB2543550A (en) 2015-10-21 2017-04-26 Hox Therapeutics Ltd Peptides
KR20180072821A (en) 2015-11-03 2018-06-29 얀센 바이오테크 인코포레이티드 Antibodies that specifically bind to PD-1 and uses thereof
EP3757119A1 (en) * 2015-11-16 2020-12-30 Ubiprotein, Corp. A method for extending half-life of a protein
EP3386536A4 (en) * 2015-12-07 2019-07-31 Opi Vi- IP Holdco LLC Composition of antibody construct-agonist conjugates and methods of use thereof
JP7064437B2 (en) 2015-12-17 2022-05-10 サイオクサス セラピューティクス リミテッド Group B adenovirus encoding an anti-TCR complex antibody or fragment
MX2018010672A (en) 2016-03-04 2019-05-27 Univ Rockefeller Antibodies to cd40 with enhanced agonist activity.
WO2017156349A1 (en) 2016-03-10 2017-09-14 Cold Genesys, Inc. Methods of treating solid or lymphatic tumors by combination therapy
WO2017157305A1 (en) 2016-03-15 2017-09-21 Generon (Shanghai) Corporation Ltd. Multispecific fab fusion proteins and use thereof
CN109069638B (en) 2016-03-24 2022-03-29 璟尚生物制药公司 Trispecific inhibitors for cancer therapy
MA43835A (en) 2016-03-25 2018-11-28 Seattle Genetics Inc PROCESS FOR THE PREPARATION OF PEGYLATED MEDICINAL PRODUCTS AND THEIR INTERMEDIARIES
US20170342169A1 (en) 2016-05-27 2017-11-30 Abbvie Biotherapeutics Inc. Bispecific binding proteins
IL263223B2 (en) 2016-05-27 2023-03-01 Abbvie Biotherapeutics Inc Anti-cd40 antibodies and their uses
CA3026880A1 (en) 2016-06-08 2017-12-14 Paul Foster Treatment of igg4-related diseases with anti-cd19 antibodies crossbinding to cd32b
EP3470424A4 (en) 2016-06-08 2020-03-04 Shanghai Jiaotong University School of Medicine Sequence of antibody heavy chain constant region for enhancing agonist antibody activity
BR112018076260A2 (en) 2016-06-20 2019-03-26 Kymab Limited antibody or fragment thereof that specifically binds to hpd-11, bispecific antibody or fusion protein, use of an antibody or fragment, method, pharmaceutical composition, modulation method, inhibition method, treatment method, nucleic acid, vector, host and immunocytocin
BR112019000512A2 (en) 2016-07-14 2019-04-24 Genmab A/S antibody, nucleic acid, expression vector, host cell, composition, methods of treating a disease, to produce a bispecific antibody and to detect if cross-linking between cd40 and cd137 expressing cells occurs in a sample, use of a multispecific antibody and kit
JP7241677B2 (en) 2016-07-19 2023-03-17 テバ・ファーマシューティカルズ・オーストラリア・ピーティワイ・リミテッド Anti-CD47 combination therapy
CN109843916B (en) 2016-08-12 2023-10-31 詹森生物科技公司 Fc-engineered anti-TNFR superfamily member antibodies with enhanced agonistic activity and methods of use thereof
CA3033661A1 (en) 2016-08-12 2018-02-15 Janssen Biotech, Inc. Engineered antibodies and other fc-domain containing molecules with enhanced agonism and effector functions
WO2018036852A1 (en) 2016-08-25 2018-03-01 F. Hoffmann-La Roche Ag Intermittent dosing of an anti-csf-1r antibody in combination with macrophage activating agent
GB201713765D0 (en) 2017-08-28 2017-10-11 Psioxus Therapeutics Ltd Modified adenovirus
US11905331B2 (en) 2016-11-11 2024-02-20 Kumho Ht, Inc. Antibody binding specifically to CD40 and use thereof
KR20190095941A (en) 2016-12-19 2019-08-16 그렌마크 파머수티칼스 에스. 아. New TNFR Agonists and Their Uses
CN110072553B (en) 2016-12-22 2023-09-15 豪夫迈·罗氏有限公司 Treatment of tumors with anti-CSF-1R antibodies in combination with anti-PD-L1 antibodies after failure of anti-PD-L1/PD 1 treatment
US10537637B2 (en) 2017-01-05 2020-01-21 Gensun Biopharma Inc. Checkpoint regulator antagonists
CA3049842A1 (en) * 2017-02-02 2018-08-09 Silverback Therapeutics, Inc. Construct-peptide compositions and methods of use thereof
KR20190115057A (en) 2017-02-10 2019-10-10 젠맵 비. 브이 Polypeptide Variants and Uses thereof
CA3056011A1 (en) 2017-03-14 2018-09-20 Amgen Inc. Control of total afucosylated glycoforms of antibodies produced in cell culture
US10259865B2 (en) 2017-03-15 2019-04-16 Adma Biologics, Inc. Anti-pneumococcal hyperimmune globulin for the treatment and prevention of pneumococcal infection
JP2020512312A (en) 2017-03-24 2020-04-23 シアトル ジェネティックス, インコーポレイテッド Process for the preparation of glucuronide drug-linker and its intermediates
CA3055433A1 (en) * 2017-03-29 2018-10-04 Glycotope Gmbh Humanized anti-cd40 antibodies
EP3604384B1 (en) 2017-03-30 2021-09-08 NOF Corporation Hydrophilic polymer derivative having self-immolative acetal linker and composite using same
EP3604385A4 (en) 2017-03-30 2021-01-20 NOF Corporation Heterobifunctional monodispersed polyethylene glycol, and complex using same
TWI704158B (en) * 2017-04-04 2020-09-11 瑞士商赫孚孟拉羅股份公司 Novel bispecific antigen binding molecules capable of specific binding to cd40 and to fap
CN110505883A (en) 2017-04-13 2019-11-26 豪夫迈·罗氏有限公司 Proleulzin immunoconjugates used in method for treating cancer, CD40 agonist, and optionally PD-1 axis binding antagonists
WO2018220207A1 (en) 2017-06-01 2018-12-06 Psioxus Therapeutics Limited Oncolytic virus and method
JP7257971B6 (en) 2017-06-01 2023-07-24 江▲蘇▼恒瑞医▲薬▼股▲フン▼有限公司 Anti-CD40 Antibodies, Antigen-Binding Fragments Thereof, and Medical Uses Thereof
TW201902462A (en) 2017-06-02 2019-01-16 瑞士商赫孚孟拉羅股份公司 Novel administration routes for immune agonists
EP3418302A1 (en) 2017-06-19 2018-12-26 F. Hoffmann-La Roche AG Administration routes for immune agonists
CU20200002A7 (en) 2017-07-14 2020-11-30 Pfizer ANTIBODIES AGAINST MADCAM
CN111511762A (en) 2017-08-21 2020-08-07 天演药业公司 anti-CD137 molecules and uses thereof
SG11202002366VA (en) 2017-09-19 2020-04-29 Mab Discovery Gmbh Agonistic cd40 antibodies
US10610585B2 (en) 2017-09-26 2020-04-07 Inserm (Institut National De La Sante Et De La Recherche Medicale) Methods and compositions for treating and preventing HIV
EP3708589A4 (en) 2017-11-08 2021-08-11 Kyowa Kirin Co., Ltd. BISPECIFIC ANTIBODY WHICH BINDS TO CD40 AND EpCAM
CN111788227A (en) 2017-12-27 2020-10-16 百时美施贵宝公司 anti-CD 40 antibodies and uses thereof
CN109971723B (en) * 2017-12-28 2023-07-07 上海细胞治疗研究院 T cells comprising CD40 antibody and muc1 specific chimeric antigen receptor gene and uses thereof
CN109971717B (en) * 2017-12-28 2023-06-20 上海细胞治疗研究院 T cells co-expressing CD40 antibody and mesothelin specific chimeric antigen receptor and uses thereof
WO2019148444A1 (en) 2018-02-02 2019-08-08 Adagene Inc. Anti-ctla4 antibodies and methods of making and using the same
JP7204078B2 (en) 2018-03-13 2023-01-16 日油株式会社 Heterobifunctional compounds with monodispersed polyethylene glycols in main and side chains
CN111954719A (en) 2018-03-26 2020-11-17 美国安进公司 Total defucosylated glycoforms of antibodies produced in cell culture
SG11202009629YA (en) 2018-04-02 2020-10-29 Amgen Inc Erenumab compositions and uses thereof
JP2021521804A (en) * 2018-04-20 2021-08-30 リビジェン バイオファーマ カンパニー リミテッド Anti-CD40 antibody and its use
US20200017596A1 (en) * 2018-05-10 2020-01-16 Abvision, Inc. Monoclonal antibodies activating cd40 and uses thereof
US20190365719A1 (en) * 2018-06-01 2019-12-05 Massachusetts Institute Of Technology Combination treatments of hsp90 inhibitors for enhancing tumor immunogenicity and methods of use thereof
WO2019241730A2 (en) 2018-06-15 2019-12-19 Flagship Pioneering Innovations V, Inc. Increasing immune activity through modulation of postcellular signaling factors
US10597453B2 (en) 2018-06-29 2020-03-24 Gensun Biopharma, Inc. Antitumor immune checkpoint regulator antagonists
TWI803682B (en) 2018-08-20 2023-06-01 美商輝瑞股份有限公司 Anti-gdf15 antibodies, compositions and methods of use
US20210355216A1 (en) 2018-10-05 2021-11-18 INSERM (Institut National de la Santé et de la Recherche Médicale Methods and systems for controlling the agonistic properties of antibody variable domains by light
KR20210097750A (en) 2018-11-30 2021-08-09 지앙수 헨그루이 메디슨 컴퍼니 리미티드 CD40 antibody pharmaceutical compositions and uses thereof
US20220025060A1 (en) 2018-11-30 2022-01-27 Jiangsu Hengrui Medicine Co., Ltd. Anti-cd40 antibody, antigen binding fragmentand pharmaceutical use thereof
US20220089758A1 (en) * 2019-01-22 2022-03-24 Revmab Biosciences Usa, Inc. Novel anti-cd40 antibodies
US10570210B1 (en) 2019-03-04 2020-02-25 Beijing Mabworks Biotech Co.Ltd Antibodies binding CD40 and uses thereof
KR20220004985A (en) 2019-03-27 2022-01-12 인쎄름 (엥스띠뛰 나씨오날 드 라 쌍떼 에 드 라 흐쉐르슈 메디깔) Recombinant protein with CD40 activation properties
AU2020256828A1 (en) 2019-04-10 2021-11-18 Nankai University Anti-CD40 antibody and use thereof
EP3962493A2 (en) 2019-05-03 2022-03-09 Flagship Pioneering Innovations V, Inc. Methods of modulating immune activity/level of irf or sting or of treating cancer, comprising the administration of a sting modulator and/or purinergic receptor modulator or postcellular signaling factor
AU2020276931A1 (en) 2019-05-15 2021-12-16 Kyowa Kirin Co., Ltd. Bispecific antibody binding to CD40 and FAP
EP3971293A4 (en) 2019-05-15 2023-02-08 Kyowa Kirin Co., Ltd. Bispecific antibody capable of binding to cd40 and gpc3
EP3990116A1 (en) 2019-06-28 2022-05-04 Gensun Biopharma Inc. ANTITUMOR ANTAGONIST CONSISTING OF A MUTATED TGFß1 - RII EXTRACELLULAR DOMAIN AND AN IMMUNOGLOBULIN SCAFFOLD
EP4027998A1 (en) 2019-09-09 2022-07-20 Basilea Pharmaceutica International AG Pharmaceutical combinations comprising a furazanobenzimidazoles and a cd40 agonist for use in the treatment of neoplastic diseases
WO2021062372A1 (en) 2019-09-26 2021-04-01 Amgen Inc. Methods of producing antibody compositions
CA3156027A1 (en) 2019-09-26 2021-04-01 Nof Corporation Heterobifunctional monodispersed polyethylene glycol having peptide linker
CA3163358A1 (en) * 2019-12-03 2021-06-10 Evotec International Gmbh Interferon-associated antigen binding proteins and uses thereof
EP4076434A1 (en) 2019-12-17 2022-10-26 Flagship Pioneering Innovations V, Inc. Combination anti-cancer therapies with inducers of iron-dependent cellular disassembly
AR120832A1 (en) 2019-12-20 2022-03-23 Amgen Inc MULTISPECIFIC ANTIBODY CONSTRUCTS CD40 AGONISTS TARGETING MESOTHELIN FOR THE TREATMENT OF SOLID TUMORS
US20210214454A1 (en) * 2020-01-10 2021-07-15 Symphogen A/S Anti-cd40 antibodies and compositions
IL272194A (en) 2020-01-22 2021-07-29 Yeda Res & Dev Multispecific antibodies for use in treating diseases
CN115702002A (en) 2020-04-23 2023-02-14 朝途生物疗法株式会社 Improved peptide vaccines
CA3178478A1 (en) 2020-05-14 2021-11-18 Valerie Perrine Calabro Recombinant cd40 binding proteins and their use
IL298168A (en) 2020-05-14 2023-01-01 Molecular Partners Ag Multispecific proteins
BR112022024063A2 (en) 2020-05-26 2023-01-31 Inst Nat Sante Rech Med CORONAVIRUS 2 POLYPEPTIDES OF SEVERE ACUTE RESPIRATORY SYNDROME (SARS-COV-2) AND USES THEREOF FOR VACCINE PURPOSES
GB202008003D0 (en) 2020-05-28 2020-07-15 Quine Medical Ab Anti-CD40 antibody
WO2021247892A1 (en) 2020-06-04 2021-12-09 Amgen Inc. Assessment of cleaning procedures of a biotherapeutic manufacturing process
EP4172323A1 (en) 2020-06-29 2023-05-03 Flagship Pioneering Innovations V, Inc. Viruses engineered to promote thanotransmission and their use in treating cancer
MX2023004364A (en) 2020-10-15 2023-05-03 Amgen Inc Relative unpaired glycans in antibody production methods.
WO2022101302A1 (en) 2020-11-12 2022-05-19 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies conjugated or fused to the receptor-binding domain of the sars-cov-2 spike protein and uses thereof for vaccine purposes
KR20230124672A (en) 2020-12-23 2023-08-25 인쎄름 (엥스띠뛰 나씨오날 드 라 쌍떼 에 드 라 흐쉐르슈 메디깔) Chlamydia vaccine based on targeting of MOMP VS4 antigen to antigen presenting cells
TW202241479A (en) 2020-12-30 2022-11-01 美商安迅生物製藥公司 Combination therapy of an oncolytic virus delivering a foreign antigen and an engineered immune cell expressing a chimeric receptor targeting the foreign antigen
CA3204291A1 (en) 2021-01-13 2022-07-21 F. Hoffmann-La Roche Ag Combination therapy
CA3209251A1 (en) 2021-01-29 2022-08-04 Inserm (Institut National De La Sante Et De La Recherche Medicale) Chlamydia trachomatis antigenic polypeptides and uses thereof for vaccine purposes
EP4284919A1 (en) 2021-01-29 2023-12-06 Iovance Biotherapeutics, Inc. Methods of making modified tumor infiltrating lymphocytes and their use in adoptive cell therapy
EP4305066A1 (en) 2021-03-09 2024-01-17 Genmab A/S Multispecific binding agents against cd40 and cd137 in therapy
JP2024512669A (en) 2021-03-31 2024-03-19 フラグシップ パイオニアリング イノベーションズ ブイ,インコーポレーテッド Tanotransmission polypeptides and their use in the treatment of cancer
WO2022261021A1 (en) 2021-06-07 2022-12-15 Amgen Inc. Using fucosidase to control afucosylation level of glycosylated proteins
KR20240026185A (en) * 2021-06-28 2024-02-27 지앙수 헨그루이 파마슈티컬스 컴퍼니 리미티드 Anti-CD40 antibodies, antigen-binding fragments and medical uses thereof
WO2023278641A1 (en) 2021-06-29 2023-01-05 Flagship Pioneering Innovations V, Inc. Immune cells engineered to promote thanotransmission and uses thereof
IL309831A (en) 2021-07-13 2024-02-01 BioNTech SE Multispecific binding agents against cd40 and cd137 in combination therapy for cancer
US20230203176A1 (en) * 2021-09-17 2023-06-29 Novartis Ag Methods For Prevention Of Graft Rejection In Xenotransplantation
AU2022358474A1 (en) * 2021-10-01 2024-03-28 Mab Discovery Gmbh Agonistic cd40 antibodies as immune stimulatory agents
CA3233279A1 (en) 2021-10-05 2023-04-13 Amagen, Inc. Fc-gamma receptor ii binding and glycan content
WO2023088968A1 (en) 2021-11-17 2023-05-25 INSERM (Institut National de la Santé et de la Recherche Médicale) Universal sarbecovirus vaccines
WO2023147488A1 (en) 2022-01-28 2023-08-03 Iovance Biotherapeutics, Inc. Cytokine associated tumor infiltrating lymphocytes compositions and methods
WO2023172890A2 (en) * 2022-03-07 2023-09-14 Agenus Inc. Anti-ilt2 antibodies and uses thereof
WO2023193239A1 (en) 2022-04-08 2023-10-12 Peter Peizhi Luo Anti-cd28 antibodies and methods of use thereof
WO2023198851A1 (en) 2022-04-14 2023-10-19 Institut National de la Santé et de la Recherche Médicale Methods for controlling the tumor cell killing by light
WO2023215725A1 (en) 2022-05-02 2023-11-09 Fred Hutchinson Cancer Center Compositions and methods for cellular immunotherapy
WO2024054992A1 (en) 2022-09-09 2024-03-14 Bristol-Myers Squibb Company Methods of separating chelator
CN115969997B (en) * 2022-12-19 2024-02-13 华润生物医药有限公司 Antibody drug conjugate targeting CLDN18.2 and application thereof
CN116375870B (en) * 2022-12-30 2023-09-15 优睿赛思(武汉)生物科技有限公司 Humanized antibody of anti-human CD40 protein, preparation method and application thereof

Family Cites Families (138)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US59427A (en) * 1866-11-06 Improvement in fences
US142358A (en) * 1873-09-02 Improvement in the purification of illuminating-gas
US439216A (en) * 1890-10-28 Maker
US4399216A (en) 1980-02-25 1983-08-16 The Trustees Of Columbia University Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
US4634665A (en) 1980-02-25 1987-01-06 The Trustees Of Columbia University In The City Of New York Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
US5179017A (en) 1980-02-25 1993-01-12 The Trustees Of Columbia University In The City Of New York Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
US4510245A (en) 1982-11-18 1985-04-09 Chiron Corporation Adenovirus promoter system
US4740461A (en) 1983-12-27 1988-04-26 Genetics Institute, Inc. Vectors and methods for transformation of eucaryotic cells
US5168062A (en) 1985-01-30 1992-12-01 University Of Iowa Research Foundation Transfer vectors and microorganisms containing human cytomegalovirus immediate-early promoter-regulatory DNA sequence
JP2532858B2 (en) 1985-04-01 1996-09-11 セルテツク リミテツド Transformed myeloma cell line
US4735210A (en) 1985-07-05 1988-04-05 Immunomedics, Inc. Lymphographic and organ imaging method and kit
US4968615A (en) 1985-12-18 1990-11-06 Ciba-Geigy Corporation Deoxyribonucleic acid segment from a virus
GB8601597D0 (en) 1986-01-23 1986-02-26 Wilson R H Nucleotide sequences
US4959455A (en) 1986-07-14 1990-09-25 Genetics Institute, Inc. Primate hematopoietic growth factors IL-3 and pharmaceutical compositions
US4912040A (en) 1986-11-14 1990-03-27 Genetics Institute, Inc. Eucaryotic expression system
US5750172A (en) 1987-06-23 1998-05-12 Pharming B.V. Transgenic non human mammal milk
GB8717430D0 (en) 1987-07-23 1987-08-26 Celltech Ltd Recombinant dna product
GB8809129D0 (en) 1988-04-18 1988-05-18 Celltech Ltd Recombinant dna methods vectors and host cells
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
GB8827305D0 (en) 1988-11-23 1988-12-29 British Bio Technology Compounds
US5175384A (en) 1988-12-05 1992-12-29 Genpharm International Transgenic mice depleted in mature t-cells and methods for making transgenic mice
US5959177A (en) 1989-10-27 1999-09-28 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
US5633076A (en) 1989-12-01 1997-05-27 Pharming Bv Method of producing a transgenic bovine or transgenic bovine embryo
US6673986B1 (en) 1990-01-12 2004-01-06 Abgenix, Inc. Generation of xenogeneic antibodies
US6713610B1 (en) * 1990-01-12 2004-03-30 Raju Kucherlapati Human antibodies derived from immunized xenomice
EP0463151B1 (en) 1990-01-12 1996-06-12 Cell Genesys, Inc. Generation of xenogeneic antibodies
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5151510A (en) 1990-04-20 1992-09-29 Applied Biosystems, Inc. Method of synethesizing sulfurized oligonucleotide analogs
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
ATE185601T1 (en) 1990-07-10 1999-10-15 Cambridge Antibody Tech METHOD FOR PRODUCING SPECIFIC BONDING PAIRS
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
US5789650A (en) 1990-08-29 1998-08-04 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
WO1992003917A1 (en) 1990-08-29 1992-03-19 Genpharm International Homologous recombination in mammalian cells
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
ES2108048T3 (en) 1990-08-29 1997-12-16 Genpharm Int PRODUCTION AND USE OF LOWER TRANSGENIC ANIMALS CAPABLE OF PRODUCING HETEROLOGICAL ANTIBODIES.
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
DE69129154T2 (en) 1990-12-03 1998-08-20 Genentech Inc METHOD FOR ENRICHING PROTEIN VARIANTS WITH CHANGED BINDING PROPERTIES
EP1820858B1 (en) 1991-03-01 2009-08-12 Dyax Corporation Chimeric protein comprising micro-protein having two or more disulfide bonds and embodiments thereof
US5416367A (en) * 1991-03-06 1995-05-16 Quicklogic Corporation Programmable application specific integrated circuit and logic cell therefor
IE921169A1 (en) 1991-04-10 1992-10-21 Scripps Research Inst Heterodimeric receptor libraries using phagemids
WO1992022653A1 (en) 1991-06-14 1992-12-23 Genentech, Inc. Method for making humanized antibodies
DE4122599C2 (en) 1991-07-08 1993-11-11 Deutsches Krebsforsch Phagemid for screening antibodies
AU2515992A (en) 1991-08-20 1993-03-16 Genpharm International, Inc. Gene targeting in animal cells using isogenic dna constructs
ATE181571T1 (en) 1991-09-23 1999-07-15 Medical Res Council METHODS FOR PRODUCING HUMANIZED ANTIBODIES
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
JPH05242071A (en) * 1991-11-20 1993-09-21 Mitsubishi Electric Corp Control or decision/estimation of object system and its device
CA2129663C (en) 1992-02-06 2005-07-05 James S. Huston Biosynthetic binding protein for cancer marker
US5874082A (en) 1992-07-09 1999-02-23 Chiron Corporation Humanized anti-CD40 monoclonal antibodies and fragments capable of blocking B cell proliferation
US5397703A (en) 1992-07-09 1995-03-14 Cetus Oncology Corporation Method for generation of antibodies to cell surface molecules
AU675661B2 (en) 1992-07-24 1997-02-13 Abgenix, Inc. Generation of xenogeneic antibodies
US6177401B1 (en) 1992-11-13 2001-01-23 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften Use of organic compounds for the inhibition of Flk-1 mediated vasculogenesis and angiogenesis
US5455258A (en) 1993-01-06 1995-10-03 Ciba-Geigy Corporation Arylsulfonamido-substituted hydroxamic acids
PT804070E (en) 1993-03-09 2000-11-30 Genzyme Corp ISOLATION OF COMPONENTS OF INTEREST FROM MILK.
CA2172376C (en) 1993-10-01 2008-11-18 William C. Fanslow, Iii Antibodies to cd40
US5827690A (en) 1993-12-20 1998-10-27 Genzyme Transgenics Corporatiion Transgenic production of antibodies in milk
NZ278740A (en) * 1993-12-23 1998-05-27 Immunex Corp Treating disease characterised by neoplastic cells expressing cd40 using a cd40 binding protein
US5612126A (en) * 1994-01-19 1997-03-18 Burlington Industries, Inc. Stiff fabric and method of forming the stiff fabric
IL112248A0 (en) 1994-01-25 1995-03-30 Warner Lambert Co Tricyclic heteroaromatic compounds and pharmaceutical compositions containing them
JPH0812700A (en) 1994-07-01 1996-01-16 Sumitomo Electric Ind Ltd Monoclonal antibody against mouse cd40
US5643763A (en) 1994-11-04 1997-07-01 Genpharm International, Inc. Method for making recombinant yeast artificial chromosomes by minimizing diploid doubling during mating
GB9425060D0 (en) * 1994-12-13 1995-02-08 Univ Birmingham Carcinoma treatment
US6046037A (en) 1994-12-30 2000-04-04 Hiatt; Andrew C. Method for producing immunoglobulins containing protection proteins in plants and their use
US5863949A (en) 1995-03-08 1999-01-26 Pfizer Inc Arylsulfonylamino hydroxamic acid derivatives
US6091001A (en) 1995-03-29 2000-07-18 Abgenix, Inc. Production of antibodies using Cre-mediated site-specific recombination
US6130364A (en) 1995-03-29 2000-10-10 Abgenix, Inc. Production of antibodies using Cre-mediated site-specific recombination
WO1996032181A1 (en) * 1995-04-11 1996-10-17 Robert Bosch Gmbh Process and device for reducing pollutants, especially nitrogen oxides in combustion exhaust gases
MX9708026A (en) 1995-04-20 1997-11-29 Pfizer Arylsulfonyl hydroxamic acid derivatives as mmp and tnf inhibitors.
CA2218489A1 (en) 1995-04-21 1996-10-24 Aya Jakobovits Generation of large genomic dna deletions
EP0822830B1 (en) 1995-04-27 2008-04-02 Amgen Fremont Inc. Human anti-IL-8 antibodies, derived from immunized xenomice
EP0823941A4 (en) 1995-04-28 2001-09-19 Abgenix Inc Human antibodies derived from immunized xenomice
US5747498A (en) 1996-05-28 1998-05-05 Pfizer Inc. Alkynyl and azido-substituted 4-anilinoquinazolines
US5880141A (en) 1995-06-07 1999-03-09 Sugen, Inc. Benzylidene-Z-indoline compounds for the treatment of disease
US5780215A (en) * 1995-07-26 1998-07-14 Konica Corporation Silver halide color photographic light-sensitive material
GB9520822D0 (en) 1995-10-11 1995-12-13 Wellcome Found Therapeutically active compounds
US6440418B1 (en) 1995-11-07 2002-08-27 Idec Pharmaceuticals Corporation Methods of treating autoimmune diseases with gp39-specific antibodies
GB9624482D0 (en) 1995-12-18 1997-01-15 Zeneca Phaema S A Chemical compounds
ATE225343T1 (en) 1995-12-20 2002-10-15 Hoffmann La Roche MATRIX METALLOPROTEASE INHIBITORS
KR100489174B1 (en) 1996-03-05 2005-09-30 제네카-파마 소시에떼아노님 4-anilinoquinazoline derivatives
US5714352A (en) 1996-03-20 1998-02-03 Xenotech Incorporated Directed switch-mediated DNA recombination
US5994619A (en) 1996-04-01 1999-11-30 University Of Massachusetts, A Public Institution Of Higher Education Of The Commonwealth Of Massachusetts, As Represented By Its Amherst Campus Production of chimeric bovine or porcine animals using cultured inner cell mass cells
EP0818442A3 (en) 1996-07-12 1998-12-30 Pfizer Inc. Cyclic sulphone derivatives as inhibitors of metalloproteinases and of the production of tumour necrosis factor
ID19609A (en) 1996-07-13 1998-07-23 Glaxo Group Ltd HETEROSICLIC COMPOUNDS
BR9710362A (en) 1996-07-13 1999-08-17 Glaxo Group Ltd Pharmaceutical formulation compound uses a compound processes of treatment of a human or animal suffering from a mediated by abnormal activity of protein tyrosine kinase and for the preparation of a compound
HRP970371A2 (en) 1996-07-13 1998-08-31 Kathryn Jane Smith Heterocyclic compounds
WO1998003516A1 (en) 1996-07-18 1998-01-29 Pfizer Inc. Phosphinate based inhibitors of matrix metalloproteases
CN1228083A (en) 1996-08-23 1999-09-08 美国辉瑞有限公司 Arylsulfonylamino hydroxamic acid derivatives
US5817690A (en) * 1996-08-27 1998-10-06 American Home Products Corporation 4-aminoethoxy indolone derivatives
ID18494A (en) 1996-10-02 1998-04-16 Novartis Ag PIRAZOLA DISTRIBUTION IN THE SEQUENCE AND THE PROCESS OF MAKING IT
US5916771A (en) 1996-10-11 1999-06-29 Abgenix, Inc. Production of a multimeric protein by cell fusion method
DK1500329T3 (en) 1996-12-03 2012-07-09 Amgen Fremont Inc Human antibodies that specifically bind TNF-alpha
DK0950059T3 (en) 1997-01-06 2004-11-01 Pfizer Cyclic sulfone derivatives
CA2279276C (en) 1997-02-03 2005-09-13 Pfizer Products Inc. Arylsulfonylamino hydroxamic acid derivatives
BR9807824A (en) 1997-02-07 2000-03-08 Pfizer Derivatives of n-hydroxy-beta-sulfonyl-propionamide and its use as matrix metalloproteinase inhibitors
CA2280151C (en) 1997-02-11 2005-12-13 Pfizer Inc. Arylsulfonyl hydroxamic acid derivatives
US6235883B1 (en) 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
CA2289102A1 (en) 1997-05-07 1998-11-12 Sugen, Inc. 2-indolinone derivatives as modulators of protein kinase activity
JP2002512624A (en) 1997-05-21 2002-04-23 バイオベーション リミテッド Method for producing non-immunogenic protein
EP0984692A4 (en) 1997-05-30 2001-02-21 Merck & Co Inc Novel angiogenesis inhibitors
DE69838172T2 (en) 1997-08-22 2008-04-10 Astrazeneca Ab OXINDOLYLCHINAZOLE DERIVATIVES AS ANGIOGENESEHEMMER
AU744939B2 (en) 1997-09-26 2002-03-07 Merck & Co., Inc. Novel angiogenesis inhibitors
CN1280580A (en) 1997-11-11 2001-01-17 辉瑞产品公司 Thienopyrimidine and thienopyridine derivatives useful as anti-cancer agents
GB9725782D0 (en) 1997-12-05 1998-02-04 Pfizer Ltd Therapeutic agents
GB9800575D0 (en) 1998-01-12 1998-03-11 Glaxo Group Ltd Heterocyclic compounds
GB9800569D0 (en) 1998-01-12 1998-03-11 Glaxo Group Ltd Heterocyclic compounds
GB9801690D0 (en) 1998-01-27 1998-03-25 Pfizer Ltd Therapeutic agents
US6051228A (en) 1998-02-19 2000-04-18 Bristol-Myers Squibb Co. Antibodies against human CD40
AU2978899A (en) 1998-03-03 1999-09-20 Abgenix, Inc. Cd147 binding molecules as therapeutics
JP4462654B2 (en) 1998-03-26 2010-05-12 ソニー株式会社 Video material selection device and video material selection method
PA8469501A1 (en) 1998-04-10 2000-09-29 Pfizer Prod Inc HYDROXAMIDES OF THE ACID (4-ARILSULFONILAMINO) -TETRAHIDROPIRAN-4-CARBOXILICO
PA8469401A1 (en) 1998-04-10 2000-05-24 Pfizer Prod Inc BICYCLE DERIVATIVES OF HYDROXAMIC ACID
US20020029391A1 (en) 1998-04-15 2002-03-07 Claude Geoffrey Davis Epitope-driven human antibody production and gene expression profiling
AU4009899A (en) * 1998-05-23 1999-12-13 Tanox, Inc. Molecules targeting cd40 and tumor cells
SK287132B6 (en) 1998-05-29 2009-12-07 Sugen, Inc. Pharmaceutical composition containing pyrrole substituted 2-indolinone, kit containing mentioned composition and use pyrrole substituted 2-indolinone
US20020142374A1 (en) 1998-08-17 2002-10-03 Michael Gallo Generation of modified molecules with increased serum half-lives
US6114361A (en) 1998-11-05 2000-09-05 Pfizer Inc. 5-oxo-pyrrolidine-2-carboxylic acid hydroxamide derivatives
WO2000034317A2 (en) 1998-12-08 2000-06-15 Biovation Limited Method for reducing immunogenicity of proteins
PT2112166T (en) 1998-12-23 2019-01-30 Pfizer Human monoclonal antibodies to ctla-4
WO2000066155A1 (en) * 1999-04-30 2000-11-09 La Jolla Institute For Allergy And Immunology Methods for preventing reactivation of latent virus and controlling virus replication
US20030118588A1 (en) * 1999-05-22 2003-06-26 Linda Diehl Induction of anti-tumor CTL immunity through in vivo triggering of 4-1BB and/or CD40
US6946129B1 (en) * 1999-06-08 2005-09-20 Seattle Genetics, Inc. Recombinant anti-CD40 antibody and uses thereof
US6482411B1 (en) * 1999-08-27 2002-11-19 Board Of Regents, The University Of Texas System Methods of reducing bone loss with CD40 ligand
US6517529B1 (en) 1999-11-24 2003-02-11 Radius International Limited Partnership Hemodialysis catheter
GB9927757D0 (en) * 1999-11-25 2000-01-26 Kennedy Rheumatology Inst Treatment of autoimmune diseases
AU3662101A (en) * 2000-02-01 2001-08-14 Tanox Inc Cd40-binding apc-activating molecules
US20030059427A1 (en) * 2000-04-28 2003-03-27 Force Walker R. Isolation and characterization of highly active anti-CD40 antibody
US7063845B2 (en) 2000-04-28 2006-06-20 Gemini Science, Inc. Human anti-CD40 antibodies
AU2001259215A1 (en) * 2000-04-28 2001-11-12 La Jolla Institute For Allergy And Immunology Human anti-cd40 antibodies and methods of making and using same
MY143465A (en) 2001-01-05 2011-05-13 Pfizer Antibodies to insulin-like growth factor i receptor
AR039067A1 (en) * 2001-11-09 2005-02-09 Pfizer Prod Inc ANTIBODIES FOR CD40
KR20120048644A (en) 2004-01-09 2012-05-15 암젠 프레몬트 인코포레이티드 Antibodies to madcam
US10181126B2 (en) 2012-03-13 2019-01-15 American Express Travel Related Services Company, Inc. Systems and methods for tailoring marketing
US10884952B2 (en) 2016-09-30 2021-01-05 Intel Corporation Enforcing memory operand types using protection keys

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
USRE45847E1 (en) 2004-01-09 2016-01-19 Pfizer Inc. Antibodies to MAdCAM
US9221915B2 (en) 2005-09-07 2015-12-29 Pfizer Inc. Human monoclonal antibodies to activin receptor-like kinase-1
US9107906B1 (en) 2014-10-28 2015-08-18 Adma Biologics, Inc. Compositions and methods for the treatment of immunodeficiency

Also Published As

Publication number Publication date
ECSP045093A (en) 2004-07-23
OA12725A (en) 2006-06-28
JP4616555B2 (en) 2011-01-19
US8388971B2 (en) 2013-03-05
CL2009001779A1 (en) 2010-02-05
KR100830086B1 (en) 2008-05-20
NO20042388L (en) 2004-08-09
HUP0402247A2 (en) 2005-01-28
US20090130715A1 (en) 2009-05-21
RS52484B (en) 2013-02-28
AU2002356926B2 (en) 2007-01-04
PL214289B1 (en) 2013-07-31
MY137641A (en) 2009-02-27
KR20050063749A (en) 2005-06-28
HRP20040525A2 (en) 2005-04-30
BR0214137A (en) 2004-09-14
IS2940B (en) 2016-03-15
ZA200404207B (en) 2005-08-31
US20160152713A1 (en) 2016-06-02
US20100098694A1 (en) 2010-04-22
AU2002356926A2 (en) 2003-05-19
BRPI0214137B1 (en) 2019-03-19
EG25801A (en) 2012-08-08
TWI331040B (en) 2010-10-01
TWI338579B (en) 2011-03-11
AU2002356926C1 (en) 2003-05-19
PL214634B1 (en) 2013-08-30
AR039067A1 (en) 2005-02-09
MXPA04004467A (en) 2005-05-16
US20120225014A1 (en) 2012-09-06
PT1476185E (en) 2013-11-05
AP1918A (en) 2008-11-13
NZ533180A (en) 2008-03-28
HRP20040525B1 (en) 2017-06-02
ME00503B (en) 2011-10-10
EP1476185B1 (en) 2013-07-31
IS7253A (en) 2004-05-06
CN1582165B (en) 2013-05-29
CN1582165A (en) 2005-02-16
GEP20074222B (en) 2007-10-25
ES2432968T3 (en) 2013-12-05
DOP2002000507A (en) 2003-05-15
EA200400654A1 (en) 2004-12-30
CY1115675T1 (en) 2017-01-25
PA8557701A1 (en) 2003-12-19
UY27537A1 (en) 2003-02-28
WO2003040170A3 (en) 2003-10-09
AP2004003034A0 (en) 2004-06-30
NI200200128A (en) 2010-01-11
TW201034686A (en) 2010-10-01
GT200200228A (en) 2008-09-26
IL161823A (en) 2014-02-27
US7288251B2 (en) 2007-10-30
SI1476185T1 (en) 2013-12-31
EA011449B1 (en) 2009-02-27
IL161823A0 (en) 2005-11-20
PL392809A1 (en) 2011-03-14
NO334339B1 (en) 2014-02-10
CU23745A3 (en) 2012-01-31
HUP0402247A3 (en) 2012-09-28
TW200306204A (en) 2003-11-16
US20030211100A1 (en) 2003-11-13
US20130024956A1 (en) 2013-01-24
US20090169563A1 (en) 2009-07-02
US20060093600A1 (en) 2006-05-04
JP2013099333A (en) 2013-05-23
EP1476185A2 (en) 2004-11-17
PL370514A1 (en) 2005-05-30
CN103450360B (en) 2016-11-02
JP2005508176A (en) 2005-03-31
PE20030846A1 (en) 2003-10-11
HK1069122A1 (en) 2005-05-13
US20070190051A1 (en) 2007-08-16
CR10878A (en) 2011-01-12
US7563442B2 (en) 2009-07-21
CN103450360A (en) 2013-12-18
RS39204A (en) 2006-12-15
TNSN04078A1 (en) 2006-06-01
MA27139A1 (en) 2005-01-03
JP2010213704A (en) 2010-09-30
DK1476185T3 (en) 2013-10-14
US7338660B2 (en) 2008-03-04
HN2002000318A (en) 2003-03-10
WO2003040170A2 (en) 2003-05-15
BRPI0214137B8 (en) 2021-05-25
US7618633B2 (en) 2009-11-17
US7626012B2 (en) 2009-12-01
JP5547532B2 (en) 2014-07-16
EP2343086A2 (en) 2011-07-13
JP5848232B2 (en) 2016-01-27
CA2466128C (en) 2013-10-29
CR7343A (en) 2004-10-28
HU229829B1 (en) 2014-09-29
EP2343086A3 (en) 2012-02-22
EP1476185A4 (en) 2007-04-04
CA2466128A1 (en) 2003-05-15

Similar Documents

Publication Publication Date Title
US7618633B2 (en) Antibodies that bind CD40 and methods of treating cancer and enhancing immune responses
AU2002356926A1 (en) Antibodies to CD40
CA2885172C (en) Antibodies to m-csf
EP1660127A1 (en) Antibodies to c-met

Legal Events

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

Ref document number: P-392/04

Country of ref document: YU

AK Designated states

Kind code of ref document: A2

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

AL Designated countries for regional patents

Kind code of ref document: A2

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 566/KOLNP/2004

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2466128

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 161823

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 1200400414

Country of ref document: VN

Ref document number: 1020047006992

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: PA/a/2004/004467

Country of ref document: MX

Ref document number: 2003542215

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: AP/P/2004/003034

Country of ref document: AP

WWE Wipo information: entry into national phase

Ref document number: 2002356926

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2004/04207

Country of ref document: ZA

Ref document number: 533180

Country of ref document: NZ

Ref document number: 200404207

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: DZP2004000146

Country of ref document: DZ

WWE Wipo information: entry into national phase

Ref document number: P20040525A

Country of ref document: HR

Ref document number: 04053734

Country of ref document: CO

Ref document number: 200400654

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 5640

Country of ref document: GE

Ref document number: 8256

Country of ref document: GE

Ref document number: 2002802898

Country of ref document: EP

Ref document number: 20028245709

Country of ref document: CN

COP Corrected version of pamphlet

Free format text: PAGES 1/19-19/19, DRAWINGS, REPLACED BY CORRECT PAGES 1/18-18/18; DUE TO LATE TRANSMITTAL BY THE RECEIVING OFFICE

WWE Wipo information: entry into national phase

Ref document number: 1-2004-500651

Country of ref document: PH

WWP Wipo information: published in national office

Ref document number: 2002802898

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 2002356926

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: CR2009-010878

Country of ref document: CR