WO2005075504A1 - IDENTIFICATION OF NOVEL IgE EPITOPES - Google Patents

IDENTIFICATION OF NOVEL IgE EPITOPES Download PDF

Info

Publication number
WO2005075504A1
WO2005075504A1 PCT/US2004/024360 US2004024360W WO2005075504A1 WO 2005075504 A1 WO2005075504 A1 WO 2005075504A1 US 2004024360 W US2004024360 W US 2004024360W WO 2005075504 A1 WO2005075504 A1 WO 2005075504A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
ige
antibodies
seq
cells
Prior art date
Application number
PCT/US2004/024360
Other languages
English (en)
French (fr)
Inventor
Sanjaya Singh
Danyang Huang
Sek Chung Michael Fung
Original Assignee
Tanox, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PCT/US2004/002894 external-priority patent/WO2004070011A2/en
Application filed by Tanox, Inc. filed Critical Tanox, Inc.
Priority to KR1020067017609A priority Critical patent/KR20070008578A/ko
Priority to KR1020137018993A priority patent/KR101562114B1/ko
Priority to JP2006551035A priority patent/JP2008507474A/ja
Priority to AU2004315197A priority patent/AU2004315197B2/en
Priority to CA002552999A priority patent/CA2552999A1/en
Priority to EP04779424A priority patent/EP1718669A4/en
Priority to KR1020147000238A priority patent/KR101581659B1/ko
Priority to KR1020127009917A priority patent/KR101365375B1/ko
Publication of WO2005075504A1 publication Critical patent/WO2005075504A1/en
Priority to IL176940A priority patent/IL176940A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/42Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins
    • C07K16/4283Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins against an allotypic or isotypic determinant on Ig
    • C07K16/4291Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins against an allotypic or isotypic determinant on Ig against IgE
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0008Antigens related to auto-immune diseases; Preparations to induce self-tolerance
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/567Framework region [FR]

Definitions

  • Allergy is a hypersensitive state induced by an exaggerated immune response to a foreign agent, such as an allergen.
  • Immediate (type I) hypersensitivity characterized by allergic reactions immediately following contact with the allergen, is mediated via B cells and is based on antigen-antibody reactions. Delayed hypersensitivity is mediated via T cells and based on mechanisms of cellular immunity.
  • allergy has become more and more synonymous with type I hypersensitivity.
  • Immediate hypersensitivity is a response based on the production of antibodies of the immunogldbulin class E (IgE antibodies) by B cells which upon exposure to an allergen differentiate into antibody secreting plasma cells.
  • the IgE induced reaction is a local event occurring at the site of the allergen's entry into the body, i.e. at mucosal surfaces and/or at local lymph nodes.
  • Locally produced IgE will first sensitize local mast cells, i.e. IgE antibodies bind with their- constant regions to Fee receptors on the surface of the mast cells, and then "spill-over" IgE enters the circulation and binds to receptors on both circulating basophils and tissue- fixed mast cells throughout the body.
  • the Fee receptors are crosslinked by binding of the allergen causing the cells to degranulate and release a number of anaphylactic mediators such as histamine, prostaglandins, leukotrienes, etc. It is the release of these substances which is responsible for the clinical symptoms typical of immediate hypersensitivity, namely contraction of smooth muscle in the respiratory tract or the intestine, the dilation of small blood vessels and the increase in their permeability to water and plasma proteins, the secretion of mucus resulting, e.g in allergic rhinitis, atopic excema and asthma, and the stimulation of nerve endings in the skin resulting in itching and pain.
  • anaphylactic mediators such as histamine, prostaglandins, leukotrienes, etc.
  • reaction upon second contact with the allergen is intensified because some B cells form a "memory pool" of surface IgE positive B cells (slgE + B cells) after the first contact with the allergen by expressing IgE on the cell surface.
  • FceRI is predominantly expressed on the surface of mast cells and basophils, but low levels of FceRI can also be found on human Langerhan's cells, dendritic cells, and monocytes, where it functions in IgE- mediated allergen presentation.
  • FceRI has been reported on human eosinophils and platelets (Hasegawa, S. et. al., Hematopoiesis, 1999, 93:2543- 2551 ). FceRI is not found on the surface of B cells, T cells, or neutrophils.
  • the low-affinity receptor, FceRII (CD23) is a lectin-like molecule comprising three identical subunits with head structures extending from a long -helical coiled stalk from the cellular plasma membrane (Dierks, A.E. et al., J. Immunol. 1993, 150:2372-2382).
  • FceRII associates with CD21 on B cells involved in the regulation of synthesis of IgE (Sanon, A. et al., J. Allergy Clin. Immunol. 1990, 86:333-344, Bonnefoy, J. et al., Eur. Resp. J. 1996, 9:63s-66s).
  • FceRII has long been recognized for allergen presentation (Sutton and Gould ,1993, Nature, 366:421-428). IgE bound to FceRII on epithelial cells is responsible for specific and rapid allergen presentation (Yang, P.P., J. Clin. Invest., 2000, 106:879-886). FceRII is present on several cell types, including B- cells, eosinophils, platelets, natural killer cells, T-cells, follicular dendritic cells, and Langerhan's cells.
  • a promising concept for the treatment of allergy involves the application of monoclonal antibodies, which are IgE isotype-specific and are thus capable of binding IgE.
  • This approach is based on the inhibition of allergic reactions by downregulating the IgE immune response, which is the earliest event in the induction of allergy and provides for the maintenance of the allergic state.
  • IgE immune response As the response of other antibody classes is not affected, both an immediate and a long lasting effect on allergic symptoms is achieved.
  • Early studies of human basophil density showed a correlation between the level of IgE in the plasma of a patient and the number of FceRI receptors per basophil (Malveaux et al., J. Clin. Invest, 1978, 62:176).
  • WO 99/62550 disclosed the use of IgE molecules and fragments, which bind to FceRI and FceRII IgE binding sites to block IgE binding to receptors.
  • effective therapies that lack deleterious side effects for the management of these allergic diseases are limited.
  • One therapeutic approach to treating allergic diseases involved using humanized anti-IgE antibody to treat allergic rhinitis and asthma (Corne, J. et al., J. Clin. lnvestA 997, 99:879-887; Racine- Poon, A. et al., Clin. Pharmcol. Ther. 1997, 62:675-690; Fahy, J.V. et al., Am. J. Resp. Crit. Care Med.
  • Antibodies suitable as anti-allergic agents should react with surface IgE positive B cells which differentiate into IgE producing plasma cells, so that they can be used to functionally eliminate those B cells.
  • antibodies to IgE in principle may also induce mediator release from IgE sensitized mast cells by crosslinking the Fee receptors, thus antagonizing the beneficial effect exerted on the serum IgE and slgE + B cell level.
  • One of the potentially dangerous problems with developing anti-IgE therapies is the possibility of IgE-crosslinking caused by the therapetic antibody binding to IgE already bound to the high affinity receptor and triggering histamine release resulting in a potentially anaphylactic reaction.
  • antibodies applicable for therapy of allergy must not be capable of reacting with IgE bound on sensitized mast cells and basophils, but should retain the capability to recognize slgE + B cells.
  • IgE isotype-specific antibodies have been described e.g. by Chang et al. (Biotechnology 8, 122-126 (1990)), in European Patent No. EP0407392, and several U.S. Patents, e.g., U.S. Patent No. 5,449,760.
  • Peptides used to generate anti-IgE antibodies also suffer from the dangerous potential to induce anaphylactic antibodies.
  • Generation of anti-IgE antibodies during active vaccination may be capable of triggering histamine release in the same way passively administered anti-IgE antibodies, if the antibodies generated during immunization bind to IgE bound to the high affinity IgE receptor or by other mechanisms.
  • the present invention relates to novel peptide epitopes derived from the CH3 domain of IgE. These peptide epitopes are recognized by high affinity antibodies that specifically bind IgE. These novel peptides may be used for both the active immunization of a mammal by administering these peptides to generate high affinity antibodies in the mammal. The peptide epitopes may also be used in generating high affinity anti-IgE antibodies in a non-human host that specifically bind to these regions of IgE and use the resulting antibodies for the passive immunization of a of a mammal. [0014] One immunogen (epitope A, Fig.
  • Epitope 11 of the present invention comprises the amino acid sequence: Asn Pro Arg Gly Val Ser Xaa Tyr Xaa Xaa Arg Xaa (SEQ ID NO. 72).
  • epitope A is: Asn Pro Arg Gly Val Ser Ala Tyr Leu Ser Arg Pro (SEQ ID NO. 73)
  • epitope B, Fig. 11 comprises the amino acid sequence: Leu Pro Arg Ala Leu Xaa Arg Ser Xaa (SEQ ID NO. 74).
  • Examples of Epitope B include: Leu Pro Arg Ala Leu Met Arg Ser Thr (SEQ ID NO.
  • Xaa may be any amino acid.
  • These peptides may be included in a composition comprising at least one of the peptides and a physiologically acceptable carrier, diluent, stabilizer or excipient, as well as an immunogenic carrier.
  • the immunogenic carrier may be, e.g., BSA, KLH, tetanus toxoid, and diphtheria toxoid.
  • the present invention also relates to polynucleotides encoding SEQ ID NOS. 72-77, vectors comprising said polynucleotides, and cells harboring said vectors. [0016]
  • the present invention also relates to antibodies that specifically bind to epitope A and/or epitope B.
  • the present invention is also directed to a method of making antibodies that specifically bind to epitope A and/or epitope B.
  • the present invention relates to the administration of peptides comprising SEQ ID NO: 72 and/or SEQ ID NO 74 to a subject suffering from an IgE-mediated disease or condition.
  • the present invention relates to the administration of high affinity antibodies generated using peptides comprising SEQ ID NO: 72 and/or SEQ ID NO 74 to a mammal suffering from an IgE-mediated disease or condition.
  • the high affinity antibody may be human, humanized, or chimeric.
  • the antibody may be polyclonal or monoclonal.
  • IgE mediated diseases or conditions include, e.g., asthma, atopic dermatitis, urticaria, allergic rhinitis and eczema.
  • Figure 1 is a schematic representation of the phage vector used in antibody cloning and screening.
  • Figure 2 is a schematic representation of oligonucleotides useful in generating antibody variants.
  • Figure 3A depicts the comparison of the light chains of the murine anti-IgE antibody TES-C21 and the combined human template of L16 and JK4.
  • Figure 3B depicts the comparison of the heavy chains of TES-C21 and the combined human template DP88 and JH4b.
  • Figure 4 presents a table of the framework residue variants having high affinity as compared to the parent TES-C21.
  • Figure 5A and B depict the ELISA titration curves for clones 4, 49, 72, 78, and 136, as compared to the parent Fab of TES-C21 and negative control (5D12).
  • Figure 6 depicts an inhibition assay of clones 2C, 5A, and 51, as compared to the parent TES-C21 and a negative control antibody.
  • Figure 7A depicts the sequences of clones having a combination of beneficial mutations which resulted in even greater affinity for IgE.
  • Figure 8A & 8B depict the framework sequences of the entire light chain variable region for clones 136, 1 , 2, 4, 8, 13, 15, 21 , 30, 31 , 35, 43, 44, 53, 81 , 90, and 1 13.
  • Figure 9A & 9B depict the framework sequences of the entire heavy chain variable region for 35 clones.
  • Figures 10 A-F depict the complete heavy and light chain sequences for clones 136, 2C, 51, 5A, 2B, and 1 136-2C.
  • Figurel 1 depicts the CH3 region amino acid sequence of human IgE and highlights Epitope "A" and Epitope "B”.
  • Figure 12 depicts the overlapping peptides used to identify Epitope B.
  • Figure 13 depicts the identification of important residues in the binding region of Epitope A.
  • Figure 14 depicts the identification of important residues in the binding region of Epitope B.
  • Figure 15 depicts a western blot analysis of MAb binding to mutant peptides.
  • Figure 16 depicts the generation of anti-IgE antibodies in a transgenic animal expressing human IgE. DETAILED DESCRIPTION OF THE INVENTION Definitions
  • substantially identical with respect to an antibody chain polypeptide sequence may be construed as an antibody chain exhibiting at least 70%, or 80%, or 90% or 95% sequence identity to the reference polypeptide sequence.
  • the term with respect to a nucleic acid sequence may be construed as a sequence of nucleotides exhibiting at least about 85%, or 90%, or 95% or 97% sequence identity to the reference nucleic acid sequence.
  • identity shall be construed to mean the percentage of amino acid residues in the candidate sequence that are identical with the residue of a corresponding sequence to which it is compared, after aligning the sequences and introducing gaps, if necessary to achieve the maximum percent identity for the entire sequence, and not considering any conservative substitutions as part of the sequence identity. Neither N- or C-terminal extensions nor insertions shall be construed as reducing identity or homology. Methods and computer programs for the alignment are well known in the art. Sequence identity may be measured using sequence analysis software.
  • antibody is used in the broadest sense, and specifically covers monoclonal antibodies (including full length monoclonal antibodies), polyclonal antibodies, and multispecific antibodies (e.g., bispecific antibodies).
  • Antibodies (Abs) and immunoglobulins (Igs) are glycoproteins having the same structural characteristics. While antibodies exhibit binding specificity to a specific target, immunoglobulins include both antibodies and other antibody-like molecules which lack target specificity.
  • Native antibodies and immunoglobulins are usually heterotetrame c glycoproteins of about 150,000 daltons, composed of two identical light (L) chains and two identical heavy (H) chains. Each heavy chain has at one end a variable domain (VH) followed by a number of constant domains. Each light chain has a variable domain at one end (VL) and a constant domain at its other end.
  • “High affinity” antibodies refers to those antibodies having a binding affinity of at least 10 "10 , preferably 10 "12 .
  • anti-human IgE antibody means an antibody which binds to human IgE in such a manner so as to inhibit or substantially reduce the binding of such IgE to the high affinity receptor, FceRI.
  • variable domains refers to the fact that certain portions of the variable domains differ extensively in sequence among antibodies and are used in the binding and specificity of each particular antibody for its particular target. However, the variability is not evenly distributed through the variable domains of antibodies. It is concentrated in three segments called complementarity determining regions (CDRs) also known as hypervariable regions both in the light chain and the heavy chain variable domains. The more highly conserved portions of variable domains are called the framework (FR).
  • CDRs complementarity determining regions
  • FR framework
  • the variable domains of native heavy and light chains each comprise four FR regions, largely a adopting a /?-sheet configuration, connected by three CDRs, which form loops connecting, and in some cases forming part of, the ?-sheet structure.
  • the CDRs in each chain are held together in close proximity by the FR regions and, with the CDRs from the other chain, contribute to the formation of the target binding site of antibodies (see Kabat et al.)
  • numbering of immunoglobulin amino acid residues is done according to the immunoglobulin amino acid residue numbering system of Kabat et al., (Sequences of Proteins of Immunological Interest, National Institute of Health, Bethesda, Md. 1987), unless otherwise indicated.
  • antibody fragment refers to a portion of a full-length antibody, generally the target binding or variable region.
  • antibody fragments include Fab, Fab', F(ab') 2 and Fv fragments.
  • the phrase "functional fragment or analog" of an antibody is a compound having qualitative biological activity in common with a full- length antibody.
  • a functional fragment or analog of an anti-IgE antibody is one which can bind to an IgE immunoglobulin in such a manner so as to prevent or substantially reduce the ability of such molecule from having the ability to bind to the high affinity receptor, FceRI.
  • “functional fragment” with respect to antibodies refers to Fv, F(ab) and F(ab') 2 fragments.
  • an “Fv” fragment is the minimum antibody fragment which contains a complete target recognition and binding site. This region consists of a dimer of one heavy and one light chain variable domain in a tight, non-covalent association (VH -VL dimer). It is in this configuration that the three CDRs of each variable domain interact to define an target binding site on the surface of the V H -VL dimer. Collectively, the six CDRs confer target binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an target) has the ability to recognize and bind target, although at a lower affinity than the entire binding site.
  • Single-chain Fv or “sFv” antibody fragments comprise the VH and VL domains of an antibody, wherein these domains are present in a single polypeptide chain.
  • the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains which enables the sFv to form the desired structure for target binding.
  • the Fab fragment contains the constant domain of the light chain and the first constant domain (CH1 ) of the heavy chain.
  • Fab' fragments differ from Fab fragments by the addition of a few residues at the carboxyl terminus of the heavy chain CH1 domain including one or more cysteines from the antibody hinge region.
  • F(ab') fragments are produced by cleavage of the disulfide bond at the hinge cysteines of the F(ab') 2 pepsin digestion product. Additional chemical couplings of antibody fragments are known to those of ordinary skill in the art.
  • the term "monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single targetic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the target. In addition to their specificity, monoclonal antibodies are advantageous in that they may be synthesized by the hybridoma culture, uncontaminated by other immunoglobulins.
  • the modifier "monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies for use with the present invention may be isolated from phage antibody libraries using the well known techniques.
  • the parent monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler and Milstein, Nature 256, 495 (1975), or may be made by recombinant methods.
  • Humanized forms of non-human (e.g. murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ab') 2 or other target-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence.
  • the humanized antibody may also comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin template chosen.
  • Fc immunoglobulin constant region
  • the terms “cell”, “cell line” and “cell culture” include progeny. It is also understood that all progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. Variant progeny that have the same function or biological property, as screened for in the originally transformed cell, are included.
  • the "host cells” used in the present invention generally are prokaryotic or eukaryotic hosts.
  • Transformation of a cellular organism with DNA means introducing DNA into an organism so that the DNA is replicable, either as an extrachromosomal element or by chromosomal integration.
  • Transfection of a cellular organism with DNA refers to the taking up of DNA, e.g., an expression vector, by the cell or organism whether or not any coding sequences are in fact expressed.
  • the terms "transfected host cell” and “transformed” refer to a cell in which DNA was introduced.
  • the cell is termed “host cell” and it may be either prokaryotic or eukaryotic. Typical prokaryotic host cells include various strains of E. coli.
  • Typical eukaryotic host cells are mammalian, such as Chinese hamster ovary or cells of human origin.
  • the introduced DNA sequence may be from the same species as the host cell of a different species from the host cell, or it may be a hybrid DNA sequence, containing some foreign and some homologous DNA.
  • the term "vector” means a DNA construct containing a DNA sequence which is operably linked to a suitable control sequence capable of effecting the expression of the DNA in a suitable host.
  • control sequences include a promoter to effect transcription, an optional operator sequence to control such transcription, a sequence encoding suitable mRNA ribosome binding sites, and sequences which control the termination of transcription and translation.
  • the vector may be a plasmid, a phage particle, or simply a potential genomic insert. Once transformed into a suitable host, the vector may replicate and function independently of the host genome, or may in some instances, integrate into the genome itself.
  • "plasmid” and “vector” are sometimes used interchangeably, as the plasmid is the most commonly used form of vector. However, the invention is intended to include such other forms of vectors which serve equivalent function as and which are, or become, known in the art.
  • control sequences refers to DNA sequences necessary for the expression of an operably linked coding sequence in a particular host organism.
  • the control sequences that are suitable for prokaryotes include a promoter, optionally an operator sequence, and a ribosome binding site.
  • Eukaryotic cells are known to utilize promoters, polyadenylation signals, and enhancers.
  • DNA for a presequence or secretory leader may be operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide; a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation.
  • "operably linked" means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading phase.
  • mammal for purposes of treatment refers to any animal classified as a mammal, including human, domestic and farm animals, nonhuman primates, and zoo, sports, or pet animals, such as dogs, horses, cats, cows, etc.
  • epitope tag polypeptide when used herein in the context of a polypeptide refers to a polypeptide fused to an "epitope tag".
  • the epitope tag polypeptide has enough residues to provide an epitope against which an antibody can be made, yet is short enough such that it does not interfere with activity of the polypeptide.
  • the epitope tag preferably also is fairly unique so that the antibody does not substantially cross-react with other epitopes.
  • Suitable tag polypeptides generally have at least 6 amino acid residues and usually between about 8-50 amino acid residues (preferably between about 9-30 residues). Examples include the flu HA tag polypeptide and its antibody 12CA5 (Field et al, Mol Cell. Biol.
  • the epitope tag may be an epitope of the Fc region of an IgG molecule (e.g., IgGI , lgG2, lgG3 or lgG4) that is responsible for increasing the in vivo serum half-life of the IgG molecule.
  • an IgG molecule e.g., IgGI , lgG2, lgG3 or lgG4
  • label when used herein refers to a detectable compound or composition which can be conjugated directly or indirectly to a molecule or protein, e.g., an antibody.
  • the label may itself be detectable (e.g., radioisotope labels or fluorescent labels) or, in the case of an enzymatic label, may catalyze chemical alteration of a substrate compound or composition which is detectable.
  • solid phase means a non-aqueous matrix to which the antibody of the present invention can adhere.
  • solid phases encompassed herein include those formed partially or entirely of glass (e.g. controlled pore glass), polysacchahdes (e.g., agarose), polyacrylamides, polystyrene, polyvinyl alcohol and silicones.
  • the solid phase can comprise the well of an assay plate; in others it is a purification column (e.g. an affinity chromatography column).
  • IgE-mediated disorder means a condition or disease which is characterized by the overproduction and/or hypersensitivity to the immunoglobulin IgE. Specifically it would be construed to include conditions associated with anaphylactic hypersensitivity and atopic allergies, including for example: asthma, allergic rhinitis & conjunctivitis (hay fever), eczema, urticaria, atopic dermatitis, and food allergies.
  • aphylactic hypersensitivity including for example: asthma, allergic rhinitis & conjunctivitis (hay fever), eczema, urticaria, atopic dermatitis, and food allergies.
  • the serious physiological condition of anaphylactic shock caused by, e.g., bee stings, snake bites, food or medication is also encompassed under the scope of this term.
  • the starting or "parent” antibody may be prepared using techniques available in the art for generating such antibodies. These techniques are well known. Exemplary methods for generating the starting antibody are described in more detail in the following sections. These descriptions are possible alternatives for making or selecting a parent antibody and in no way limit the methods by which such a molecule may be generated.
  • the antibody's binding affinity is determined prior to generating a high affinity antibody of the present invention.
  • the antibody may be subjected to other biological activity assays, e.g., in order to evaluate effectiveness as a therapeutic.
  • Such assays are known in the art and depend on the target and intended use for the antibody.
  • a routine cross-blocking assay such as that described in "Antibodies: A Laboratory Manual” (Cold Spring Harbor Laboratory, Ed Harlow and David Lane (1988)) can be performed.
  • epitope mapping can be performed to determine where the antibody binds an epitope of interest.
  • the binding affinity of the antibody for a homolog of the target used to generate the antibody may be assessed using techniques known in the art.
  • the other species is a nonhuman mammal to which the antibody will be administered in preclinical studies.
  • the species may be a nonhuman primate, such as rhesus, cynomolgus, baboon, chimpanzee and macaque.
  • the species may be a rodent, cat or dog, for example.
  • the parent antibody is altered according to the present invention so as to generate an antibody which has a higher or stronger binding affinity for the target than the parent antibody.
  • Antibody specificity results from the unique interface that is formed between the antibody and its target; the surfaces complement each other to produce a unique fit (Jones, S. & Thornton, J. M. (1996) Proc. Natl. Acad. Sci. USA 93: 13-20).
  • the overall affinity can increase as a result of the lower energy cost needed to favor the association of the binding partners.
  • the binding surface of the antibody is generally composed of six complemetarity determining regions (CDRs) which are loops that extend out from the core.
  • CDRs complemetarity determining regions
  • the CDRs are composed of amino acids having a sequence that is unique for binding to the specific target.
  • the environment around these amino acids must become more favorable by introducing or improving various noncovalent forces, which ultimately lowers the energetics of the interaction, resulting in higher affinity.
  • Van der Waals forces are noncovalent interactions which occur between two electrically neutral molecules (Voet, D. & Voet, J. G. (1990) Biochemistry John Wiley and Sons, NY, NY). Associations can occur between two surfaces from electrostatic interactions that arise from permanent or induced dipoles. These dipoles can exist along the ends of a-helices or near polar amino acids. By increasing the number of van der Waals forces along a binding interface, a more favorable association will result.
  • the resulting high affinity antibody preferably has a binding affinity for the target which is at least about 10 fold higher, or at least about 20 fold higher, or at least about 500 fold higher or may be 1000 to 5000 fold higher, than the binding affinity of the parent antibody for the target.
  • the degree of enhancement in binding affinity necessary or desired will depend on the initial binding affinity of the parent antibody.
  • the method for making high affinity antibodies from a parent antibody involves the following steps:
  • [0066] Obtaining or selecting a parent antibody which binds the target of interest, which comprises heavy and light chain variable domains. This may be done by traditional hybridoma techniques, phage-display techniques, or any other method of generating a target specific antibody.
  • a framework sequence which is close in sequence to the parent framework preferably a human template sequence.
  • This template may be chosen based on, e.g., its comparative overall length, the size of the CDRs, the amino acid residues located at the junction between the framework and the CDRs, overall homology, etc.
  • the template chosen can be a mixture of more than one sequence or may be a consensus template.
  • [0068] Generating a library of clones by making random amino acid substitutions at each and every possible CDR position.
  • One may also randomly substitute the amino acids in the human framework template that are, e-.g., adjacent to the CDRs or that may affect binding or folding, with all possible amino acids, generating a library of framework substitutions. These framework substitutions can be assessed for their potential effect on target binding and antibody folding.
  • the substitution of amino acids in the framework may be done either simultaneously or sequentially with the substitution of the amino acids in the CDRs.
  • step (3) Constructing an expression vector comprising the heavy and/or light chain variants generated in step (3) which may comprise the formulas: FRH1-CDRH1- FRH2-CDRH2-FRH3-CDRH3-FRH4(I) and FRL1-CDRL1-FRL2-CDRL2-FRL3- CDRL3-FRL4 (II), wherein FRL1 , FRL2, FRL3, FRL4, FRH1 , FRH2, FRH3 and FRH4 represent the variants of the framework template light chain and heavy chain sequences chosen in step 3 and the CDRs represent the variant CDRs of the parent antibody CDRs.
  • An example of a vector containing such light and heavy chain sequences is depicted in Figure 1.
  • Soluble targets or fragments thereof can be used as immunogens for generating antibodies.
  • the antibody is directed against the target of interest.
  • the target is a biologically important polypeptide and administration of the antibody to a mammal suffering from a disease or disorder can result in a therapeutic benefit in that mammal.
  • antibodies may be directed against non polypeptide targets.
  • the target is a polypeptide, it may be a transmem rane molecule (e.g. receptor) or ligand such as a growth factor.
  • One target of the present invention is IgE. Whole cells may be used as the immunogen for making antibodies.
  • the target may be produced recombinantly or made using synthetic methods.
  • the target may also be isolated from a natural source.
  • Antigens used in producing antibodies of the invention may include polypeptides and polypeptide fragments of the invention, including epitope A and/or B.
  • a polypeptide used to immunize an animal can be obtained by standard recombinant, chemical synthetic, or purification methods.
  • an antigen in order to increase immunogenicity, an antigen can be conjugated to a carrier protein.
  • Commonly used carriers include, but are not limited to, keyhole limpet hemocyanin (KLH), thyroglobulin, bovine serum albumin (BSA), and tetanus toxoid.
  • KLH keyhole limpet hemocyanin
  • BSA bovine serum albumin
  • tetanus toxoid tetanus toxoid.
  • the coupled peptide is then used to immunize an animal (e.g., a mouse, a rat, or a rabbit).
  • well known adjuvants can be administered with the antigen to facilitate induction of
  • Polyclonal antibodies are usually generated in non-human mammals by multiple subcutaneous (sc) or intraperitoneal (ip) injections of the relevant target in combination with an adjuvant. Numerous agents capable of eliciting an immunological response are well known in the art.
  • the mammalian antibody selected will normally have a sufficiently strong binding affinity for the target.
  • the antibody may bind the human anti-IgE target with a binding affinity (Kd) value of about 1 x 10 "8 M.
  • Bind binding affinity
  • Antibody affinities may be determined by saturation binding; enzyme-linked immunoabso bant assay (ELISA); and competition assays (e.g., radioimmunoassays).
  • Monoclonal antibodies are antibodies which recognize a single antigenic site. Their uniform specificity makes monoclonal antibodies much more useful than polyclonal antibodies, which usually contain antibodies that recognize a variety of different antigenic sites. Monoclonal antibodies may be made using the hybridoma method first described by Kohler et al., Nature, 256: 495 (1975), or may be made by recombinant DNA methods.
  • a mouse or other appropriate host animal such as a rodent
  • lymphocytes that produce or are capable of producing antibodies that will specifically bind to the protein used for immunization.
  • lymphocytes may be immunized in vitro. Lymphocytes then are fused with myeloma cells using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principals and Practice, pp. 590-103 (Academic Press, 1986)).
  • the hybridoma cells thus prepared are seeded and grown in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells.
  • a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells.
  • the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (HAT medium), substances which prevent the growth of HGPRT-deficient cells.
  • HAT medium hypoxanthine, aminopterin, and thymidine
  • Preferred myeloma cells are those that fuse efficiently, support stable high-level production of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium.
  • the clones may be subcloned by limiting dilution procedures and grown by standard methods (Goding, Monoclonal Antibodies: Principals and Practice, pp. 59-103, Academic Press, 1986)). Suitable culture media for this purpose include.
  • the monoclonal antibodies secreted by the subclones are suitably separated from the culture medium by conventional immunoglobulin purification procedures such as, for example, protein A- Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
  • DNA encoding the monoclonal antibodies is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the monoclonal antibodies).
  • the hybridoma cells serve as a source of such DNA.
  • the DNA may be placed into expression vectors, which are then transferred into host cells such as E. coli cells, NS0 cells, Chinese hamster ovary (CHO) cells, or myeloma cells to obtain the synthesis of monoclonal antibodies in the recombinant host cells.
  • the DNA also may be modified, for example, by substituting the coding sequence for human heavy- and light-chain constant domains in place of the homologous murine sequences (U.S. Pat. No. 4,816,567; Morrison et al., Proc. Natl Acad. Sci. USA 81 : 6851 (1984)), or by covalently joining to the immunoglobulin polypeptide.
  • Humanized Antibodies for example, by substituting the coding sequence for human heavy- and light-chain constant domains in place of the homologous murine sequences (U.S. Pat. No. 4,816,567; Morrison et al., Proc. Natl Acad. Sci. USA 81 : 6851 (1984)), or by covalently joining to the immunoglobulin polypeptide.
  • Humanization is a technique for making a chimeric antibody wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species.
  • a humanized antibody has one or more amino acid residues introduced into it from a source which is non- human. These non-human amino acid residues are often referred to as "import" residues, which are typically taken from an "import" variable domain.
  • Humanization can be essentially performed following the method of Winter and co-workers (Jones et al, Nature 321: 522-525 (1986); Riechman et al., Nature 332: 323-327 (1988); Verhoeyens et al., Science 239: 1534-1536 (1988)), by substituting non-human CDR's or CDR sequences for the corresponding sequences in a human antibody (See, e.g., U.S. Pat. No. 4,816,567).
  • the humanized antibody may have some CDR residues and some FR residues substituted by residues from analogous sites in murine antibodies.
  • variable domains both light and heavy
  • sequence of the variable domain of a non-human antibody is compared with the library of known human variable- domain sequences.
  • the human sequence which is closest to that of the non- human parent antibody is then accepted as the human framework for the humanized antibody (Sims et al., J. Immunol. 151 : 2296 (1993); Chothia et al., J. Mol. Biol. 196: 901 (1987)).
  • Another method uses a particular framework derived from the consensus sequence of all human antibodies of a particular subgroup of light or heavy chains.
  • F(ab') 2 fragments can be isolated directly from recombinant host cell culture.
  • Other techniques for the production of antibody fragments will be apparent to the skilled practitioner.
  • the antibody of choice is a single chain Fv fragment (scFv). (PCT patent application WO 93/16185). PREPARATION OF HIGH AFFINITY ANTIBODIES
  • one or more amino acid residues may be altered in one or more of the variable regions of the parent antibody.
  • one or more substitutions of framework residues may be introduced in the parent antibody where these result in an improvement in the binding affinity of the antibody, for example, for human IgE.
  • framework region residues to modify include those which non- covalently bind target directly (Amit et al. Science 233: 747-753 (1986)); interact with/effect the conformation of CDR (Chothia et al. J. Mol. Biol. 196: 901-917 (1987)); and/or participate in the VL-VH interface (EP 239 400 B1 ).
  • modification of one or more of such framework region residues results in an enhancement of the binding affinity of the antibody for the target of interest.
  • Modifications in the antibodies' biological properties may be accomplished by selecting substitutions that differ significantly in their effect on maintaining, e.g., (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation; (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain.
  • substitutions that differ significantly in their effect on maintaining, e.g., (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation; (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain.
  • Non-conservative substitutions will entail exchanging a member of one of these classes for another class.
  • Nucleic acid molecules encoding amino acid sequence variants are prepared by a variety of methods known in the art. These methods include, but are not limited to, oligonucleotide-mediated (or site-directed) mutagenesis, PCR mutagenesis, and cassette mutagenesis of an earlier prepared variant or a non-variant version of the species-dependent antibody.
  • the preferred method for generating variants is an oligonucleotide-mediated synthesis.
  • the antibody variant will only have a single hypervariable region residue substituted, e.g. from about two to about fifteen hypervariable region substitutions.
  • One method for generating the library of variants is by oligonucleotide mediated synthesis according to the scheme depicted in Figure 2.
  • Three oligonucleotides of approximately 100 nucleotides each may be synthesized spanning the entire light chain or heavy chain variable region.
  • Each oligonucleotide may comprise: (1 ) a 60 amino acid stretch generated by the triplet (NNK) 20 where N is any nucleotide and K is G or T, and (2) an approximately 15-30 nucleotide overlap with either the next oligo or with the vector sequence at each end.
  • the polymerase Upon annealing of these three oligonucleotides in a PCR reaction, the polymerase will fill in the opposite strand generating a complete double stranded heavy chain or light chain variable region sequence.
  • the number of triplets may be adjusted to any length of repeats and their position within the oligonucleotide may be chosen so as to only substitute amino acds in a given CDR or framework region.
  • NNK all twenty amino acids are possible at each position in the encoded variants.
  • the overlapping sequence of 5-10 amino acids (15-30 nucloetides) will not be subtituted, but this may be chosen to fall within the stacking regions of the framework, or may substituted by a separate or subsequent round of synthesis.
  • the library of heavy and light chain variants can be constructed in any expression vector, such as a bacteriophage, specifically the vector of Fig.1 , each of which contains DNA encoding a particular heavy and light chain variant.
  • the biological activity of variant relative to the parent antibody is determined. As noted above, this involves determining the binding affinity of the variant for the target. Numerous high- throughput methods exist for rapidly screen antibody variants for their ability to bind the target of interest.
  • One or more of the antibody variants selected from this initial screen may then be screened for enhanced binding affinity relative to the parent antibody.
  • One common method for determining binding affinity is by assessing the association and dissociation rate constants using a BIAcoreTM surface plasmon resonance system (BIAcore, Inc.).
  • a biosensor chip is activated for covalent coupling of the target according to the manufacturer's (BIAcore) instructions.
  • the target is then diluted and injected over the chip to obtain a signal in response units (RU) of immobilized material. Since the signal in RU is proportional to the mass of immobilized material, this represents a range of immobilized target densities on the matrix.
  • Dissociation data are fit to a one-site model to obtain k 0ff +/- s.d. (standard deviation of measurements).
  • Pseudo-first order rate constant (k s ) are calculated for each association curve, and plotted as a function of protein concentration to obtain k o ⁇ +/- s.e. (standard error of fit).
  • Equilibrium dissociation constants for binding, K D 'S are calculated from SPR measurements as ko ff /k on - Since the equilibrium dissociation constant, K D , is inversely proportional to k 0ff , an estimate of affinity improvement can be made assuming the association rate (k on ) is a constant for all variants.
  • the resulting candidate(s) with high affinity may optionally be subjected to one or more further biological activity assays to confirm that the antibody variant(s) with enhanced binding affinity still retain the desired therapeutic attributes.
  • the optimal antibody variant retains the ability to bind the target with a binding affinity significantly higher than the parent antibody.
  • the antibody variant(s) so selected may be subjected to further modifications oftentimes depending upon the intended use of the antibody. Such modifications may involve further alteration of the amino acid sequence, fusion to heterologous polypeptide(s) and/or covalent modifications such as those elaborated below.
  • any cysteine residues not involved in maintaining the proper conformation of the antibody variant may be substituted, generally with serine, to improve the oxidative stability of the molecule and prevent aberrant cross linking.
  • cysteine bond(s) may be added to the antibody to improve its stability (particularly where the antibody is an antibody fragment such as an Fv fragment).
  • the invention also provides isolated nucleic acid encoding an antibody variant as disclosed herein, vectors and host cells comprising the nucleic acid, and recombinant techniques for the production of the antibody variant.
  • the nucleic acid encoding it is isolated and inserted into a replicable vector for further cloning (amplification of the DNA) or for expression.
  • DNA encoding the antibody variant is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the antibody variant).
  • the vector components generally include, but are not limited to, one or more of the following: a signal sequence, an origin of replication, one or more marker genes, an enhancer element, a promoter, and a transcription termination sequence.
  • the phage expression vector depicted in Figure 1 is comprised of a commonly used M13 vector and M13's own gene III viral secretion signal for rapid secretion and screening variant Fabs for proper binding specificity and minimal affinity criteria.
  • This vector does not use the entire gene ill sequence, so there is no display on the surface of the bacterial cell, but rather the Fabs are secreted into the periplasmic space.
  • the Fabs could be expressed in the cytoplasm and isolated.
  • the heavy and light chains each have their own viral secretion signal, but are dependently expressed from a single strong inducible promoter.
  • the vector in Figure 1 also provides a His tag and a myc tag for easy purification, as well as detection.
  • the Fabs could be independently expressed from separate promoters or that the secretion signal need not be the viral sequence chosen, but could be a prokaryotic or eukaryotic signal sequence suitable for the secretion of the antibody fragments from the chosen host cell. It should also be recognized that the heavy and light chains may reside on different vectors.
  • the antibody variant of this invention may be produced recombinantly.
  • the variant may also be expressed as a fusion polypeptide fused with a heterologous polypeptide, which is preferably a signal sequence or other polypeptide having a specific cleavage site at the N-terminus of the mature protein or polypeptide.
  • a heterologous polypeptide which is preferably a signal sequence or other polypeptide having a specific cleavage site at the N-terminus of the mature protein or polypeptide.
  • the heterologous signal sequence selected preferably is one that is recognized and processed (i.e., cleaved by signal peptidase) by the host cell.
  • the signal sequence may be substituted by a prokaryotic signal sequence selected, for example, from the group of the alkaline phosphatase, penicillinase, Ipp, or heat-stable enterotoxin II leaders. Or in the case of the vector of Figure 1 , the signal sequence chosen was a viral signal sequence from gene III.
  • the native signal sequence may be substituted by, e g., the yeast invertase leader, ⁇ -factor leader (including Saccharomyces and Kluyveromyces ⁇ - factor leaders), or acid phosphatase leader, the C.
  • Vectors usually contain a nucleic acid sequence that enables the vector to replicate in one or more selected host cells. Generally, this sequence is one that enables the vector to replicate independently of the host chromosomal DNA, and includes origins of replication or autonomously replicating sequences. Such sequences are well known for a variety of bacteria, yeast, and viruses.
  • the origin of replication from the plasmid pBR322 is suitable for most Gram-negative bacteria, the 2 ⁇ plasmid origin is suitable for yeast, and various viral origins (SV40, polyoma, adenovirus, VSV or BPV) are useful for vectors in mammalian cells.
  • the origin of replication component is not needed for mammalian expression vectors (the SV40 origin may typically be used only because it contains the early promoter).
  • Vectors may contain a selection gene, also termed a selectable marker.
  • Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., ampicillin, neomycin, methotrexate, or tetracycline, (b) complement auxotrophic deficiencies, or (c) supply critical nutrients not available from complex media, e.g., the gene encoding D-alanine racemase for Bacilli.
  • One example of a selection scheme utilizes a drug to arrest growth of a host cell. Those cells that are successfully transformed with a heterologous gene produce a protein conferring drug resistance and thus survive the selection regimen. Examples of such dominant selection use the drugs neomycin, mycophenolic acid and hygromycin.
  • Another example of suitable selectable markers for mammalian cells are those that enable the identification of cells competent to take up the antibody nucleic acid, such as DHFR, thymidine kinase, metallothionein-l and -II, preferably primate metallothionein genes, adenosine deaminase, omithine decarboxylase, etc.
  • cells transformed with the DHFR selection gene are first identified by culturing all of the transformants in a culture medium that contain methotrexate (Mtx), a competitive antagonist of DHFR.
  • Mtx methotrexate
  • An appropriate host cell when wild-type DHFR is employed is the Chinese hamster ovary (CHO) cell line deficient in DHFR activity.
  • host cells particularly wild-type hosts that contain endogenous DHFR transformed or co-transformed with DNA sequences encoding antibody, wild-type DHFR protein, and another selectable marker such as aminoglycoside 3'-phosphotransferase (APH) can be selected by cell growth in medium containing a selection agent for the selectable marker such as an aminoglycosidic antibiotic, e.g., kanamycin, neomycin, or G418. (U.S. Pat. No. 4,965,199).
  • APH aminoglycoside 3'-phosphotransferase
  • a suitable selection gene for use in yeast is the trpl gene present in the yeast plasmid Yrp7 (Stinchcomb et al., Nature 282: 39 (1979)).
  • the trpl gene provides a selection marker for a variant strain of yeast lacking the ability to grow in typtophan, for example, ATCC No. 44076 or PEP4-1 . Jones, Genetics 85: 12 (1977).
  • the presence of the trpl lesion in the yeast host cell genome then provides an effective environment for detecting transformation by growth in the absence of tryptophan.
  • Leu2-deficient yeast strains (ATCC 20,622 or 38,626) are complemented by known plasmids bearing the Leu2 gene.
  • Expression and cloning vectors usually contain a promoter that is recognized by the host organism and is operably linked to the antibody nucleic acid.
  • Promoters suitable for use with prokaryotic hosts include the phoA promoter, /?-lactamase and lactose promoter systems, alkaline phosphatase, a tryptophan (trp) promoter system, and hybrid promoters such as the tac promoter.
  • phoA promoter /?-lactamase and lactose promoter systems
  • alkaline phosphatase alkaline phosphatase
  • trp tryptophan
  • Other known bacterial promoters are suitable.
  • Promoters for use in bacterial systems may also contain a Shine-Dalgarno (S.D.) sequence operably linked to the DNA encoding the antibody.
  • S.D. Shine-Dalgarno
  • Virtually all eukaryotic genes have an AT-rich region located approximately 25 to 30 bases upstream from the site where transcription is initiated. Another sequence found 70 to 80 bases upstream from the start of transcription of many genes is a CNCAAT region where N may be any nucleotide. At the 3' end of most eukaryotic genes is an AATAAA sequence that may be the signal for addition of the poly A tail to the 3' end of the coding sequence. All of these sequences are suitably inserted into eukaryotic expression vectors.
  • suitable promotor sequences for use with yeast hosts include the promoters for 3-phosphoglycerate kinase or other glycolytic enzymes, such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, 3- phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase.
  • 3-phosphoglycerate kinase or other glycolytic enzymes such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, 3- phosphoglycerate mutase, pyruv
  • yeast promoters which are inducible promoters having the additional advantage of transcription controlled by growth conditions, are the promoter regions for alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, degradative enzymes associated with nitrogen metabolism, metallothionein, glyceraldehyde-3-phosphate dehydrogenase, and enzymes responsible for maltose and galactose utilization.
  • Suitable vectors and promoters for use in yeast expression are further described in EP 73,657.
  • Yeast enhancers also are advantageously used with yeast promoters.
  • Antibody transcription from vectors in mammalian host cells is controlled, for example, by promoters obtained from the genomes of viruses such as polyoma virus, fowlpox virus, adenovirus (such as Adenovirus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and most preferably Simian virus 40 (SV40), from heterologous mammalian promoters, e.g., the actin promoter or an immunoglobulin promoter, from heat-shock promoters- provided such promoters are compatible with the host cell systems.
  • viruses such as polyoma virus, fowlpox virus, adenovirus (such as Adenovirus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and most preferably Simian virus 40 (SV40), from
  • the early and late promoters of the SV40 virus are conveniently obtained as an SV40 restriction fragment that also contains the SV40 viral origin of replication.
  • the immediate early promoter of the human cytomegalovirus is conveniently obtained as a Hindi II E restriction fragment.
  • a system for expressing DNA in mammalian hosts using the bovine papilloma virus as a vector is disclosed in U.S. Pat. No. 4,419,446. A modification of this system is described in U.S. Pat. No. 4,601 ,978.
  • human /?-interferon cDNA has been expressed in mouse cells under the control of a thymidine kinase promoter from herpes simplex virus.
  • the rous sarcoma virus long terminal repeat can be used as the promoter.
  • Enhancer sequences are now known from mammalian genes (globin, elastase, albumin, ⁇ -fetoprotein, and insulin). Typically, however, one will use an enhancer from a eukaryotic cell virus. Examples include the SV40 enhancer on the late side of the replication origin (bp 100-270), the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers.
  • the enhancer may be spliced into the vector at a position 5' or 3' to the antibody-encoding sequence, but is preferably located at a site 5' from the promoter.
  • Expression vectors used in eukaryotic host cells may also contain sequences necessary for the termination of transcription and for stabilizing the mRNA. Such sequences are commonly available from the 5' and, occasionally 3', untranslated regions of eukaryotic or viral DNAs or cDNAs. These regions contain nucleotide segments transcribed as polyadenylated fragments in the untranslated portion of the mRNA encoding the antibody.
  • One useful transcription termination component is the bovine growth hormone polyadenylation region. See e.g.,WO94/1 1026. SELECTION AND TRANSFORMATION OF HOST CELLS
  • Suitable host cells for cloning or expressing the DNA in the vectors herein are prokaryotic, yeast, or higher eukaryotic cells.
  • Suitable prokaryotes for this purpose include both Gram-negative and Gram-positive organisms, for example, Enterobacteria such as E. coli, Enterobacter, Erwinia, Klebsiella, Proteus, Salmonella, Serratia, and Shigella, as well as Bacilli, Pseudomonas, and Streptomyces.
  • E. coli cloning host is E. coli 294 (ATCC 31 ,446), although other strains such as E. coli B, E. coli X1776 (ATCC 31 ,537), and E. coli W31 10 (ATCC 27,325) are suitable. These examples are illustrative rather than limiting.
  • eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for antibody-encoding vectors.
  • Saccharomyces cerevisiae is the most commonly used among lower eukaryotic host microorganisms.
  • a number of other genera, species, and strains are commonly available and useful herein, such as Schizosaccharomyces pombe; Kluyveromyces; Candida; Trichoderma; Neurospora crassa; and filamentous fungi such as e.g., Neurospora, Penicillium, Tolypocladium, and Aspergillus hosts, such as A. nidulans and A. niger.
  • Suitable host cells for the expression of glycosylated antibodies are derived from multicellular organisms. In principal, any higher eukaryotic cell culture is workable, whether from vertebrate or invertebrate culture. Examples of invertebrate cells include plant and insect cells, Luckow et al., Bio/Technology 6, 47-55 (1988); Miller et al., Genetic Engineering, Setlow et al. eds. Vol. 8, pp. 277-279 (Plenam publishing 1986); Mseda et al., Nature 315, 592-594 (1985).
  • baculoviral strains and variants and corresponding permissive insect host cells from hosts such as Spodoptera frugiperda (caterpillar), Aedes (mosquito), Drosophila melanogaster (fruitfly), and Bombyx mori have been identified.
  • a variety of viral strains for transfection are publicly available, e.g., the L-1 variant of Autographa califomica NPV and the Bm-5 strain of Bombyx mori NPV, and such viruses may be used as the virus herein according to the present invention, particularly for transfection of Spodoptera frugiperda cells.
  • plant cells cultures of cotton, corn, potato, soybean, petunia, tomato, and tobacco and also be utilized as hosts.
  • Vertebrate cells and propagation of vertebrate cells, in culture has become routine. See Tissue Culture, Academic Press, Kruse and Patterson, eds. (1973).
  • useful mammalian host cell lines are monkey kidney; human embryonic kidney line; baby hamster kidney cells; Chinese hamster ovary cells/- DHFR (CHO, Urlaub et al., Proc. Natl. Acad. Sci. USA 77: 4216 (1980)); mouse sertoli cells; human cervical carcinoma cells (HELA); canine kidney cells; human lung cells; human liver cells; mouse mammary tumor; and NSO cells.
  • Host cells are transformed with the above-described vectors for antibody production and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences.
  • the host cells used to produce the antibody variant of this invention may be cultured in a variety of media.
  • Commercially available media such as Ham's F10 (Sigma), Minimal Essential Medium (MEM, Sigma), RPMI-1640 (Sigma), and Dulbecco's Modified Eagle's Medium (DMEM, Sigma) are suitable for culturing host cells.
  • 4,767,704; 4,657,866; 4,560,655; 5,122,469; 5,712,163; or 6,048,728 may be used as culture media for the host cells. Any of these media may be supplemented as necessary with hormones and/or other growth factors (such as insulin, transferrin, or epidermal growth factor), salts (such as X-chlorides, where X is sodium, calcium, magnesium; and phosphates), buffers (such as HEPES), nucleotides (such as adenosine and thymidine), antibiotics (such as GENTAMYCIN.TM. drug), trace elements (defined as inorganic compounds usually present at final concentrations in the micromolar range), and glucose or an equivalent energy source.
  • hormones and/or other growth factors such as insulin, transferrin, or epidermal growth factor
  • salts such as X-chlorides, where X is sodium, calcium, magnesium; and phosphates
  • buffers such as HEPES
  • the antibody variant can be produced intracellulariy, in the periplasmic space, or directly secreted into the medium. If the antibody variant is produced intracellulariy, as a first step, the particulate debris, either host cells or lysed fragments, may be removed, for example, by centrifugation or ultrafiltration. Carter et al., Bio/Technology 10: 163-167 (1992) describe a procedure for isolating antibodies which are secreted to the periplasmic space of E. coli. Briefly, cell paste is thawed in the presence of sodium acetate (pH 3.5), EDTA, and phenylmethylsulfonylfluoride (PMSF) over about 30 minutes.
  • sodium acetate pH 3.5
  • EDTA EDTA
  • PMSF phenylmethylsulfonylfluoride
  • Cell debris can be removed by centrifugation.
  • supernatants from such expression systems are generally first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit.
  • a protease inhibitor such as PMSF may be included in any of the foregoing steps to inhibit pr ⁇ teolysis and antibiotics may be included to prevent the growth of adventitious contaminants.
  • the antibody composition prepared from the cells can be purified using, for example, hydroxylapatite chromatography, gel elecrophoresis, dialysis, and affinity chromatography, with affinity chromatography being the preferred purification technique.
  • affinity chromatography is the preferred purification technique.
  • the suitability of protein A as an affinity ligand depends on the species and isotype of any immunoglobulin Fc domain that is present in the antibody variant. Protein A can be used to purify antibodies that are based on human IgGI , lgG2 or lgG4 heavy chains (Lindmark et al., J. Immunol Meth. 62: 1- 13 (1983)).
  • Protein G is recommended for all mouse isotypes and for human lgG3 (Guss et al., EMBO J. 5: 1567-1575 (1986)).
  • the matrix to which the affinity ligand is attached is most often agarose, but other matrices are available. Mechanically stable matrices such as controlled pore glass or poly(styrenedivinyl)benzene allow for faster flow rates and shorter processing times than can be achieved with agarose.
  • the antibody variant comprises a CH3 domain
  • the Bakerbond ABXTM resin J. T. Baker, Phillipsburg, N.J.
  • the mixture comprising the antibody variant of interest and contaminants may be subjected to low pH hydrophobic interaction chromatography using an elution buffer at a pH between about 2.5-4.5, preferably performed at low salt concentrations (e.g., from about 0-0.25M salt).
  • low salt concentrations e.g., from about 0-0.25M salt.
  • Therapeutic formulations of the polypeptide or antibody may be prepared for storage as lyophilized formulations or aqueous solutions by mixing the polypeptide having the desired degree of purity with optional "pharmaceutically-acceptable" carriers, excipients or stabilizers typically employed in the art (all of which are termed “excipients”).
  • excipients typically employed in the art
  • buffering agents, stabilizing agents, preservatives, isotonifiers, non-ionic detergents, antioxidants and other miscellaneous additives See Remington's Pharmaceutical Sciences, 16th edition, A. Osol, Ed. (1980)).
  • Such additives must be nontoxic to the recipients at the dosages and concentrations employed.
  • Buffering agents help to maintain the pH in the range which approximates physiological conditions. They are preferably present at concentration ranging from about 2 mM to about 50 mM.
  • Suitable buffering agents for use with the present invention include both organic and inorganic acids and salts thereof such as citrate buffers (e.g., monosodium citrate-disodium citrate mixture, citric acid- trisodium citrate mixture, citric acid-monosodium citrate mixture, etc.), succinate buffers (e.g., succinic acid-monosodium succinate mixture, succinic acid-sodium hydroxide mixture, succinic acid-disodium succinate mixture, etc.), tartrate buffers (e.g., tartaric acid-sodium tartrate mixture, tartaric acid-potassium tartrate mixture, tartaric acid-sodium hydroxide mixture, etc.), fumarate buffers (e.g., fumaric acid- monosodium fumarate mixture, etc.), fuma
  • Preservatives may be added to retard microbial growth, and may be added in amounts ranging from 0.2%-1 % (w/v).
  • Suitable preservatives for use with the present invention include phenol, benzyl alcohol, meta-cresol, methyl paraben, propyl paraben, octadecyldimethylbenzyl ammonium chloride, benzalconium halides (e.g., chloride, bromide, iodide), hexamethonium chloride, alkyl parabens such as methyl or propyl paraben, catechol, resorcinol, cyclohexanol, and 3- pentanol.
  • Isotonicifiers sometimes known as “stabilizers” may be added to ensure isotonicity of liquid compositions of the present invention and include polhydric sugar alcohols, preferably trihydric or higher sugar alcohols, such as glycerin, erythritol, arabitol, xylitol, sorbitol and mannitol.
  • polhydric sugar alcohols preferably trihydric or higher sugar alcohols, such as glycerin, erythritol, arabitol, xylitol, sorbitol and mannitol.
  • Stabilizers refer to a broad category of excipients which can range in function from a bulking agent to an additive which solubilizes the therapeutic agent or helps to prevent denaturation or adherence to the container wall.
  • Typical stabilizers can be polyhydric sugar alcohols (enumerated above); amino acids such as arginine, lysine, glycine, glutamine, asparagine, histidine, alanine, ornithine, L-leucine, 2- phenylalanine, glutamic acid, threonine, etc., organic sugars or sugar alcohols, such as lactose, trehalose, stachyose, mannitol, sorbitol, xylitol, ribitol, myoinisitol, galactitol, glycerol and the like, including cyclitols such as inositol; polyethylene glycol; amino acid polymers; sulfur containing reducing agents, such as
  • proteins such as human serum albumin, bovine serum albumin, gelatin or immunoglobulins
  • hydrophylic polymers such as polyvinylpyrrolidone monosaccharides, such as xylose, mannose, fructose, glucose; disaccharides such as lactose, maltose, sucrose and trisaccacharides such as raffinose; polysaccharides such as dextran.
  • Stabilizers may be present in the range from 0.1 to 10,000 weights per part of weight active protein.
  • Non-ionic surfactants or detergents may be added to help solubilize the therapeutic agent as well as to protect the therapeutic protein against agitation-induced aggregation, which also permits the formulation to be exposed to shear surface stressed without causing denaturation of the protein.
  • Suitable non-ionic surfactants include polysorbates (20, 80, etc.), polyoxamers (184, 188 etc.), Pluronic® polyols, polyoxyethylene sorbitan monoethers (TWEEN ⁇ -20, TWEEN®-80, etc.).
  • Non-ionic surfactants may be present in a range of about 0.05 mg/m! to about 1.0 mg/ml, preferably about 0.07 mg/ml to about 0.2 mg/ml.
  • Additional miscellaneous excipients include bulking agents, (e.g. starch), chelating agents (e.g. EDTA), antioxidants (e.g., ascorbic acid, methionine, vitamin E), and co-solvents.
  • the formulation herein may also contain more than one active compound as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other. For example, it may be desirable to further provide an immunosuppressive agent.
  • Such molecules are suitably present in combination in amounts that are effective for the purpose intended.
  • the active ingredients may also be entrapped in microcapsule prepared, for example, by coascervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsule and poly-(methylmethacylate) microcapsule, respectively, in colloidal drug delivery systems (for example, liposomes, albumin micropheres, microemulsions, nano- particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin micropheres, microemulsions, nano- particles and nanocapsules
  • sustained-release preparations include semi-permeable matrices of solid hydrophobic polymers containing the antibody variant, which matrices are in the form of shaped articles, e.g., films, or microcapsules.
  • sustained- release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl- methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No.
  • copolymers of L-glutamic acid and ethyl-L-glutamate non-degradable ethylene- vinyl acetate
  • degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOTTM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate)
  • poly-D-(-)-3-hydroxybutyric acid While polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for over 100 days, certain hydrogels release proteins for shorter time periods.
  • encapsulated antibodies When encapsulated antibodies remain in the body for a long time, they may denature or aggregate as a result of exposure to moisture at 37° C resulting in a loss of biological activity and possible changes in immunogenicity. Rational strategies can be devised for stabilization depending on the mechanism involved. For example, if the aggregation mechanism is discovered to be intermolecular S--S bond formation through thio-disulfide interchange, stabilization may be achieved by modifying sulfhydryl residues, lyophilizing from acidic solutions, controlling moisture content, using appropriate additives, and developing specific polymer matrix compositions.
  • the amount of therapeutic polypeptide, antibody or fragment thereof which will be effective in the treatment of a particular disorder or condition will depend on the nature of the disorder or condition, and can be determined by standard clinical techniques. Where possible, it is desirable to determine the dose-response curve and the pharmaceutical compositions of the invention first in vitro, and then in useful animal model systems prior to testing in humans.
  • an aqueous solution of therapeutic polypeptide, antibody or fragment thereof is administered by subcutaneous injection.
  • Each dose may range from about 0.5 ⁇ g to about 50 ⁇ g per kilogram of body weight, or more preferably, from about 3 ⁇ g to about 30 ⁇ g per kilogram body weight.
  • the dosing schedule for subcutaneous administration may vary form once a month to daily depending on a number of clinical factors, including the type of disease, severity of disease, and the subject's sensitivity to the therapeutic agent. USES FOR THE ANTIBODY VARIANT
  • the antibody variants of the invention may be used as affinity purification agents.
  • the antibodies are immobilized on a solid phase such as SEPHADEXTM resin or filter paper, using methods well known in the art.
  • the immobilized antibody variant is contacted with a sample containing the target to be purified, and thereafter the support is washed with a suitable solvent that will remove substantially all the material in the sample except the target to be purified, which is bound to the immobilized antibody variant. Finally, the support is washed with another suitable solvent, such as glycine buffer, that will release the target from the antibody variant.
  • the variant antibodies may also be useful in diagnostic assays, e.g., for detecting expression of a target of interest in specific cells, tissues, or serum.
  • the antibody variant typically will be labeled with a detectable moiety.
  • Numerous labels are available Techniques for quantifying a change in fluorescence are described above.
  • the chemiluminescent substrate becomes electronically excited by a chemical reaction and may then emit light which can be measured (using a chemiluminometer, for example) or donates energy to a fluorescent acceptor.
  • Examples of enzymatic labels include luciferases (e.g., firefly luciferase and bacterial luciferase; U.S. Pat. No.
  • luciferin 2,3- dihydrophthalazinediones, malate dehydrogenase, urease, peroxidase such as horseradish peroxidase (HRPO), alkaline phosphatase, .beta.-galactosidase, glucoamylase, lysozyme, saccharide oxidases (e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase), heterocyclic oxidases (such as uricase and xanthine oxidase), lactoperoxidase, microperoxidase, and the like.
  • HRPO horseradish peroxidase
  • alkaline phosphatase .beta.-galactosidase
  • glucoamylase lysozyme
  • saccharide oxidases e.g., glucose oxidase, galactose
  • Biotin binds selectively to avidin and thus, the label can be conjugated with the antibody variant in this indirect manner.
  • the antibody variant is conjugated with a small hapten (e.g. digloxin) and one of the different types of labels mentioned above is conjugated with an anti-hapten antibody variant (e.g. anti-digioxin antibody).
  • a small hapten e.g. digloxin
  • an anti-hapten antibody variant e.g. anti-digioxin antibody
  • the antibody variant need not be labeled, and the presence thereof can be detected using a labeled antibody which binds to the antibody variant.
  • the antibodies of the present invention may be employed in any known assay method, such as competitive binding assays, direct and indirect sandwich assays, and immunoprecipitation assays. Zola, Monoclonal Antibodies: A Manual of Techniques, pp. 147-158 (CRC Press, Inc. 1987).
  • Competitive binding assays rely on the ability of a labeled standard to compete with the test sample for binding with a limited amount of antibody variant. The amount of target in the test sample is inversely proportional to the amount of standard that becomes bound to the antibodies. To facilitate determining the amount of standard that becomes bound, the antibodies generally are insolubilized before or after the competition. As a result, the standard and test sample that are bound to the antibodies may conveniently be separated from the standard and test sample which remain unbound.
  • Sandwich assays involve the use of two antibodies, each capable of binding to a different immunogenic portion, or epitope, or the protein to be detected.
  • the test sample to be analyzed is bound by a first antibody which is immobilized on a solid support, and thereafter a second antibody binds to the test sample, thus forming an insoluble three-part complex.
  • the second antibody may itself be labeled with a detectable moiety (direct sandwich assays) or may be measured using an anti-immunoglobulin antibody that is labeled with a detectable moiety (indirect sandwich assay).
  • sandwich assay is an ELISA assay, in which case the detectable moiety is an enzyme.
  • the tumor sample may be fresh or frozen or may be embedded in paraffin and fixed with a preservative such as formalin, for example.
  • the antibodies may also be used for in vivo diagnostic assays.
  • the antibody variant is labeled with a radionucleotide (such as .sup.111 In, .sup.99 Tc, .sup.14 C, .sup.131 I, .sup.3 H, .sup.32 P or .sup.35 S) so that the tumor can be localized using immunoscintiography.
  • a high affinity anti-IgE antibody of the present invention may be used to detect the amount of IgE present in, e.g., the lungs of an asthmatic patient.
  • the antibody of the present invention can be provided in a kit, i.e., packaged combination of reagents in predetermined amounts with instructions for performing the diagnostic assay.
  • the kit may include substrates and cofactors required by the enzyme , (e.g., a substrate precursor which provides the detectable chromophore or fluorophore).
  • substrates and cofactors required by the enzyme e.g., a substrate precursor which provides the detectable chromophore or fluorophore.
  • other additives may be included such as stabilizers, buffers (e.g., a block buffer or lysis buffer) and the like.
  • the relative amounts of various reagents may be varied widely to provide for concentrations in solution the reagents which substantially optimize the sensitivity of the asssy.
  • the reagents may be provided as dry powders, usually lyophilized, eluding excipients which on dissolution will provide a reagent solution having the ppropriate concentration.
  • the antibodies of the present invention may be used to treat a mammal.
  • the antibody is administered to a nonhuman mammal for the purposes of obtaining preclinical data, for example.
  • exemplary nonhuman mammals to be treated include nonhuman primates, *dogs, cats, rodents and other mammals in which preclinical studies are perf ormed.
  • Such mammals may be established animal models for a disease to b «e treated with the antibody or may be used to study toxicity of the antibody of interest.
  • dose escalation studies may be perform d on the mammal.
  • the antibody or polypeptide is administered by any suitable means, including parenteral, subcutaneous, intraperitoneal, intrapulmonary, an ⁇ l intranasal, and, if desired for local immunosuppressive treatment, intralesional administration.
  • Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration.
  • the antibody variant is suitably administered by pulse infusion, particularly with declining doses of the antibody variant.
  • the dosing is given by injectio s, most preferably intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic.
  • the appropriate dosage of the antibody or polypeptide will depend on the type of disease to be treated, the severity and course of the disease, whether the antibody variant is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody variant, and the discretion of e attending physician.
  • the very high affinity anti-human IgE antibodies of the invention may be suitably administered to the patient at one time or over a series of treatments.
  • about 0.1 mg/kg to 150 mg7kg (e.g., 0.1-20 mg/kg) of antibody is an initial candidates dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion.
  • a typical daily dosage might range from about 1 mg/kg to 100 mg/kg or more, depending on the factors mentioned above.
  • the treatment is sustained until a desired suppression of disease symptoms occurs.
  • other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
  • An exemplary dosing regimen for an anti-LFA-1 or anti-ICAM-1 antibody is disclosed in WO 94/04188. 150] The antibody variant composition will be formulated, dosed and administered in a manner consistent with good medical practice.
  • Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • the "therapeutically effective amount" of the antibody variant to be administered will be governed by such considerations, and is the minimum amount necessary to prevent, ameliorate, or treat a disease or disorder.
  • the antibody variant need not be, but is optionally formulated with one or more agents currently used to prevent or treat the disorder in question. The effective amount of such other agents depends on the amount of antibody present in the formulation, the type of disorder or treatment, and other factors discussed above.
  • the antibodies of the present invention which recognize IgE as their target may be used to treat "IgE-mediated disorders". These include diseases such as asthma, allergic rhinitis & conjunctivitis (hay fever), eczema, urticaria, atopic dermatitis, and food allergies.
  • IgE-mediated disorders include diseases such as asthma, allergic rhinitis & conjunctivitis (hay fever), eczema, urticaria, atopic dermatitis, and food allergies.
  • the serious physiological condition of anaphylactic shock caused by, e.g., bee stings, snake bites, food or medication is also encompassed under the scope of this invention.
  • epitope refers to a site on an antigen to which B and/or T cells respond.
  • B-cell epitopes can be formed both from contiguous amino acids or noncontiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids are typically retained on exposure to denaturing solvents, whereas epitopes formed by tertiary folding are typically lost on treatment with denaturing solvents.
  • An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation.
  • Antibodies that recognize the same epitope can be identified in a simple immunoassay showing the ability of one antibody to block the binding of another antibody to a target antigen.
  • Epitope mapping of the binding site for high affinity antibodies IgE of the present invention involved Western blot analysis for binding, a peptide scan of the CH3 domain of IgE, an alanine scan of the regions that showed binding, amino acid replacement from corresponding regions of IgGI and site directed mutagenesis.
  • the peptide scan of the entire CH3 domain of IgE required seventy-three overlapping peptides. Each peptide was subjected to binding by labeled anti-IgE antibodies of the present invention to determine the specific epitope(s) of IgE that block binding of IgE to its high affinity receptor.
  • the peptide scan identified two peptides as potential anti-IgE MAb contact sites on IgE, designated Epitope 'A' and Epitope 'B' (See Figure 11 ). Although the Epitope 'A' and Epitope 'B' sequences are about 80 amino acids apart in the linear sequence, they are positioned close to each other in the three dimensional structure of IgE.
  • Figure 12 illustrates the binding region of Epitope B as determined by ELISA using peptide scan.
  • the invention also relates to pharmaceutical compositions, e.g., vaccines, comprising the peptide immunogen molecules of the present invention, including SEQ ID NO 72 and/or SEQ ID NO 74, and diluents, excipients, adjuvants, or carriers. It further concerns a process for the preparation of an immunogen of the invention, comprising covalently coupling at least one peptide of the invention with a moiety capable of eliciting an immune response against that peptide.
  • immunogenic peptides as defined above, for use as a pharmaceutical, e.g. in the treatment of IgE-mediated diseases or conditions, such as allergy and atopic dermatitis.
  • It further relates to a method of immunizing a mammal against IgE-mediated diseases or conditions, such as allergies and atopic dermatitis, comprising the administration of a therapeutically effective amount of the immunogenic peptides as defined above to a patient in need of such treatment.
  • IgE-mediated diseases or conditions such as allergies and atopic dermatitis
  • the immunogenic peptides of the present invention while being substantially incapable of mediating non-cytolytic histamine release, are capable of eliciting antibodies with strong serological cross-reactivity with the target amino acid sequences of Epitope A and/or Epitope B.
  • the initial dose of peptide may be administered, for example, intramuscularly, followed by repeat (booster) doses of the same at 14 to 28 days later. Doses will of course depend to some extent on the age, weight and general health of the patient. Immunization may be "active" or “passive”. In “active” immunization the subject receives an immunogenic peptide of the present invention and an anti-IgE response is actively induced by the subject's immune system.
  • immunogenic carrier includes those materials which have the property of independently eliciting an immunogenic response in a host animal and which can be covalently coupled to polypeptide either directly via formation of peptide or ester bonds between free carboxyl, amino or hydroxyl groups in the polypeptide and corresponding groups on the immunogenic carrier material, or alternatively by bonding through a conventional bifunctional linking group, or as a fusion protein.
  • immunogenic carriers examples include: albumins, such as BSA; globulins; thyroglobulins; hemoglobins; hemocyanins (particularly Keyhole Limpet Hemocyanin [KLH]); proteins extracted from ascaris, e.g. ascaris extracts such as those described in J. Immunol. 111 [1973] 260-268, J. Immunol. 122 [1979] 302- 308, J. Immun. 98 [1967] 893-900, a Am. J. Physiol.
  • Vaccines have been produced using diphteria toxoid or tetanus toxoid as immunogenic carrier material (Lepow M. L. et al., J. of Infectious Diseases 150 [1984] 402-406; Coen Beuvery, E. et al., Infection and Immunity 40 [1983] 39-45) and these toxoid materials can also be used in the present invention.
  • the purified protein derivative of tuberculin is particularly preferred for utilization in the "active" immunization scheme since (I) it does not induce a T-cell response itself (i.e. it is in effect a "T-cell hapten"), and yet behaves as a fully processed antigen and is recognized by T- cells as such; (2) it is known to be one of the most powerful hapten "carriers" in the linked recognition mode; and (3) it can be used in humans without further testing.
  • the present invention also relates to polynucleotides encoding the peptides of the present invention, vectors comprising said polynucleotides, and cells harboring said vectors.
  • active immunization may be achieved by administering the polynucelotides encoding the peptides of the present invention.
  • Vectors suitable for such therapy are known in the art and include, e.g., adenovirus vectors.
  • Passive immunization is achieved by administering anti-IgE antibodies of the present invention, to a patient suffering from IgE-mediated disease or condition.
  • These antibodies can be prepared by administering an immunogenic peptide of the present invention to a non-human mammal and collecting the resultant antiserum. Improved titres can be obtained by repeated injections over a period of time.
  • mammals which may be used for eliciting antibodies; it is generally preferred to use rabbits or guinea pigs, but horses, cats, dogs, goats, pigs, rats, cows, sheep, etc., can also be used.
  • Antibody is recovered by collecting blood from the immunized animal after the passage of 1 to 2 weeks subsequently to the last administration, centrifuging the blood and isolating serum from the blood.
  • Monoclonal antibodies may e.g. be human or murine.
  • an antibody of the present invention can be introduced into the mammal by, e.g., intramuscular injection.
  • any form of antibody administration may be used.
  • Any conventional liquid or solid vehicle may be employed which is acceptable to the subject and does not have adverse side effects.
  • Phosphate-buffered saline (PBS) at a physiological pH, e.g. about pH 6.8 to 7.2, preferably about pH 7.0, may be used as a vehicle, alone or with a suitable adjuvant, such as an aluminium hydroxide-based adjuvant.
  • PBS Phosphate-buffered saline
  • 0167 The following examples are offered by way of illustration and not by way of limitation.
  • the human template chosen for the VH chain was a combination of DP88 (aa residues 1-95) and JH4b (aa residues 103-113) (See Figure 3B).
  • the human template chosen for the V L chain was a combination of L16 (VK subgroup III, aa residues 1-87) combined with JK4 (aa residues 98-107) (See Figure 3A).
  • the framework homology between the murine sequence and the human template was about 70% for VH and about 74% for V . 0170]
  • a Fab library was constructed by DNA synthesis and overlapping PCR as described above and depicted in Fig.2.
  • the library was composed of synthesized TES-C21 CDRs synthesized with the respective chosen human templates, DP88/JH4b and L16/JK4.
  • the complexity of the library was 4096 (- 2 12 ).
  • the overlapping nucleotides encoding partial VH and V sequences were synthesized in the range of about 63 to about 76 nucleotides with 18 to 21 nucleotide overlaps.
  • PCR amplification of VL and VH gene was performed using a biotinylated forward primer containing the specific sequence to the framework region FR1 and an overhanging sequence annealed to the end of leader sequence (Genelll) and a reverse primer from the conserved constant region (C/ or CH1 ) under standard PCR conditions.
  • the PCR product was purified by agarose gel electrophoresis, or by commercial PCR purification kit to remove unincorporated biotinylated primers and non-specific PCR.
  • the beads were sedimented and washed twice with 200 ⁇ L 2x B&W buffer.
  • the non-biotinylated ssDNA (minus strand) was eluted with 300 ⁇ L freshly prepared 0.15M NaOH at RT for 10 min with mild shaking. A second NaOH elution can increase the yield slightly (optional).
  • the eluant was centrifuged to remove any trace beads.
  • the ssDNA was precipitated from the supernatant by adding 1 ⁇ L glycogen (10mg/mL), 1/10 volume of 3M NaOAc (pH 5.2), and 2.5 volumes of EtOH. The precipitated ssDNA was then washed with 70% EtOH followed by lyophilizing for 3 min and dissolving in 20 ⁇ L ddH 2 O. The ssDNA was quantitated by spotting on an ethidium bromide (EtBr) agarose plate with DNA standards, or by measuring OD 260 .
  • EtBr ethidium bromide
  • Uridinylated templates were prepared by infecting CJ236 E. coli strain (duf ung " ) with M13-based phage (phage-expression vector TN003). 177] The following components [200 ng of uridinylated phage vector (8.49 kb); 92 ng phosphorylated single-stranded H chain (489 bases); 100 ng phosphorylated single-stranded L chain (525 bases); 1 ⁇ L 10X annealing buffer; adjust volume with ddH 2 O to 10 ⁇ l] were annealed (at about 8-fold molar ratio of insert to vector) by PCR holding the temperature at 85°C for 5 min (denaturation) and then ramping to 55°C over 1 hour.
  • Plating Phage Library 180 The phage library was diluted in LB media to achieve the desired number of plaques per plate. High titer phage was mixed with 200 ⁇ L XL-1 B cell culture. 3mL LB top agar was mixed, poured onto an LB plate, and allowed to sit at room temperature for 10 minutes. The plate was incubated overnight at 37°C. B.
  • Phage Elution 181 100 ⁇ L of phage elution buffer (10mM Tris-CI, pH 7.5, 10mM EDTA, 100mM NaCI) was added to each well of a sterile U-bottom 96 well plate. A single phage plaque from the overnight library plate was transferred with a filtered pipette tip to a well. The phage elution plate was incubated at 37°C for 1 hour. The plate may be stored at 4°C following incubation. C. Culture for Deep Well Plates
  • XL1 B cells from 50mL culture were added to 2xYT media at a 1 :100 dilution. The cells were grown at 37°C in a shaker until the A 6 oo was between 0.9 to 1.2.
  • IPTG 1 M IPTG (1 :2000) was added to the XL1 B culture. The final concentration of IPTG was 0.5mM. 750 ⁇ L of cell culture was transferred to each well of a 96 well - deep well plate (Fisher Scientific). Each well was inoculated with 25 ⁇ L of eluted phage. The deep well plate was placed in the shaker (250rpm) and incubated overnight at 37°C. D. Preparing Supernatant for ELISA Screening
  • IPTG was added at a final concentration of 0.5mM and 15mL of the culture was tranferred to a 50mL conical tube for each clone to be characterized.
  • the cells were pelleted in an IEC centrifuge at 4,500 rpm for 20 minutes. Culture medium was removed the pellet was resuspend in 650 ⁇ L of resuspension buffer (50mM Tris, pH 8.0 containing 1 mM EDTA and 500mM sucrose), vortexed, and placed on ice for 1 hour with gentle shaking. Cellular debris was removed by centrifugation at 9,000 rpm for 10 minutes at 4°C. The supernatant containing the soluble Fabs was collected and stored at 4°C.
  • resuspension buffer 50mM Tris, pH 8.0 containing 1 mM EDTA and 500mM sucrose
  • the framework residues that differed between the TES-C21 sequence and the human template were randomly substituted as described above and then assessed for their potential affect on target binding, and antibody folding. Potential framework residues that may have affected the binding were identified. In this case, they were residues 12, 27, 43, 48, 67, 69 in V H , and 1 , 3, 4, 49, 60, 85 in VL (Kabat number system). (See Figure 4) It was later demonstrated that only positions 27 and 69 significantly affected binding in the VH region (clone number 1136-2C).
  • the primary screen used was a single point ELISA (SPE) using culture media (See description below).
  • SPE single point ELISA
  • the primary screen selected clones that that bind to the antibody's target molecule. Clones that gave equal or better signal than the parent molecule were selected for the next round of screening.
  • the constructed library was subjected to an ELISA screen for improved binding to the recombinant human IgE, SE44.
  • Clones with binding affinity greater than murine TES-C21 Fab were isolated and sequenced. Clone ID #4, 49, 72, 76, and 136 were further characterized. ELISA titration curves for clone 4, 49, 72, 78, and 136 are shown in Figure 5A and 5B indicating that their affinity is similar to the parent, TES-C21. These clones compete with murine TES-C21 for binding to human IgE indicating that the binding epitope was not changed during the humanization process.
  • Humanized Fabs did not bind to Fc ⁇ RI-bound IgE suggesting that it is less likely that the humanized antibodies will crosslink the receptor to cause histamine release when they were constructed into divalent IgG.
  • Plates were coated with 2ug/mL sheep anti-human Fd in carbonate coating buffer overnight at 4°C. The coating solution was removedand the plates were blocked with 200uL/well 3% BSA/PBS for 1 hour at 37°C. After washing the plates 4x with PBS/0.1 % TWEEN ® (PBST), 50uL/well Fab sample (i.e., supernatant containing high titer phage and secreted Fab or periplasmic prep from DMB block, or 15mL prep) was added. Plates were incubated for 1 hour at room temperature followed by washing 4x with PBST.
  • PBST PBS/0.1 % TWEEN ®
  • Plates were coated with 0.25ug/mL (for purified Fab 0.1 ug/ml) SE44 in carbonate coating buffer overnight at 4°C. Coating solution was removed and the plates were blocked with 200uL/well 3 % BSA PBS for 1 hour at 37°C.
  • Buffer A (1 liter) 50mM NaH 2 PO 4 6.9 g NaH 2 PO 4 H 2 O (or 6 g NaH 2 PO 4 ) 300mM NaCI 17.54 g NaCI 10mM imidazole 0.68 g imidazole (MW 68.08) adjust pH to 8.0 using NaOH Lysis buffer: Mix 25 mL of Buffer A with one tablet of Complete Protease Inhibitor Cocktail (Roche, Basel, Switzerland). 0201] Resuspended cells were transferred into a 50mL conical tube and lysed with 100 ⁇ L 100 mg/mL lysozyme by inverting the tube several times until the mixture moves together as a blob (due to the lysis).
  • the column was washed with 20 mL buffer (50mM NaH 2 PO , 300mM NaCI, 15mM imidazole, adjust pH to 8.0 with NaOH) followed by a 20 mL wash with 50mM NaH 2 PO 4 , 300mM NaCI, 20mM imidazole.
  • Fabs were eluted with 6 x 500 ⁇ L elution buffer (50mM NaH 2 PO 4 , 300mM NaCI, 450mM imidazole, adjust pH to 8.0 with NaOH) and analyzed by SDS PAGE. Column fractions were stored at 4 °C.
  • Example 8 Soluble Receptor Assay 203 A 96 well assay plate suitable for ELISA was coated with 0.05 mL 0.5 ⁇ g/mL FceRI alpha-chain receptor coating buffer (50 mM carbonate/bicarbonate, pH 9.6) for 12 hours at 4-8°C. The wells were aspirated and 250 ⁇ L blocking buffer (PBS, 1% BSA, pH 7.2) was added and incubated for 1 hour at 37°C.
  • FceRI alpha-chain receptor coating buffer 50 mM carbonate/bicarbonate, pH 9.6
  • the samples and reference TES-C21 MAbs were titered from 200 to 0.001 ⁇ g/mL by 1 :4 dilutions with assay buffer (0.5% BSA and 0.05% Tween 20, PBS, pH 7.2) and an equal volume of 100ng/mL biotinylated IgE was added and the plate incubated for 2-3 hours at 25°C.
  • assay buffer (0.5% BSA and 0.05% Tween 20, PBS, pH 7.2
  • biotinylated IgE was added and the plate incubated for 2-3 hours at 25°C.
  • the FceRI - coated wells were washed three times with PBS and 0.05% TWEEN 20 and 50 ⁇ L from the sample wells were transferred and incubated with agitation for 30 minutes at 25°C.
  • Each candidate was assayed for binding affinity and positive clones were sequenced. Antibody variants having beneficial mutations in CDR regions that increase binding affinity were further characterized. Assays included Biacore analysis; inhibition of IgE binding to its receptor; and cross linking of receptor bound IgE.
  • High affinity MAbs candidates were generated.
  • the heavy and light chains variable regions were PCR amplified from phage vectors templates and subcloned separately into H- and L-chain expression vectors under the expression of a CMV promoter.
  • Six full antibody clones were constructed and are represented in Figure 10 A-F.
  • Appropriate heavy and light chain plasmids were co-transfected into the mouse myeloma cell line NS0 using electroporation by techniques well known in the art. See, e.g., Liou et al. J Immunol. 143(12):3967-75 (1989).
  • Antibodies were purified from the single stable cell line supernatants using protein A-sepharose (Pharmacia). The concentration of the antibody was determined using spectrophotometer at 280nm and FCA assay (IDEXX).
  • HRP horseradish peroxidase
  • peroxidase conjugation kit Zymed Labs, San Francisco, CA
  • the titer of each conjugated anti-IgE MAb was determined using ELISA with plates coated with a monoclonal human IgE (SE44).
  • SE44 monoclonal human IgE
  • 73 overlapping peptides were synthesized which encompassed amino acid residues 141 to 368 of human IgE, which included the entire CH3 domain. Each peptide consisted of 12 amino acid residues having a 3 amino acid overlap with the 3' end of the previous peptide.
  • SPOTs membranes were synthesized with fluorenylmethoxycarboyl (Fmoc) amino acids on cellulose membrane. The membranes were rinsed in methanol and then washed in TBS (pH 7.5) 3X for 10 min.
  • Lysates from this in-vitro transcription and translation coupled system (10 ul reaction mix) were subjected to SDS-PAGE (12 %) and then transferred to nitrocellulalose membranes.
  • the membranes were blocked with 5% dry milk in Tris-buffered saline (TBS) and subsequently stained with the primary antibody, anti-IgE MAbs.
  • Specific reactive bands were detected using a goat anti-human IgG Fc conjugated to horseradish peroxidase (Jackson Labs, Bar Harbor, Maine) and the immunoreactive bands were visualized by the SuperSignal Western blotting detection kit (Pierce).
  • Anti-V5 antibodies were used as a positive control that detected the V5 tag introduced at the C-terminus of these peptides.
  • Transgenic mice that constitutively express human IgE were used to demonstrate the active production of antibodies to a human immunogenic peptide of Epitope B.
  • Two fusion peptides, each comprising an immunogenic peptide of the invention, a cysteine residue and KLH, were chemically synthesized.
  • the sequence of peptide 1 was : (KLH-Cys) - Leu Pro Arg Ala Leu Met Arg Ser Thr and the sequence of peptide 2 was: Leu Pro Arg Ala Leu Met Arg Ser Thr - (Cys-KLH)
  • mice were injected subcutaneously with 20 ⁇ g of the immunogenic peptide in complete Freund's adjuvant (Difco Laboratories, Detroit, Ml) in 200 ⁇ L of PBS pH 7.4. At two-week intervals the mice were twice injected subcutaneously with 20 ⁇ g of the peptide immunogen in incomplete Freund's adjuvant. Then, two weeks later and three days prior to sacrifice, the mice were again injected intraperitoneally with 20 ⁇ g of the same immunogen in PBS. Serum was collected and tested for the presence of anti-IgE antibodies specific for Epitope B. As seen in Figure 16, the peptide elicited anti-IgE antibodies in these transgenic mice.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Pulmonology (AREA)
  • Genetics & Genomics (AREA)
  • Microbiology (AREA)
  • Dermatology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Rheumatology (AREA)
  • Otolaryngology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Saccharide Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
PCT/US2004/024360 2003-02-01 2004-07-29 IDENTIFICATION OF NOVEL IgE EPITOPES WO2005075504A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
KR1020067017609A KR20070008578A (ko) 2004-02-02 2004-07-29 신규한 IgE 에피토프의 확인
KR1020137018993A KR101562114B1 (ko) 2004-02-02 2004-07-29 신규한 IgE 에피토프의 확인
JP2006551035A JP2008507474A (ja) 2004-02-02 2004-07-29 新規のIgEエピトープの同定
AU2004315197A AU2004315197B2 (en) 2004-02-02 2004-07-29 Identification of novel IgE epitopes
CA002552999A CA2552999A1 (en) 2004-02-02 2004-07-29 Identification of novel ige epitopes
EP04779424A EP1718669A4 (en) 2004-02-02 2004-07-29 IDENTIFICATION OF NEW IGE EPITOPES
KR1020147000238A KR101581659B1 (ko) 2004-02-02 2004-07-29 신규한 IgE 에피토프의 확인
KR1020127009917A KR101365375B1 (ko) 2004-02-02 2004-07-29 신규한 IgE 에피토프의 확인
IL176940A IL176940A (en) 2003-02-01 2006-07-18 An isolated antibody or antigen-binding fragment thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
USPCT/US04/02892 2004-02-02
PCT/US2004/002894 WO2004070011A2 (en) 2003-02-01 2004-02-02 HIGH AFFINITY ANTI-HUMAN IgE ANTIBODIES
USPCT/US04/02894 2004-02-02
PCT/US2004/002892 WO2004070010A2 (en) 2003-02-01 2004-02-02 A method for generating high affinity antibodies

Publications (1)

Publication Number Publication Date
WO2005075504A1 true WO2005075504A1 (en) 2005-08-18

Family

ID=34841291

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/024360 WO2005075504A1 (en) 2003-02-01 2004-07-29 IDENTIFICATION OF NOVEL IgE EPITOPES

Country Status (8)

Country Link
EP (1) EP1718669A4 (ko)
JP (1) JP2008507474A (ko)
KR (4) KR101562114B1 (ko)
CN (1) CN102702359A (ko)
AU (2) AU2004315197B2 (ko)
CA (1) CA2552999A1 (ko)
SG (2) SG183683A1 (ko)
WO (1) WO2005075504A1 (ko)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008099188A1 (en) * 2007-02-15 2008-08-21 Astrazeneca Ab Binding members for ige molecules
WO2008099178A2 (en) * 2007-02-15 2008-08-21 Astrazeneca Ab Binding members for ige molecules
EP2056849A2 (en) * 2006-08-04 2009-05-13 The Trustees of the University of Pennsylvania Methods and compositions for treating ige-mediated diseases
JP2010505416A (ja) * 2006-10-02 2010-02-25 アムジェン インコーポレイテッド Il−17受容体a抗原結合タンパク質
WO2010067286A2 (en) 2008-12-09 2010-06-17 Pfizer Vaccines Llc IgE CH3 PEPTIDE VACCINE
JP2011507816A (ja) * 2007-12-21 2011-03-10 グラクソスミスクライン バイオロジカルズ ソシエテ アノニム マラリア用ワクチン
US8722053B2 (en) 2010-06-07 2014-05-13 Pfizer Vaccines Llc IgE CH3 peptide vaccine
JP2015121536A (ja) * 2007-05-31 2015-07-02 メディジーン アーゲー ワクチンとしての変異型パルボウイルス構造タンパク質
WO2019186369A1 (en) * 2018-03-26 2019-10-03 Novartis Ag Methods of treating chronic spontaneous urticaria using ligelizumab
WO2022264021A1 (en) 2021-06-14 2022-12-22 Novartis Ag PHARMACEUTICAL FORMULATION CONTAINING AN ANTI-IgE ANTIBODY

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3565587A4 (en) 2017-01-06 2020-12-16 The Regents of The University of California THERAPEUTIC ANTI-IGE ANTIBODIES AND RELATED METHODS AND COMPOSITIONS

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999067293A1 (en) * 1998-06-20 1999-12-29 United Biomedical Inc. Peptide composition as immunogen for the treatment of allergy
US6180370B1 (en) * 1988-12-28 2001-01-30 Protein Design Labs, Inc. Humanized immunoglobulins and methods of making the same

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS63225397A (ja) * 1986-10-03 1988-09-20 Dainippon Pharmaceut Co Ltd 抗アレルギー作用を有する新規ペプチド
DE69333484T2 (de) * 1992-09-24 2005-03-24 Novartis Ag Umgestaltete monoklonale Antikörper gegen ein Immunglobulinisotyp
US6172213B1 (en) * 1997-07-02 2001-01-09 Genentech, Inc. Anti-IgE antibodies and method of improving polypeptides
US7393529B2 (en) * 1998-04-09 2008-07-01 Idexx Laboratories, Inc. Methods and compositions for inhibiting binding of IgE to a high affinity receptor
EP0957111B1 (en) * 1998-04-09 2004-12-29 Idexx Laboratories, Inc. Specific binding proteins for treating canine allergy
GB9822763D0 (en) * 1998-10-20 1998-12-16 Univ Sheffield Immunoglobin variant
MXPA01008612A (es) * 1999-02-25 2003-06-24 Smithkline Beecham Biolog Epitopos o mimotopos derivados de los dominos c-epsilon-3 o c-epsilon-4 de ige, antagonistas de los mismos y sus usos terapeuticos.
GB9913327D0 (en) * 1999-06-08 1999-08-11 Smithkline Beecham Biolog Vaccine
GB0020717D0 (en) * 2000-08-22 2000-10-11 Smithkline Beecham Biolog Novel compounds and process
US6887472B2 (en) * 2000-08-30 2005-05-03 Pfizer Inc. Anti-IgE vaccines
GB0026334D0 (en) * 2000-10-27 2000-12-13 Smithkline Beecham Biolog Vaccine
JP2003047482A (ja) * 2001-05-22 2003-02-18 Pfizer Prod Inc 非アナフィラキシー誘発性IgEワクチン
GB0209878D0 (en) * 2002-04-30 2002-06-05 Glaxosmithkline Biolog Sa Vaccine
EP1592777A4 (en) * 2003-02-01 2008-06-04 Tanox Inc METHOD OF GENERATING HIGH-ANTIBODY ANTIBODIES

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6180370B1 (en) * 1988-12-28 2001-01-30 Protein Design Labs, Inc. Humanized immunoglobulins and methods of making the same
WO1999067293A1 (en) * 1998-06-20 1999-12-29 United Biomedical Inc. Peptide composition as immunogen for the treatment of allergy

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP1718669A4 *

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2056849A2 (en) * 2006-08-04 2009-05-13 The Trustees of the University of Pennsylvania Methods and compositions for treating ige-mediated diseases
EP2056849A4 (en) * 2006-08-04 2010-09-08 Univ Pennsylvania METHODS AND COMPOSITIONS FOR TREATING IGE MEDIATED DISEASES
JP2010505416A (ja) * 2006-10-02 2010-02-25 アムジェン インコーポレイテッド Il−17受容体a抗原結合タンパク質
US8389704B2 (en) 2007-02-15 2013-03-05 Medimmune Limited Binding members for IgE molecules
EP2620452A1 (en) * 2007-02-15 2013-07-31 Medimmune Limited Binding members for IGE molecules
WO2008099178A2 (en) * 2007-02-15 2008-08-21 Astrazeneca Ab Binding members for ige molecules
US7959917B2 (en) 2007-02-15 2011-06-14 Medimmune Limited Binding members for IgE molecules
AU2008216008B2 (en) * 2007-02-15 2012-07-05 Astrazeneca Ab Binding members for IgE molecules
AU2008215926B2 (en) * 2007-02-15 2012-07-19 Astrazeneca Ab Binding members for IgE molecules
WO2008099188A1 (en) * 2007-02-15 2008-08-21 Astrazeneca Ab Binding members for ige molecules
EP2592094A1 (en) * 2007-02-15 2013-05-15 AstraZeneca AB Binding members for IgE molecules
WO2008099178A3 (en) * 2007-02-15 2008-10-16 Astrazeneca Ab Binding members for ige molecules
JP2018109636A (ja) * 2007-05-31 2018-07-12 メディジーン アーゲー ワクチンとしての変異型パルボウイルス構造タンパク質
JP2015121536A (ja) * 2007-05-31 2015-07-02 メディジーン アーゲー ワクチンとしての変異型パルボウイルス構造タンパク質
JP2011507816A (ja) * 2007-12-21 2011-03-10 グラクソスミスクライン バイオロジカルズ ソシエテ アノニム マラリア用ワクチン
US8475801B2 (en) 2008-12-09 2013-07-02 Pfizer Vaccines, LCC IgE CH3 peptide vaccine
EP2865389A1 (en) 2008-12-09 2015-04-29 Pfizer Vaccines LLC IgE CH3 peptide vaccine
US8298547B2 (en) 2008-12-09 2012-10-30 Pfizer Vaccines, LLC IgE CH3 peptide vaccine
US9216229B2 (en) 2008-12-09 2015-12-22 Pfizer Vaccines Llc IgE CH3 peptide vaccine
WO2010067286A2 (en) 2008-12-09 2010-06-17 Pfizer Vaccines Llc IgE CH3 PEPTIDE VACCINE
US8722053B2 (en) 2010-06-07 2014-05-13 Pfizer Vaccines Llc IgE CH3 peptide vaccine
US9249233B2 (en) 2010-06-07 2016-02-02 Pfizer Vaccines Llc IgE CH3 peptide vaccine
WO2019186369A1 (en) * 2018-03-26 2019-10-03 Novartis Ag Methods of treating chronic spontaneous urticaria using ligelizumab
WO2022264021A1 (en) 2021-06-14 2022-12-22 Novartis Ag PHARMACEUTICAL FORMULATION CONTAINING AN ANTI-IgE ANTIBODY

Also Published As

Publication number Publication date
CN102702359A (zh) 2012-10-03
KR101365375B1 (ko) 2014-02-19
KR20120056297A (ko) 2012-06-01
EP1718669A1 (en) 2006-11-08
EP1718669A4 (en) 2007-10-10
AU2004315197B2 (en) 2009-06-04
JP2008507474A (ja) 2008-03-13
KR20130088198A (ko) 2013-08-07
SG149892A1 (en) 2009-02-27
AU2004315197A1 (en) 2005-08-18
SG183683A1 (en) 2012-09-27
CA2552999A1 (en) 2005-08-18
KR101562114B1 (ko) 2015-11-20
AU2009202408A1 (en) 2009-07-09
KR101581659B1 (ko) 2015-12-31
KR20140021058A (ko) 2014-02-19
KR20070008578A (ko) 2007-01-17

Similar Documents

Publication Publication Date Title
US9156908B2 (en) High affinity, anti-human IgE antibodies
AU2009202408A1 (en) Identification of novel IgE epitopes
US20110200604A1 (en) IDENTIFICATION OF NOVEL IgE EPITOPES
AU2012261739A1 (en) Identification of novel IgE epitopes
AU2013263817A1 (en) High affinity anti-human IgE antibodies
MXPA06008690A (es) Identificacion de epitopos ige novedosos

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG 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 NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY 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: A1

Designated state(s): GM KE LS MW MZ NA 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 HU IE IT LU MC NL PL PT RO SE SI 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
WWE Wipo information: entry into national phase

Ref document number: 2552999

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 176940

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2006551035

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2004315197

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: PA/a/2006/008690

Country of ref document: MX

Ref document number: 200480041292.8

Country of ref document: CN

NENP Non-entry into the national phase

Ref country code: DE

WWW Wipo information: withdrawn in national office

Country of ref document: DE

ENP Entry into the national phase

Ref document number: 2004315197

Country of ref document: AU

Date of ref document: 20040729

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2004315197

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2004779424

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 1020067017609

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2004779424

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1020067017609

Country of ref document: KR