SG183683A1 - Identification of novel ige epitopes - Google Patents

Identification of novel ige epitopes Download PDF

Info

Publication number
SG183683A1
SG183683A1 SG2012057212A SG2012057212A SG183683A1 SG 183683 A1 SG183683 A1 SG 183683A1 SG 2012057212 A SG2012057212 A SG 2012057212A SG 2012057212 A SG2012057212 A SG 2012057212A SG 183683 A1 SG183683 A1 SG 183683A1
Authority
SG
Singapore
Prior art keywords
ser
antibody
thr
gly
leu
Prior art date
Application number
SG2012057212A
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
Publication of SG183683A1 publication Critical patent/SG183683A1/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]

Abstract

The present invention relates to novel peptide epitopes derived from the CH3 domain of IgE which are recognized by high affinity antibodies that specifically bind IgE. These novel peptides may be used for both active immunization of a subject by administering these peptides to generate high affinity antibodies in a subject, as well as for generating high affinity anti-IgE antibodies in non-human hosts that specifically bind to these regions of IgE for passive immunization of a subject. (No suitable figure)

Description

Identification of Novel IgE Epitopes
CROSS RELATED APPLICATIONS . 10001] The present application claims priority to PCT Application No. PCT/US04/02882 and PCT/US04/02894 filed Feb. 2, 2004, all of which are incorporated by reference herein.
BACKGROUND OF THE INVENTION
[0002] 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, 7 is mediaied via B cells and is based on antigen-antibody reactions. Delayed ” hypersensitivity is mediated via T cells and based on mechanisms of celiular immunity. In recent years, the term "allergy" has become more and more. synonymous with type I hypersensitivity. 10003) Immediate hypersensitivity is a response based on the production of antibodies of the immunoglobulin class E (igk 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 aliergen's entry into the body, i.e. at mucosal surfaces and/or at local lymph nodes. Locally produced IgE wilt first sensitize local mast cells, Le. Igk antibodies bind with their constant regions to Foe receptors on the surface of the mast cells, and then “spill-over" [gE enters the circuiation and binds to receptors on both circulating basophils and tissue- fixed mast cells throughout the body. When the bound IgE is subsequently
Fa contacted with the allergen, the Foe recepiors are crosslinked by binding of the
Se allergen causing the cells fc degranulate and release a number of anaphyiactic mediaiors such as histamine, prostagianding, leukotrienes, etc. it is the release of these substances which is responsibie 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. In addition, the reaction upon second contact with the allergen is intensified because some B cells form a "memory pool" of surface IgE positive B cells (sigE™ B cells) after the first contact with the allergen by expressing IgE on the cell surface.
[0004] There are two major receptors for IgE, the high affinity receptor FceR! and the low- affinity receptor FoeRIl, FoeRlis predominantly expressed on the surface of mast cells and basophils, bul low levels of FoeRl can also be found on human
Langerhan’s cells, dendritic cells, and monocytes, where it functions in IgE- mediated allergen presentation. In addition, FceR! has been reporied on human eosinophils and plaieslsts (Hasegawa, 8. et. al., Hematopoiesis, 1998, 83:2543- 2551). FoeRl is not found on the surface of B cells, T cells, or neutrophils, The expression of FeeRl on Langerhan's cells and dermal dendritic cells is functionally £ and biologically important for IgE-bound antigen presentation in allergic individuals oo {Klubal R. st al, J. Invest. Dermatol, 1897, 108 (3):336-42),
[60053] The low-affinity receptor, FceRIl (CD23) is a lectin-like molecuie comprising three identical subunits with head structures extending from a long o-helical coiled stalk : from the cellular plasma membrane (Dierks, AE, ef al, J. Immunol. 1893, 150:2372-2382). Upon binding to IgE, FeeRll associates with CD21 on B cells involved in the regulation of synthesis of IgE (Sanon, A. et al, J. Allergy Clin.
Immunol. 1990, 886:333-344, Bonnefoy, J. ef al, Eur, Resp. J. 1898, £:83s-66s).
FceRI has long been recognized for allergen presentation {Sutton and Gould 1983, Nature, 366:421-428). IgE bound fo FceRIl on epithelial cells is responsible for specific and rapid allergen presentation (Yang, P.P., J. Clin. invest., 2000, 106:878-886). FceRlIl is present on several cell types, including B- cells, eosinophils, platelets, natural Killer cells, T-cells, follicular dendritic cells, and
Oo Langerhan's cells. or [0006] The structural entities on the IgE molecule that interact with FceR! and FoeRl} have also been identified. Mutagenesis studies have indicated that the CH3 domain mediates IgE interaction with both FoeR! (Presta af al, J. Biol. Cham. 1084, 26©:28388-26373; Henry A. et al, Biochemistry, 1997, 36:155688-15578) and FceRl {Sutlon and Gould, Nature, 1893, 366; 421-428: Shi, J. ef al,
Biochemistry, 1887, 36:2112-2122}. The binding sites for both high- and low- affinity receptors are located symmetrically along a central rotational axis through the two CH3 domains. The FeeRl-binding site is located in a CH3 domain on the z outward side near the junction of the CH2 dornain, whereas the FeeRIl-binding site is on the carboxy!-terminus of CHB.
[0007] 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 tha induction of allergy and provides for the maintenance of the allergic state. As the response of other antibody classes is not affected, bath 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 FceRl receptors ver basophil (Malveaux ef al., J. Clin. Invest, 1878, 62:176). They noted that the FceR!i densities in allergic and non-allergic ) parsons range from 10” to 10° receptors per basophil. Later it was shown that treatment of allergic diseases with anti-IgE decreased the amount of circulating tok to 1% of pretreatment levels (MacGlashan ef af, J. Immunol. 1987, 158:1438-1448), MacGlashan analyzed serum obtained from patients treated with whale anti-igE antibody, which binds free IgE circulating in the serum of the patient. They reported that lowering the level of circulating IgE in a patient resultedin a ower number of receptors present on the surface of basophils.
Thus, they hypothesized that FceRI densify on the surface of basophils and mast cells is directly or indirectly regulated by the level of circulating IgE antibody.
[0608] More recently, WO 90/62550 disclosed the use of IgE molecules and fragments, which bind to FeeRl and FeeRlIl IgE binding sites to block IgE binding to receptors. { | However, effective therapies that lack deleterious side effects for the management i of these allergic diseasas are limited. One therapeutic approach to treating allergic diseases involved using humanized anti-igE antibody to treat allergic rhinitis and asthma (Cone, J. ef al., J. Clin. Invest.1997, 99:879-887: Racine- oo
Poon, A. ef al, Clin. Pharmeol. Ther. 1897, 62:675-690; Fahy, JV. et al, Am. J.
Resp. Crit. Care Med. 1997, 155:1824-18234; Boulet, L.. P. ef al., Am. J. Resp. Crit.
Care Med., 1987, 155:1835-1 840; Miigrom, E. et af., N. Engl. J. Med, 1989, 341:1966-1973). These clinical data demonstrate that inhibition of IE binding to its receptors is an effective approach to fresting allergic diseases.
[0009] Antibodies suitabie as anti-allergic agents should react with surface Igk positive B cells which differentiate Into IgE producing plasma cells, so that they can be used to functionally eliminate those B cells. However, antibodies to IgE in principle may also induce mediator release from IgE sensitized mast cells by crosslinking the
Fee racapiors, thus antagonizing the beneficial effect exerted on the serum lok and slgE” B cell level. One of the potentially dangerous problems with developing anti-IgE therapies is the possiblity of igE-crosslinking caused by the therapetic : antibody binding to IgE already bound to the high affinity receptor and triggering nistamine release resuliing in a potentially anaphylactic reaction.
[0610] Therefore, antibodies applicable for therapy of allergy must not be capable of r ) reacting with IgE bound on sensitized mast cells and basophils, but should retain the capability to recognize sige” B calls. Such IgE isctype-specific antibodies have been described e.g. by Chang et al. {Biotechnology 8, 122-126 (1990)}, in
European Patent No. EP0407382, and several U.S. Patents, e.g., U.S. Patent No. 5,448,760.
[0011] Peptides used to generate anti-igE antibodies also suffer from the dangerous notential to induce anaphylactic antibodies, Generation of anti-igE antibodies during active vaccination may be capable of triggering histamine reisase inthe same way passively administered anti-IgE antibodies, if the antibodies generated during immunization bind fo IgE bound to the high affinity IgE receptor or by other. mechanisms, 10012] Thus, there is a need for higher affinity non-anaphylactic antibodies that bind specifically to IgE but do not bind IgE already bound to its high affinity receptor, as ( well as peptides for active immunization that do not induce anaphylactic antibodies. The inventors have identified the specific epitope of igE that provides high affinity binding of antibodies without binding to IgE on mast cells or basophils. : These specific epitopes can in turn be used to gnerate specific peptides for active immunization to generate antibodies to IgE that only bind to the region of IgE that binds to the receptor, ensuring that the antibodies do not crosslink IgE already bound to the receptor and thus are non-anaphylactic.
Ca
: SUMMARY OF THE INVENTION
[6013] The present invention relates to novel peptide epitopes derived from the CH3 : domain of IgE, These peptide epitopes ars 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 peplide epitopes may also be used in generating high affinity anti-IgE antibodies in a non-human host that specifically hind to these regions of IgE and use the resulting antibodies for the passive immunization of a2 of a mammal.
[0014] One immunogen (epitope A, Fig. 11) of the present invention comprises the amino ( acid sequence: :
Asn Pro Arg Gly Val Ser Xaa Tyr Xaa Xaa Arg Xaa (SEQ ID NC. 72).
One example of epitope A is:
Asn Pro Arg Gly Val Ser Ala Tyr Leu Ser Arg Pro (SEQ 1D NC. 73)
Another immunogen {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 includes:
Leu Pro Arg Ala Leu Met Arg Ser Thr (SEQ ID NO. 75)
His Pro His Leu Pro Arg Ala Leu Met Arg Ser Thr (SEQ ID NO 76)
Leu Pro Arg Ala Leu Met Arg Ser Thr Thr Lys Thr (SEQ ID NO 77).
In either SEQ ID NO: 72 or SEQ ID NO: 74, Xsa may be any amino acid.
[0015] 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 diphtharia toxoid. The present invention also relates to polynucleotides encoding SEQ ID NOS. 72-77, vectors comprising said . polynucleotides, and cells harboring said vectors, 10016) The present invention also refates 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 io epitope A and/or epitope B.
[6017] 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.
[0618] The present invention relates to the administration of high affinity antibodies generaied using peptides comprising SEQ 1D NO: 72 and/or SEQ ID NO 74 toa 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, Such IgE mediated diseases or conditions include, e.g., asthma, atopic dermatiis, urticaria, allergic rhinitis and eczema.
BRIEF DESCRIPTION OF THE FIGURES
[06019] Figure 1 is a schematic representation of the phage vector used in antibody cloning and screening. 10020] Figure 2 is a schematic representation of ofigonuciectides useful in generating
Co antibody variants.
[0021] Figure 3A depicts the comparison of the light chains of the murine anti-lg= antibody TES-C21 and the combined human template of L16 and JK4.
[0022] Figure 3B depicts the comparison of the heavy chains of TES-C21 and the combinad human template DP88 and JH4b. ©0023] Figure 4 presents a table of the framework residue variants having high affinity as - compared io the parent TES-C21. 10624] Figure 5A and B depict the ELISA titration curves for clones 4, 49, 72, 78, and 138, as compared to the parent Fab of TES-C21 and negative contra! (5012).
[0025] Figure 6 depicts an inhibition assay of clones 2C, 5A, and 51, as compared to the parent TES-C21 and a negative control antibody. [60261 Figure 7A depicts the sequences of clones having a combination of beneficial mutations which resulted in even greater affinity for IgE. } i [0027] Figure 8A & 8B depict the framework sequences of the entire fight chain variable region for clones 138, 1, 2, 4, 8, 13, 15, 21, 30, 31, 35, 43, 44, 33, 81, 80, and 113.
[0028] Figure 9A & 8B depict the framework sequences of the entire heavy chain variabie region for 35 clones, 10029] Figures 10 A-F depict the complete heavy and light chain sequences for clones 138, 2C, 51, BA, 2B, and 1136-2C,
[0030] Figure11 depicts the CH3 region amino acid sequence of human IgE and highlights Epitope "A" and Epitope "B”.
[0031] Figure 12 depicts the overlapping peptides used fo identify Epitope B.
: [0032] Figure 13 depicts the identification of important residues in the binding region of
Epitope A.
[033] Figure 14 depicts the identification of important residues in the binding region of
Epitope B.
[0034] Figure 15 depicts a western blot analysis of MAD binding to mutant peptides.
[6635] Figure 16 depicts the generation of anti-IgE antibodies in a transgenic animal expressing human IgE. :
DETAILED DESCRIPTION OF THE INVENTION Definitions
[0036] Terms used throughout! this application are io be construed with ordinary and
Oy typical meaning to those of ordinary skill in the art. However, Applicants desire oo that the following terms be given the particular definition as defined below. 10037] The phrase "substantially identical” with respect to an antibody chain polypeptide sequence may be construed as an antibody chain exhibiting at least 70%, or 80%, ar 90% or 85% sequence identity to the reference polypeptide sequence. The term with respect to a nucleic acid seguence may be construed as a sequence of nucieofides exhibiting at least about 85%, or 90%, or 85% or 87% sequence identity to the reference nucieic acid sequence.
[0038] The term “identity” or “homology” 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 io achieve the maximum percent identity for the entire sequence, and not considering any conservative substitutions as part of 0 the sequence identity. Neither N- or C-terminal extensions nor inserlions shall be construed as reducing identity or homology. Methods and computer programs for the alignment are well known in the art. Seguence identity may be measured using sequence analysis software, 10039] The term "antibody" is used in the broadest sense, and specifically covers menocional antibodies {including full length monoclonal antibodies), polyclonal antibodies, and multispecific antibodies (e.g., bispecific antibodies). Antibodies (Abs) and immunoglobulins {igs} are givcoproteins having the same structural characteristics, While anfibodies exhibit binding specificity to a specific target, immunoglobulins inctude both antibodies and other antibody-like molecules which lack target specificity, Native antibodies and immunoglobulins are usually heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light (L) chains and two identical heavy {M) chains. Each heavy chain has at one end a variable domain {Vy} followed by a number of constant domains.
Each light chain has a variable domain at one end (VV) and a constant domain at its other end. “High affinity” antibodies refers to those antibodies having a binding affinity of at least 107°, preferably 10712, 0040) As used herein, “anti-human igE antibody” means an antibody which binds fo human IgE in such a manner 50 as fo inhibit or substantially reduce the binding of such {gE to the high affinity receptor, FeeRl, { [0041] The term "variable" in the context of variable domain of antibodies, 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 fight chain and the heavy chain variable domains. The more * highly conserved portions of variable domains are called the framework (FR). The : variable domains of native heavy and light chains each comprise four FR regions, largely a adopting a f-shest configuration, connected by three CDRs, which form loops connecting, and in some cases forming part of, the g-shest structure. The
CDRs in sach chain are heid together in close proximity by the FR regions and, with the CDRs from the othar chain, contribute to the formation of the target ; binding site of antibodies (see Kabat et al.) As used herain, numbering of immunoglobufin 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. 1887}, uniess otherwise indicated. | :
[0042] - The term "antibody fragment” refers io a portion of a fullength antibody, generally the target binding or variable region. Examples of antibody fragments include Fab,
Fab’, F(ab’); and Fv fragments. The phrase "functional fragment or analog” of an antibody is a compound having qualitative biological activity in common with a fuli- length antibody. For example, a functional fragment or anaiog of an anti-igE antibody is one which can bind to an igk 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, FeeRl. As used herein, “functional fragment” with respect to antibodies, refers to Fv, F(ab) and F{ab"); fragments. An "By" fragment is the minimum antibody fragment which sontains a complete target recognition and binding site. This region consists of a dimer of one heavy and ons light chain variable domain in a tight, non-covalent association (Vy Vy dimer). It is in this configuration that the three CDRs of each variable domain interact to define an target binding site on the swface of the Vy -Vy dimer. Collectively, the six
CDRs confer target binding specificity io 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 ata lower affinity than the enfire binding site, "Single-chain Fv" ar "sFv" antibody fragments comprise the
Vp and Vi domains of an antibody, wherein these domains are present in a single polypeptide chain. Generally, the Fv polypeptide further comprises a polypeptide © linker between the Vy and V| domains which enables the sFv to form the desired structure for target binding.
[0043] 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 carboxy! tetminus 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"); pepsin digestion product, Additional chemical
LL ) couplings of antibody fragments are known to thase of ordinary skill in the art. : : [0044] 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 ccouriing mutations that may be present in minor amounts. Monoclonal aniibodies 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 monaclonal antibody is directed against a single determinant on the target. in addition to their specificity, monoclonal antibodies are advantageous in that they oo 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. For exampie, the monocicnal 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 Kohter and Milstein, Nature 258, 495 (1875), or may be made by recombinant methods. : : ( [0045] "Humanized" forms of non-human {e.g. murine) antibodies are chimeric immunogiobulins, immunoglobulin chains or fragments thereof {such as Fv, Fab,
Fab’, F(ab"), or other targei-binding subsequences of antibodies) which contain : minimal sequence derived from non-human immunoglobulin, In genaral, the humanized antibody will comprise substantially all of at least ong, and typically twa, 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 2 human immunoglobulin consensus sequence. The : humanized antibody may also comprise at isast a portion of an immunoglcbulin constant region (Fg), typically that of a human immunoglobulin template chosen. 10046) 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
C ; 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.
[0047] "Transformation" of & cellular organism with DNA means introducing DNA into an organism so that the DNA is replicable, either as an exirachromosomal element or by chromosomal integration. “Transfection” of a cellular organism with DNA refers “ to the faking up of DNA, e.g., an expression vector, by the cell or organism whether ar not any coding sequences are in fact expressed. The terms : "transfected host call" and "transformed" refer to a cell in which DNA was iniroduced. The cell is fermed "host cell" and it may be either prokaryetic or eukaryotic. Typical prokaryotic host cells include various strains of E. coll. Typical eukaryotic host celis are mammalian, such as Chinese hamster ovary or cells of human origin, The iniroduced 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, oo [0048] The term "vector” means & DNA construct containing a DNA sequence which is operably linked fo a suitable control sequence capable of effecting the expression of the DNA in a suitable host. Such control sequences include 5 promoter io effect transcription, an optional operator sequence to control such transcription, a
Bh saguence encoding suitable mRNA ribosome binding sites, and sequences which control the termination of transcription and translation. The vector maybe 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. In the present specification, "plasmid" and "vector" are sometimes used interchangaably, 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 sguivalent function as and which are, or become, Known in the art, 10049] The expression "control sequences” refers io 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, for example, include a promoter, optionally an operator sequence, and a ribosome binding site,
Eukaryotic cells are known to ulilize promoters, potyadenviation signals, and enhancers. DNA for a presequence or secrefory leader may be operably linked to 7 } DNA for a polypeptide if it is expressed as a preprotein that participates in ths secretion of the polypeptide; a promoter or enhancer is operably linked to & coding sequence if it affects the transcription of the sequence; or a ribesome 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. Generally, "operably linked" means that the DNA sequences being linked are contiguous, and, In the case of a secretory leader, contiguous and in reading phase. However, enhancers do not have io be contiguous,
10030] "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.
[0051] The term "epitope tagged" when used herein in the context of a polypeptide refers to a polypeptide fused fo 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 ths fiu HA . tag polypeptide and iis antibody 12CAS (Field et al, Mo! Cell, Biol, 8: 2158-2165 {1988))); the c-myc tag and the 8F9, 3C7, 8E10, G4, B7 and 9510 antibodies thereagainst (Evan et al, Mal Cell. Biol. 5(12): 3810-3616 (1985)); and the Herpes
Simplex virus glycoprotein D (gD) tag and its antibody (Paborsky et al., Protein
Engineering 3(8}: 547-553 (1880). in certain embodiments, the epitope tag may be an epitope of the Fe region of an IgG molecule (2.g., 1gG1, 1932, 1gG3 or IgG4) that is responsible for increasing the in vive serum half-iife of the IgG molecule.
[0052] The word "label" when used herein refers to a detectable compound or composition which can be conjugated directly or indirectly to a molecuie 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 iabel, may catalyze chemical alteration of a substrate compound ar composition which is detectable.
Lo [0053] As used herein, "solid phase” means a non-aqueous matrix to which the antibody ~ of the present inverition can adhere. Example of solid phases encompassed herein Inchide those formed partially or entirety of glass (e.g. controlied pore glass), polysaccharides (e.g., agarose), polyacrylamides, polystyrene, nolyvinyi alcohol and silicones. In certain embodiments, depending on the context, the solid phase can comprise the well of an assay plate; in others ii is a purification column (e.g. an affinity chromatography column).
[6054] As used herein, the term "lgE-mediated disorder” means a condition or disease which is characterized by the overproduction and/or hypersensitivity fo the immunoglobulin 1gE=. Specifically if would be construed fo include conditions associated with anaphylactic hypersensitivity and atopic allergies, including for example: asthma, aliergic rhinitis & conjunctivitis (hay fever), eczema, urticaria, stopic dermatitis, and food aliergies. The serious physiological condition of anaphylactic shock caused by, e.g., bee stings, shake bites, food or medication, is also encompassed under the scope of this term.
Generation of Antibodies [0055) The starting or “parent” antibody may be prepared using technigues 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 4 ; making or seleciing a parent antibody and in no way limi the methods by which such a molecule may be generated. 0056] The antibody's binding affinity is determined prior to generating a high affinity antibody of the present invention. Also, the antibody may be subjected to other biological activity assays, £.g., in order te evaluate effectivensss as a therapeutic,
Such assays are known in the art and depend on the target and intended use for the antibody. :
[0057] To screen for antibodies which bind fo a particular epitope (&.g., those which block binding of IgE to its high affinity receptor), 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. Alternatively, epitope mapping can be performed io determine where the antibody binds an epitope of interest. Optionally, the binding affinity of the antibody for 2 homolog of { pg the target ussd to generate the antibody (where the homolog is from a differant - | species) may be assessed using techniques known in the ari. In one embodiment, the other species is a nonhuman mammal to which the antibody will be administered in preclinical studies. Accordingly, the species may be a nonhuman primate, such as rhesus, cynomolgus, baboon, chimpanzee and macaque. In other embodiments, the species may be a rodent, cat or dog, for example.
[0058] The parent antibody is altered according to tha present invention so ag fo generate an antibody which has a higher or stronger binding affinity for {he target than the parent antibody. Antibody specificity results from the unigue interface that is formed between the antibody and its target; the surfaces complement sach other to produce a unique fit (Jones, S. & Thomiton, J. M. (1886) Proc. Natl. Acad.
Sci. USA 83; 13-20). By further improving the contacts aiong this interface, the overall affinity can increase as a result of the lower energy cost needed fo favor the association of the binding partners.
[0659] The binding surface of the antibody is generally composed of six complemetarity determining regions (CDRs) which are loops that extend out from the core. The
CDRs are composed of amino acids having a sequence that is unique for binding to the spacific target. To increase the affinity of an antibody for its antigen, the environment around these amino acids must become more favorabie by introducing or improving various noncovalent forces, which ultimately lowers the { E enargetics of the interaction, resulting in higher affinity. 10060] Van dar Waals forces are noncovalent interactions which occur betwesn two electrically neutral motecules (Voet, D. & Voet, J. G. (1880) Biochemistry John + Wiley and Sons, NY, NY), Associations can occur between two surfaces from electrostatic interactions that arise from permanant 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 associafion will. result,
[0061] introducing hydrogen bonds will also increase the specificity of an interaction between an antibody and ifs antigen. Common donors and acceptors involved in hydrogen bonding are nitrogen, oxygen and sulfer atoms, of which amino acids are predominantly composed {See Vost, et al., supra). Hydrogen bonds tend cross only short distances {usually 2.7 to 3.1 A hence the binding partners must ( : come within close proximity for these interactions to oceur. Thus, one manner by — which affinity can be improved is fo bring potential donar and acceptor molecules into close contact to establish hydrogen bonds.
[0062] + Finally, improving the hydrophobic interactions will also increase the favorable energetics between two binding partners. The nonpolar residues that lie close to the binding surface should be surrounded by other nonpolar residues, and thus will exist in a favorable environment. By satisfying the burial of nonpolar side chains, the energetics of the interaction are favorable for a strong binding interface.
10063] Interactions that stabilize the protein-protein interface lower the energetic cost of maintaining these contacts and thus will increase the overall affinity. By improving the environment around individual amina acids that are near the binding interface, 2 more favorable environment is produced resulting in higher binding affinity.
Therefore, by introducing favorable contacts and improving the interface through further complementation, the overall binding interaction between antibady and antigen will be greatly improved.
[0064] The resulting high affinity antibody preferably has a binding affinity for the target which is at jeast about 10 fold higher, or at least about 20 fold higher, or at isast = about 500 fold higher or may be 1000 to 5000 foid higher, than the binding affinity { of the parent antibody for the target. The degres of enhancement in binding affinity necessary or desired will depend on the initial binding affinity of the parent antibody. 10065] in general, the method for making high affinity antibodies from a parent antibody involves the following steps:
[0066] 1. 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 fechnigues, phags-display techniques, or any other method of generating a target specific antibody.
[0067] 2. Selecting a framework sequence which is close in sequence {0 the parent framework, preferably a human template seguence. This template may be chosen based on, &.g., its comparative overall length, the size of the CDRs, the
Lo amine acid residues located at the junction between ihe framework and the CDRs, { oo overall homology, etc. The template chosen can be a mixture of more than one - sequence of may be a consensus template.
[0068] 3. Generating a library of clones by making random amino acid substitutions at each and every possible CDR position. One may also randomiy 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, generaling a iinrary 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, Cne method for generating the library of variants by oligonucleotide synthesis. : 10069] 4, Constructing an expression vector comprising the heavy and/or light chain variants generated in step (3) which may comprise the formulas: FRH1-CDRH1-
FRH2-CORH2-FRH3-CORH3-FRH4(l} and FRL1-CDRL1-FRL2-CDRL2-FRL3-
CORL3-FRL4 (Il), wherein FRL1, FRL2, FRL3, FRL4, FRH1, FRH2, FRH3 and
FRH4 represent the variants of the framework template light chain and heavy chain segusnces chosen in siep 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. i [0070 5. Screening the library of clones against the specific target and those clones “that bind the target ara screened for improved binding affinity. Those clones that bind with greater affinity than the parent molecule may be selected. The optimal high affinity candidate will have the greatest binding affinity possible compared to the parent antibody, preferably greater then 20 fold, 100 fold, 1000 fold or 5000 fold. If the chosen variant contains certain amino acids that are undesirable, such &s a glycosylation site that has been introduced or a potentially immunogenic site, : these amino acids may be replaced with more beneficial amino acid residues and the binding affinity reassessed.
[0071] One may also use this method to generate high affinity antibodies from & fully human parent antibody by randomly substituting only the CDR regions, isaving the human framework intact.
[0072] Due to improved high throughput screening techniguas and vectors such as the { one depicted in Figure 1, an artisan can rapidly and efficiently screen a ~ comprehensive library of substitutions at all sites in a given CDR and/or framework region. By randomly substituting all amino acids at all positions simultaneously, one is able to screen possible combinations that significantly increase affinity that would not have been anticipated or identified by individual substitution due to, e.g., synergy.
PARENT ANTIBODY PREPARATION
Target Preparation
[0073] Soluble targets or fragments thereof can be used as immunogens for gsnerating antibodies. The antibody is directed against the target of interest. Preferably, the target is a biologically important polypeptide and administration of the antibody to a mammal suffering from a disease or disorder can resull in a therapeutic benefit in that mammal. However, antibodies may be directed against non polypeptide targets. Where the target is a polypeptide, it may be a transmsambrans 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 oroduced recombinantly or made using synthetic methods. The target may also be isolated from a natural source. 10074) Antigens used in producing antibodies of the invention may include polypeptides and polypeptide fragments of the invention, including epitope A and/or B. A
Cr polypeptide used to immunize an animal can be obtained by stand ard recombinant, chemical synthetic, or purification methods, As is well known in the art, in order to increase immunogenicity, an antigen can be conjugaled to a carrier protein, Commoniy used carriers include, but are rot limited to, keyhole limpet hemocyanin (KLH}, thyroglobulin, bovine serum slbumin (BSA), and iefanus toxoid. The coupled peptide is then used to immunize an animal (2.g., a mouse, a rat, or a rabbit). in addition to such carriers, well known adjuvants can be administered with the antigen to facilitate induction of a strong immune response.
Polyclonal Antibodies
[0075] Polyclonai 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. [ 10076] Animals are immunized against the target, immunogenic conjugates, or . o derivatives by combining the protein or conjugate (for rabbits or mice, respectively) with Freund's complete adjuvant and injecting the sol ution intradermally. One month later the animals are boosted with 1/5 to 1/10 the ’ original amount of peptide or conjugate in Freund's incomplete adjuvant by subcutanecus injection at multiple sites. Seven io 14 days later the animals are bled and the serum is assayed for antibody titer, Animals are boosted until the titer plateaus.
[0677] The mammalian antibody selected will normally have a sufficiently strong binding affinity for the target. For example, the antibody may bind the human anii-igE target with a binding affinity (Kd) vaiue of about 1 x 10° M. Antibody affinities may be determined by saturation binding; enzyme-linked immunoabsorbant assay {ELISA}, and competition assays (e.g., radioimmunoassays).
[0078] To screen for antibodies that bind the target of interest, a routine cross-linking assay such as that described in Antibodies, A Laboratory Manual, Cold Spring
Harbor Laboratory, Ed Harlow and David Lane (1888) can be performed.
Alternatively, epitope mapping, &.g., as described in Champe, et al. J. Biol, Chem. 270: 1388-1394 (1985), can be performed to determine binding.
Monoclonal Antibodies
[0679] Monoclonal antibodies are antibodies which recognizes 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. Moncclonal antibodies may be made using the hybridoma method first described by Kohler et al, Nature, 256: 405 (1975), or may be made by recombinant DNA methods,
[0080] In the hybridoma method, & mouse or other appropriate host animal, such as a rodent, is immunized as hereinabove described io elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind io the protein used for immunization. Alternatively, lymphocytes may be immunized in vitro.
Lymphooyies then are fused with myeloma cells using a suitable fusing agent, such as polyethylene glveo!, to form a hybridoma cell (Goding, Monoclonal
Antibodies: Principals and Practice, pp. 590-103 (Academic Press, 1956).
[0081] The hybridoma ceiis thus prepared are seeded and grown in a suitable culture ¢ 5 medium that preferably contains one or more substances that inhibit the growth or :
N survival of the unfused, parental myeloma calls. For example, if the parental myeloma cells lack the enzyme hypoxanthine guanine phophoribosy! transferases {HGPRT or HPRT), the culture medium far the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (HAT medium), substances which prevent the growth of HGPRT-deficient ceils. Preferred myeioma cells are those : that fuse efficiently, support stabie high-level production of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT msdium. Human myeloma and mouse-human heieromyeioma cell lines have been described for the production of human monoclonal antibodies (Kozbar, J.
immunal. 133: 3001 (1984); Brodeur et al., Monocional Antibody Production
Techniques and Applications, pp. 51-63 {Marcel Dekker, Inc., New York, 1987)).
[0082] After hybridoma cells are identified that produce antibodies of the desired specificity, affinity, and/or activity, 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, 1586). Suitable culture media for this purpose include. The monocional antibodies secreted by the subclanes are suitably separated from the culture medium by conventional immunoglobulin purification procedures such as, for example, protein A-
Sepharose, hydroxylapatite chromatography, gel glecirophoresis, dialysis, or « affinity chromatography.
[0083] DNA encoding the monosional 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 fight chains of the monoclonal antibodies). The hybridoma cells serve as a source of such DNA.
Once isolated, the DNA may be placed into expression vectors, which are then transferred into host cells such as E, coli celis, NSO cells, Chinese hamster ovary (CHO) cells, or myeloma cells to oblain the synthesis of monocional antibodies in the recombinant host cells. The DNA alse may be modified, for example, by substituting the coding sequence for human haavy- and light-chain constant domains in place of the homologous murine sequences (U.S. Pat, No. 4,816,567,
Morrison ef al, Proc. Nat! Acad. Sci, USA 81; 6851 (1984), or by covalently joining to the immunoglobulin polypeptide. ( y Humanized Antibodies “ [0084] Humanization is a technique for making a chimeric antibody wherein substantially lags 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 amine acid residues introduced info it from a source which is norni- human. These non-human amino acid residues are often referred to as “import” residues, which are typicaily {aken from an “import” variable domain,
Humanization can be essentially performed following the method of Winter end co-workers (Jones et al, Nature 32%: 522-525 (1888); Riechman ef al, Nature 332: 323-327 (1988); Verhoeyens et al, Science 239: 1534-1536 {1988}), by substituting non-human CDR's or COR sequences for the corresponding sequences in a human antibody (Ses, e.g, U.S. Pat. No. 4,818,567). As practiced in the pressnt invention, the humanized antibody may have some CDR residues and some FR residues substituted by residues from analogous sites in murine aniibodies.
[6085] The choice of human variable domains, both light and heavy, fo be used in making the humanized antibodies is very important to reduce antigenicity.
According to the so-called "best fit" method, the sequence of the variable domain of & 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- { h | human parent antibody is then accepted as the human framework for the 3 humanized antibody (Sims et al., J. Immunol, 151: 2286 (1993); Chothiz et al,, J.
Mol, Biol, 196; 901 (1887). Ancther method uses a particutar framework derived from the consensus sequence of all human antibodies of a particular subgroup of light or heavy chains. The same framework may be used for several different : humanized antibodies {Carter et al, Proc. Natl. Acad. Sci. USA, 89: 4285 (1282);
Presta et al, J. Immunol. 151: 2623 (1883).
Antibody Fragments : [D086] Various techniques have been developed for the production of antibody fragments. Traditionally, these fragments were derived via proteolytic digestion of intact antibodies (see, e.g, Morimoto &t al., Journal of Biochemical and :
Biophysical Methods 24: 107-1 17 (1882) and Brennan et al., Science 228: 81 (1988)}. However, these fragments can now be produced directly by recombinant { host cells. For example, the antibody fragments can be isolated from an aniibody phage library. Alternatively, F(ab’). -SH fragments can be directly recovered from
E. coli and chemically coupled fo form F(ab"). fragments (Carter et al,
Bio/Technology 10: 163-167 {1982}). According to another approach, Fiabe 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. In other embodiments, the antibody of choise is a single chain Fv fragment (scFv). (PCT patent application WO 93/16188),
PREPARATION OF HIGH AFFINITY ANTIBODIES
[0087] Once the parent antibody has been identified and isolated, one or more amino acid residues may be altered in one or more of the variable regions of the parent antibody. Alternatively, or in addition, one or more substitutions of framework residues may be introduced in the parent antibody where these resultin an improvement in the binding affinity of the antibody, for example, for human igk.
Exampies of framework region residues to modify include those which non- covalently bind target directly (Amit et al. Science 233: 747-753 (10868)}; interact with/effect the conformation of CDR {Chothia et al. J. Mol. Biol. 186: 801-817
BN (1987); and/or participate in the VL-VH interface (EP 239 400 B1). In certain £ embodiments, 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.
[0088] 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 sxample, as a sheet or helical conformation; (b) the charge or hydrophobicity of the molecuie at the target site, or (¢) the bulk of the side chain. Non-conservative substitutions will entail exchanging a member of one of these classes for another class.
[0089] Nucleic acid molecules encoding amino acid sequence varianis are prepared by 2 variety of methods known in the art. These methods include, but are not {imited to, » oligonuclectide-mediatad (or site-directed) mutagenesis, PCR mutagenesis, and i. : 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 oligonucizotide-mediated synthesis. In certain embodiments, the antibody variant will only have a single hypervariabls region residue substituted, e.g. from about two fo about fifteen hypervariable region subsfitutions.
[0090] One method for generating the library of variants is by oligonucieofide mediated synthesis according to the scheme depicted in Figure 2, Three oligonuclentides of : approximaiely 100 nucleotides each may be synthesized spanning the entire light chain or heavy chain variable region. Each oligonucleotide may comprise: (1) a 50 amino acid stretch generated by the triplet (NNK)o where N is any nucleotide and Kis Gor T, and (2) an approximately 15-30 nucleotide overlap with either the next oligo or with the vector sequence at each end. 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 ic only - substitute amino acds in a given CDR or framework region, By using (NNK), al twenty amino acids are possible at each position in the encoded variants. The overlapping sequence of 5-10 amino acids (15-30 nuclostides) will not be subtituted, but this may be chosen io fall within the stacking regions of the (7 framework, or may substituted by a separate or subsequent round of synthesis.
Methods for synthesizing oligonucleotides are well known in the art and are also commercially available, Methods for generating the antibody variants from these oligonucleotides are alse well known in the art, e.g., PCR. 16691] The library of heavy and light chain variants, differing at random positions in their sequence, 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.
[0092] Following production of the antibody variants, 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. « : 10093] One or more of the antibody varianis selected from this iniiial screen may then be ~ scraenad 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 BlAcore™ surface plasmon resonance system (BlAcors, Inc.). A biosensor chip is activated for covalent coupling of the target according to the manufacturer's (BlAcore) instructions. The targst 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 immobiiized material, this represents a range of immobilized target densities on the matrix. Dissociation data are fit to & one-slie model to obtain Kes +/- 5.0.
(standard deviation of measurements). Pseudo-first order rate constant (ks) are calculated for each association curve, and plotted as a function of protein concentration to obtain ke, +/~ 8.8. (standard arror of fit). Equilibrium dissociation constants for binding, Kp's, are calculated from SPR measurements as Kew/Kon.
Since the equilibrium dissociation constant, Kp, is inversely proportional to keg, an estimate of affinity improvement can be made assuming the association rate (Ken) is a constant for all variants.
[0094] The resulting candidate(s) with high affinity may optionally be subjected to one or more further biological activity 2ssays to confirm that the antibody variant(s) with enhanced binding affinity still retain the desired therapeutic aftributes. For {0 example, in the case of an anti-IgE antibody, ons may screen for those that block binding of IgE to its receptor and inhibit the release of histamine. The optimal antibody variant retains the ability to bind the farget with a binding affinity significantly higher than the parent antibody.
[0095] 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 nolypeptidels) and/or covalent modifications such as those elaborated below. For sxample, any cysteine residues not involved in maintaining the proper conformation of the antibody variant may be substituted, generally with serine, 0 improve the oxidative stability of the molecule and prevent aberrant cross linking.
Conversely, (a) 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 {0 fragment). ~ VECTORS 10096] 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. For recombinant 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 sequancad using conventional procedures (e.g. by using oligonuciectide probes that are capable of binding specifically to genes encoding the heavy and light chains of the antibody variant}.
[0097] Many vectors are available. 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.
[0698] The phage expression vector depicted in Figure 1 is comprised of a commonly used M13 vector and M13's own gene ll viral secretion signal for rapid secretion and screening variant Fabs for proper binding spacificity 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 call, but rather the Fabs are secreted into the periplasmic space. Alternatively, the Fabs could be expressed in the : cytoplasm and isolated. The heavy and light chains each have their own viral secration signal, but are dependently expressed from a single strong inducible promoier. 10099] The vector in Figure 1 also provides a His tag and a myc tag for easy purification, as well as detection. A skilled artisan would recognize that 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 prokaryctic or eukaryotic signal sequence suitable for the secretion of the antibody fragments from the chosen host cell. 1 should also be recognized that the heavy and light chains may reside on different vectors,
A. Signal Sequence Component
Co [0100] 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 polypeplide 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. For prokaryotic host cells that do not recognize and process the native antibody signal sequence, the signal sequence may be substituted by a prokaryotic signal sequence selected, for example, from the group of the alkaline phosphatase, penicillinase, lpp, or heat-stable enterotoxin ll leaders, Orin the case of the vecior of Figure 1, the signal sequence chosen was a viral signal sequence from gene lll. For yeast . secretion the native signal sequence may be substituted by, e g., the yeast invertzee leader, o-factor isader {including Saccharomyces and Kluyveromyces a- factor leaders), or acid phosphatase leader, the C. albicans glucoamylase leader, or a signal described in e.g., WO 80/13848. In mammalian cell expression, mammalian signal sequences as well as viral secretory leaders, for example, the herpes simplex gD signal, are available. The DNA for such precursor region is ligated in reading frame to DNA encoding the antibody variant. :
B. Origin of Replication Component
[01061] Vectors usually contain a nucleic acid sequence that enables the vector fo 4 ) replicate in one or more selected host cells. Generally, this sequence is one that enables the vector io replicate independently of the hest 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 2x plasmid origin is suitable for yeast, and various viral origins © (8V40, polyoma, adenovirus, VV or BPV) are useful for vectors in mammalian cells. Generally, the origin of replication component is not needed for mammalian expression vectors (the 8V40 origin may typically be used only because it contains the early promoter).
C. Selection Gene Component { p [0102] Vectors may contain a selection gene, also termed a selectable marker. Typical a selection genes encode proteins that (a) confer resistance fo antibiotics or other toxing, e.g., ampiciliin, neomycin, methotrexate, or tetracycline, (b) complement auxotrophic deficiencies, or (c) supply critical nutrients not available from complex media, e.g., the gens encoding D-alani ne racemase for Basil,
[0103] One example of a selection scheme utifizes a drug to arrest growth of a host cell.
These 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.
[01041 Another example of suitable selectable markers for mammalian cells are these that enable the identification of cells competent to take up the antibody nucleic © acid, such as DHFR, thymidine kinase, metallothionein and «II, preferably primate metallothionein genes, adencsine deaminase, ornithine decarboxylase, elo.
[0105] For example, cells transformed with the DHFR selection gene are first identified by culturing all of the transformants in a culture medium that contain methotrexate (Mt), a competitive antagonist of DHFR. An appropriate host cell when wild-type
DHFR is employed is the Chinese hamster ovary (CHO) cell line deficlent in ~ DHFR activity. ‘ : [0106] Alternatively, host cells {particularly wiid-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-phosphoiransferase (APH) can be selected by cell growth in medium containing z selection agent for the selectable marker such as an aminogiycosidic antibiotic, e.g., kanamycin, neomycin, or G418. (U.8. Pat. No. 4,865,199).
[0107] | A suitable selection gene for use in yeast is the tp1 gene present in the yeast plasmid Yrp7 (Stinchcomb et al., Nature 282: 39 (1979). The trpt 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 trp? lesion in the yeast host cell genome then provides an effective environment for detecting transformation by growth in the ] absence of fryptophan. Similarly, Leu2-deficient ysast strains (ATCC 20,622 or { 38,826) are complemented by known plasmids bearing the Leu2 gene. we D. Promoter Component f0108] 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, f-laciamase and jactose promoter systems, alkaline phosphatase, a tryptophan (trp) promoter system, and hybrid promoters such as the tac promoter. However, other known oo bacterial promoters are suitable. Promoters for use in bacterial systems may also contain a Shine-Dalgarno (S.D.) sequence operably linked fo the DNA encoding the antibody. :
[0109] Promoter sequences are known for eukaryotes, Virtually all eukaryotic genes have an AT-rich region located approximately 25 to 30 bases upsiream 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 tall to the 3' end of the coding sequence. All of these sequences are suitably inserted into eukaryotic expression vactors. 101103 Examples of suitable promeior sequences for use with yeast hosts include the promoters for 3-phosphoaglycerate Kinase or other glycolytic enzymes, such as 4 } : enclase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-8-phosphate isomerase, 3- phosphoglycerate mutase, pyruvate Kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase, 10111] Other veast promoters, which are inducible promoters having the additional advantage of transcription controlied by growth conditions, are the promoter regions for alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, degradalive enzymes associated with nitrogen metabolism, metalicthionain, giyceraldehyde-3-phosphale dehydrogenase, and enzymes responsible for maltose and galactose utilization. Sultable vectors and promoters for Use in yeast axpression are further described in EP 73,657. Yeast enhancers also are advantageously used with yeast promoters.
[0112] 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 oreferably Simian virus 40 (SV40), from heterologous mammalian promoters, 2.0. the actin promoter or an immunogiobulin promoter, from heat-shock promoters— provided such promoters are compatible with the host cell systems.
[0113] The early and late promoters of the 3V40 virus ars 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 chbiained as a Hindlll E restriction fragment. A syslem for expressing DNA in mammalian hosts using the bovine papilioma 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. Aliernatively, human g-interferon cDNA has been sxpressad in mouse celis under the control of a thymidine kinase promoter from herpes simplex virus.
Alternatively, the rous sarcoma virus long terminal repeat can be used as the promoter.
E. Enhancer Element Component
[0114] Transcription of a DNA encoding the antibody of this invention by higher sukaryoies is often increased by inserting an enhancer saguence nic tha vector.
Many enhancer s8qUENCESs a78 NOW known from mammalian genes (globin, 4 i elastase, albumin, o-fetoprotein, and insulin). Typically, however, one will use an enhancer from a eukaryotic cell virus, Examples include the SV40 enhancer on the {ate sids 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. See also Yaniv, Nature 297; 17-18 (1882) on
So enhancing elements for activation of eukaryotic promoters. The enhancer may be soliced into the vactor at a position 5 or 2' io the antibody-encoding sequence, but is preferably located at & site 5' from the promoter.
F. Transcription Termination Component
[01135] Expression vectors used in eukaryaiic host celts {veast, fungl, insect, plant, animal, human, or nucleated cells from other multicellular organisms) may also contain sequences necessary for the termination of transcription and for stabilizing the mRNA. Such seguences are commonly available from the 5' and, occasionally 3 3, untranslated regions of eukaryotic or viral DNAs or cDNAs. These regions ee contain nuclectide segments transcribed as polyadenylated fragments in the untransiaied portion of the mRNA encoding the antibody. One useful transcription termination component is the bovine growth hormone polyadenylation region. See e.g. W0g4/11028.
SELECTION AND TRANSFORMATION OF HOST CELLS
0116] Suitable host cells for cloning or expressing the DNA in the veciors herein ars prokaryotic, yeast, or higher eukaryotic cells, Suitable prokaryotes for this purpose include both Gram-negative and Gram-positive organisms, for example,
Enterobacieria such as E. coll, Enterobacter, Erwinia, Klabsiella, Proteus,
Salmonella, Serratia, and Shigsalla, as well as Bacilli, Pssudomones, and
Streptomyoes. One preferrad E£. coli cloning host is E. coli 204 (ATCC 31,446), alihough other strains such as E. coil B, E. coli X1776 (ATCC 31,537), and E. coi
W3110 (ATCC 27,325) are suitable, These examples are illustrative rather than limiting.
[0117] In addition to prokaryotes, eukaryotic microbes such as filamentous fungi or yeast “ere suitable cloning or expression hosts for antibody-encoding vectors,
Saccharomyces cerevisiae is the most commonly used among lower eukaryotic host microorganisms. However, a number of other genera, species, and strains are commonly availiable and useful herein, such as Schizosaccharomyces pombe, { - Kluyveromyces; Candida; Trichoderma; Neurospora crassa; and filamentous fungi such as e.g., Neurospora, Penicillium, Tolvpociadium, and Aspergilius hosts, such as A. nidujans and A. higer. [011.8] Suktabie 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, Ssetlow et al. eds. Vol. 8, pp. 277-278 (Plenam publishing 1986), Mseda et al., Nature 315, 562-594 (1985). Numerous baculoviral strains and variants and corresponding permissive insect host cells from hosts such as Spodoptera frugiperda (caterpillar), Aedes (mesquite),
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 californica NPV and the Bm-5 strain of Bombyx mori NPV, and such — viruses may be used as the virus hersin according to the present invention, particularly for transfection of Spodopters frugiperda cells. Moreover, plant cells cultures of cotton, corn, potato, soybean, petunia, tomato, and tobacco and also be utilized as hosts. 0119] Vertebrate cells and propagation of vertebrate cells, in culture {tissue culture) has become routine. See Tissue Culfure, Academic Press, Kruse and Patterson, eds. (1973). Examples of useful mammalian host cell ines are monkey Kidney; human embryonic kidnay line; baby hamster kidney cells; Chinese hamster ovary cells/-
DHFR (CHO, Urlaub et al., Proc. Natl. Acad. Sel. USA 77: 4216 (1980)); mouse sertoll cells; human cervical carcinoma cells (HELA); canine kidney cells; human lung cells; human liver cells; mouse mammary tumor; and N30 cells,
[0120] Host cells are transformed with the above-described vectors for antibody production and cultured in conventional nutrient medie modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences.
[0121] The host cells used to produce the antibody variant of this invention may be cultured in & variety of media. Commercially available media such as Ham's F10 (Sigma), Minimal Essential Medium (MEM, Sigma), RPMI-184C (Sigma), and
Dulbecco's Modified Eagle's Medium (DMEM, Sigma) are suitable for culturing
O host calis. in addition, any of the media described in Ham &t al,, Meth. Enzymol. 58; 44 (1979), Barnes st al., Anal. Biochem. 102; 255 {1980}, U.S, Pat. Nos, 4,787,704; 4,657,866; 4,560,855; 5,122,489; 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 faciors (such as insuiin, transferrin, or epidermal growth factor), salts {such as X-chicrides, where
X is sodium, calcium, magnesium, and phosphates), buffers {such zs HEPES), nucleotides (such as adenosine and thymidine), antibiotics (such as
GENTAMYCIN. TM. drug), {race elements (defined as inorganic compounds usually present at final concentrations in the micromolar range), and glucese or an equivalent energy source. Any other necessary supplements may also be included ai appropriate concentrations that would be known io these skilied in the oo art. The culture conditions, such as temperature, pH, and the ike, are those
C previously used with the host cell selected for expression, and will be apparent to _ the ordinarily skilled artisan.
ANTIBODY PURIFICATION
0122] When using recombinant techniques, the antibody variant can be produced : intraceliularty, in the periplasmic space, or directly secreted into the medium. if the antibody variant is produced infracelivlarly, as a first step, the particulate debris, either host cells or lysed fragmants, may be removed, for example, by centrifugation or ultrafiltration. Carter et al., Bio/Technology 10: 163-167 (1822) describe a procedure for isclating antibodies which are secreted fo the seriplasmic space of E, coli. Briefly, cell paste is thawed in the presence of sodium acsiate :
(pH 3.5), EDTA, and phenylmethyisulfonyifluoride (PMSF) ovar about 30 minutes.
Cell debris can be removed by centrifugation. Where the antibody variant is : secreted into the medium, supernatants from such expression systems are generatly first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ulirafiitration unit, A protease inhibitor such as PMSF may be included in any of the foregoing steps to inhibit proteolysis and antibiotics may be included to prevent the growth of adventitious contaminants.
[0123] The antibody composition prepared fron the cells can be purified using, for example, hydroxylapatite chromatography, gel elscrophoresis, dialysis, and Co ; i affinity chromatography, with affinity chromatography being the preferred ourification technigue. The suitability of protein A as an affinity ligand depends on the species and isotype of any immunoglobuiin Fc domain that is present in the antibody variant. Protein A can be used fo purify antibodies that are based on human igG1, 1gG2 or igG4 heavy chains (Lindmark et al., J. Immuno! Meth. 62: 1- 13 {1883)). Protein"G is recommendsd for all mousse isotypes and for human IgG3 (Guss et al, EMBO J. 5: 1567-1575 {1888)). Tha matrix to which the affinity ligand is attached is most often agarose, but other matrices are available. Mechanically stable matrices such as controlied pore glass or poly(styrenedivinyl)benzene allow for faster flow rates and shorter processing times than can be achieved with agarose. Where the antibody variant comprises a CH3 domain, the Bakerbond
ABXTM resin (J, T. Baker, Phillipsburg, N.J.} is useful for purification. Cther techniques for protein purification such as fractionation on an ion-exchange { column, ethanol precipitation, Reverse Phase HPLC, chromatography on silica, chromatography on heparin SEPHARCSE ™ chromatography on an anion or cation exchange resin (such as a polyaspartic acid column), chromatofocusing,
SDS-PAGE, and ammonium sulfate precipitation are also available depending on the antibody variant to be recovered.
[0124] Following any preliminary purification step(s}, the mixture comprising the antibody variant of interest and contaminants may be subjected tc 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).
PHARMACEUTICAL FORMULATIONS
©0128) 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 "pharmacsutically-acceptable” carriers, excipients or stabilizers typically employed in the art (all of which are termed "excipients". For example, buffering agents, stabilizing agents, preservatives, isotonifiers, non-ionic detergents, antioxidants and other miscellaneous additives. (See Remington's Pharmaceutical Sciences, 16th edition, A. Oso}, Ed. {1980)). Such additives must be nontoxic {o the recipients at the dosages and concentrations employed, {5 0126] 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 salis thereof such as citrate buffers {e.g., monosodium citrate-disodium ciirate mixture, citric acid- trisodium citrate mixture, citric acid-monosodium diirate mixture, ec.) succinate buffers {e.g., succinic acid-monescdium succinate mixture, succinic acid-sodium hydroxide mixture, succinic acid-disodium succinate mixture, efe.), 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- monesadium fumarate mixture, etc}, fumaraie buffers (2.9., fumaric acid- monosodium fumarate mixture, fumaric acid-disodium fumarate mixture, monosodium fumarate-disodium fumarate mixture, etc.), giuconate buffers (e.g., {5 giuconic acid-sodium glyconate mixture, gluconic acid-sodium hydroxide mixture, ae giuconic acid-potassium givuconate mixture, etc.}, oxalate buffer (e.g., oxalic acid~ sodium oxalate mixture, oxalic acid-sodium hydroxide mixture, oxalic acid- potassium oxalate mixture, etc), lactate buffers (e.g., lactic acid-sodium lactate mixture, lactic acid-sodium hydroxide mixture, jactic acid-potassium lactate mixture, etc.) and acetate buffers (s.g., acetic acid-sodium acetate mixture, acetic acid-sodium hydroxide mixiure, etc.) Additionally, there may be mentioned phosphete buffers, histidine buffers and {rimethylamine salts such as Tris. 0127) Preservatives may be added io 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, methy! paraben, : ‘propyl paraben, octadecyldimethylbenzyl ammonium chloride, benzalconium halides (2.¢., chloride, bromide, iodide}, hexamethonium chioride, alkyl parabens such as methyl or propy! paraben, catechol, resorcinol, cyciohaxanol, and 3- pentanol.
[0128] Isotonicifiers someiimes known as “stabilizers” may be added to ensure isotonicity of liquid compositions of the present invention and include polhydric sugar alcohols, preferably trinydric or higher sugar alcohols, such as giycerin, erythritol, arabitol, xylitol, sorbitol and mannitol. 10129} Stabifizers refer to a broad category of excipients which can range in function from
Co a bulking agent fo an additive which solubiiizes 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-ieucine, 2- phenylalanine, glutamic acid, threonine, etc., organic sugars or sugar alcohols, such as lactose, trehalose, stachyose, mannitol, sorbitol, xylitol, ribitol, myoinisitol, gaiactitol, glycerol and the like, including cyclitols such as inositol; pelysthylene glveol; amino acid polymers, sulfur containing reducing agents, such as ures, glutathione, thioctic acid, sodium thioglycolate, thioglycerol, e-monothiogiycerol and sodium thio sulfate; low molecular weight nolypeplides (l.e, <10 residues); proteins such as human serum albumin, bovine serum albumin, gelatin or immunogicbuling; hydrophylic polymers, such as polyvinylpyrrolidone . monosaccharides, such as xylose, mannose, fructose, glucose; disaccharides
C 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.
[0130] Non-ionic surfactants or detergents (also known as "wetling agents") may bs 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 polysarbates (20, 80, etc), polyoxamers (184, 188 elc.), Pluronic® polyols, polyoxyethylene sorbitan monoethers (TWEEN®-20, TWEEN®-80, etc.). Non-ionic surfactants may be present in a range of about 0.05 mg/ml to about 1.0 mg/ml, preferably about 0.07 mg/ml to about 0.2 mg/ml. 10131] Additional misceliansous excipients include bulking agents, {&.g. starch}, chelating : agenis (e.g. EDTA), antioxidants (e.g., ascorbic acid, methionine, vitamin £), and so-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 { E microcapsule prepared, for example, by coascervaiion technigues or by interfaciai : polymerization, for example, hydroxymethylceliulose or gelafi n-microcapsule and poly-{methyimethacylate) microcapsuie, respectively, in colloidal drug delivery systems {for example, liposomes, albumin micropheres, microemulsions, nanc- particles and nanocapsules) or in macroemuisions. Such techniques are disclosed in Remington's Pharmaceutical Sciences, 16th edition, A. Osal, £4. (1880).
[0132] The formulations to be used for in vive administration must be sterile. This is readily accomplished, for example, by filtration through sterile filtration membranes. Sustained-release preparations may be prepared, Suitable gxamples of 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. Examples of sustained relesse matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyi-
C ; methacrylate), or poly(vinylalcohol)), polyiactides (U.S. Pal. No. 3,773,919), - copolymers of L-glutamic acid and ethyi-L-glutamate, non-degradable sthyiene- vinyi acetste, degradable lactic acid-glycolic acid copolymers such as the
LUPRON DEPOT ™ (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and 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. When encapsulated antibodies remain in the body for a long time, they may denature or aggregate as a result of exposure to moisture af 37°C resutting 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 bond formation through thio-disuifide interchange, stabilization may be achieved by modifying sulfhydryl residues, lvophilizing from acidic solutions, controlling moisture content, using appropriate additives, and developing specific polymer matrix compositions. 101333 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 gurve a and the pharmaceutical compositions of the invention first in vitro, and then in a useful animal model systems prior fo testing in humans. 10134) in a preferred embodiment, an agueous solution of therapeutic polypeptide, antibody or fragment thereof is administered by subcutaneous injection. Each dose may range from about 0.5 yg to about 50 pg per kilogram of body weight, or more preferably, from about 3 ug to about 30 pg per kilogram body weight. 10135] The dosing schedule for subcutanaous administration may vary form once a= month fo daily depending on a number of clinical factors, including the type of disease, severity of disease, and the subject's sensitivity to the therapsutic agent,
USES FOR THE ANTIBODY VARIANT : [¢136] The antibody variants of the invention may be used as affinity purification agents. : In this process, the antibodies are immobilized on a solid phase such as
SEPHADEX™ resin or filter paper, using methods well known in the art. The
On immobilized antibody variant is contacted with a sample containing the targst to o be purified, and thereafter the support is washed with a suitable solvent that will remove substantially al 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.
[0137] 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. For diagnostic applications, the antibody variant typically will be labeled with a detectable moiety.
Numerous fabels 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 measurad (using a chemiluminometer, for example) or donates energy fo a fluorescent acceptor. Examples of enzymatic labels include luciferases (e.q., firefly luciferase and bacterial luciferase; L1.S. Pat. No. 4,737,458), luciferin, 2.2- 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-phosphale dehydrogenase), heterocyslic oxidases (such as uricase and santhine oxidase), lactoperoxidase, microperoxidase, and the like. ( Techniques for conjugating enzymes io antibodies are described in O'Sullivan et al., Metheds for the Preparation of Enzyme-Antibody Conjugates for Use in ) Enzyme Immunoassay, in Methods in Enzym. (Ed. J. Langone & H. Van Vunakis),
Academic press, New York, 73: 147-165 (1981). 10138] Sometimes, the label is indirectly conjugated with the antibody variant. The skilied artisan The skilled artisan will be aware of varibus techniques for achieving this.
For example, the antibody variant can be conjugated with blotin and any of the three broad categories of labels mentioned above can be conjugaled with avidin, or vice versa, Biotin binds selectively fo avidin and thus, the label can be conjugated with the antibody variant in this indirect manner. Alternatively, fo achieve indirect conjugation of the label with the antibody variant, the antibody variant Is conjugated with a small hapten (e.g. digloxin) and one of the different types of iabels mentioned above is conjugatad with an anti-hapten antibody ( variant (e.g. anti-digioxin antibody). Thus, indirect conjugation of the label with the - antibody variant can be achieved.
[0139] in another embodiment of the invention, the aniibody variant need not be labelad, and the presence thereof can be detected using a labeled antibody which binds to the antibody variant. 10140] The antibodies of the present Invention may be empicyed in any known assay method, such as competilive binding assays, direct and indirect sandwich assays, © and Immunoprecipiiation assays. Zola, Monoclonal Antibodies: A Manual of
Tachnigues, pp. 147-158 (CRC Press, inc. 1987).
[0141] Competitive binding assays rely on the ability of a labeled standard fo 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 fo the amount of standard that becomes bound to the antibodies, To facilitate determining the amount of standard that becomes bound, the antibodies generally are insclubilized before or after the competition. As a result, the standard and test sample that are bound fo the antibodies may conveniently be separated from the standard and test sample which remain unbound, 0142] Sandwich assays involve the use of two antibodies, sach capabie of binding to a different immunogenic portion, or epitope, or the protein to be detected. Ina
C sandwich assay, the test sample to be analyzed is bound by a first antibody which ~ 5 immobilized on a solid support, and thereafter a second antibody binds fo the test sample, thus forming an insclubie three-part complex. See &.g., U.S, Pat. No. 4,376,110. The second antibody may Esalf ba labeled with a detectable moisty : (direct sandwich assays) or may be measured using an anti-immunoglobulin antibody that is labeled with a detectable moiety (indirect sandwich assay). For example, one type of sandwich assay is an ELISA assay, in which case the detectable moiety is an enzyme. 10143] For immunshistochemistry, the tumor sample may be fresh or frozen or may be embedded in paraffin and fixed with a preservative such as formalin, for example.
[0144] The antibodies may also be used for in vivo diagnostic assays. Generally, the antibody variant is labeled with a radionuclectide (such as sup.t11 In, sup.B8 Te, .sup.14 C, .sup.131 1, .sup.3 H, .5up.32 P or .sup.35 8) so that the tumor can be r E localized using immunescintiography. For example, a high affinity anti-lge antibody of the present invention may be used to detect the amount of IgE present in, e.g., the lungs of an asthmatic patient.
[0145] 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. Where the antibody variant is labeled with an enzyme, the kit may include substrates znd cofactors required by the enzyme (e.g., a substrate precursor which provides the detectable chromophore or fluorophores). In addition, 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 concenirations in solution the reagents which substantially eptimize the sensitivity of the asssay. wrticularly, the reagents may be provided as dry powders, usually Invophilizad, cluding excipients which on dissolution will provide a reagent solution having the phropriate concentration.
HN VIVO USES FOR THE ANTIBODY tis contemplated that the antibodies of the present invention may be used to treat “a mammal. In one embodiment, the antibody is administered to 2 nonhuman mammal for the purposes of obtaining preclinical data, for examp fe. Exemplary nonhuman mammals fo be treated include nonhuman primates, dogs, cats, - rodents and other mammals in which preclinical studies are perf armed. Such ) mammals may be established anima! models for a disease (0 be {reated with the ) antibody or may be used fo study foxicity of the antibody of interest, in each of these embodiments, dose escalation studies may be performa d on the mammal.
The antibody or polypeptide is administered by any suitable means, including sarenieral, subcutaneous, intraperiioneal, intrapulmonary, and intranasal, and, if desired for local immunosuppressive treatment, intralesional administration.
Parenteral infusions include intramuscular, Intravenous, intraarierial, intraperitoneal, or subcutaneous adminisiration. In addition, the antibody variant is suitably administered by pulse infusion, particularly with declining doses of the antibody variant, Preferably the dosing is given by injactiome, most preferably intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic. { For the prevention or treatment of disease, the appropriate dosage of the antibody o or polypeptide will depend on the type of disease to be treated, the severity and course of the disease, whether the antibody variant is aciministered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody variant, and the discration of the attending physician. Co
The very high affinity anti-human gE antibodies of the invention may be suitably administered to the patient at one Hime or over a series of treatments.
Depending on the type and severity of the disease, about 0.1 mg/kg io 150 mg/kg le.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, ot by continuous infusion. A typical dally dosage might range from about 1 mg/kg to 100 mg/kg or more, depending on the factors mentioned above. For repeated administrations over several days or fonger, depending on the condition, the treatment is sustained until a desired suppression of disease symptoms occurs.
However, 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 24/04188.
[0150] The antibody variant composition wili be formulated, dosed and administered in a manner consistent with goed medical practice. Faclors for consideration in this context inciude the particular disorder being treated, the particular mammal being { treated, the dlinical 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 ba governed by such considerations, and is the minimum amount necessary to srevent, ameliorate, or treat a disease or disorder, The aniibody variant need not be, but is optionally formulated with one or more agents currently used to prevent ar treat the disorder in question. The effective amount of such other agents depends on the amount of antibody present in the formulation, the type of disorder or treatment, and other factors discussed above, These are genarally used in the same dosages and with administration routes as used hereinbefore or about from 1 io 98% of the heretofore employed dosages. “ 10151] The antibodies of the present invention which recognize Ig as their target may be ; used io treat "IgE-mediated disorders”. These inciude diseases such as asthma, ) n allergic rhinitis & conjunctivitis (hay fever), eczema, urticaria, atopic dermatitis, and food allergies. The serious physiological condition of anaphylactic shock caused by, e.0., bes stings, shake bites, food or medication, is also encompassed under the scope of this invention,
ARTIBODY EPITOPE MAPPING
0152} The term "apitope” refers {o a site on an antigen to which B and/or T celis respond. B-cell epitopes can be formed both from contiguous amine acids or noncontiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amine acids are typically retained on exposure fo denaturing solvents, whereas epitopes formed by tertiary folding are typically lost. on treatment with denaturing solvents, An epitope typically includes at least 3, and mare usually, at least 5 or 8-10 amine acids in a unigus spatial conformation. - Antibodies that recognize the same epitope can be identified in a simple immunoassay showing the ability of one antibody fo block the binding of another antibody to a target antigen.
[0153] 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 igG1 and site directed mutagenesis. { ; 10154] 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 biock binding of IgE to its high affinity recaptar, The peplide scan identified two peptides as potential anti-igE MAD contact sites on IgE, designated Epitope AT and Epitope 'B' (See Figure 11). Although the Epitope ‘A’ and Epitope B' sequences are about 80 amino acids apart in the linear sequences, they are positioned close to each other in the three dimensional structure of IgE. Both are surface exposed, they overiap the FoeRi binding site of IgE, and in both peptides, there are positively charged residues of Arg and hydrophobic residues of Pro.
Figure 12 illustrates the binding region of Epfiope B as determined by ELISA using peptide scan.
[0155] A determination of which amino acid residues are critical for binding of high affinity
C5 antibodies within these epitopss was performed by alanine scanning mutagenesis. — "(Cunningham et al, "High-Resolution Epitope Mapping of hGH-Receptor
Interactions by Alanine-Scanning Mutagenesis” Science 244:1081-1085), Alanine was substituted for each. residue of Epitope A and Epitope B and binding of high affinity monoclonal antibodies was determined. (See Example 12 below and
Figures 13-and 14).
ACTIVE AND PASSIVE IMMUNIZATION : 0156} 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 peplide of the invention with a moiety capable of eliciting an immune response against that peptide. © [0157] it also relates to immunogenic peptides as defined above, for Lse as a pharmaceutical, e.g. in the treatment of igE-mediated diseases or conditions, such as allergy and atopic dermatiiis.
[0158] - 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 io z patient in need of such treatment. eo [6159] The immunogenic peptides of the present invention, while being substantially h | 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. 10169] The initial dose of paplide (e.g. from about 0.2 mg to about 5 mg) 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 ihe age, weight and general health of the patient. Immunization may be "active" or "massive". In "active" immunization the subject receives an immunogenic peptide of the present invention and an anli-IlgE response is actively induced by the subject's immune system. 0161] “Active” immunization, this is preferred for human use, but other mammalian species may be treated similarly, as e.g. in the dog. The term “immunogenic a carrier" herein includes those materials which have the property of independently
B eliciting an immunogenic response in & host animal and which can be covalently coupled to polypeptide either directly via formation of peptide or ester bonds between free carboxyl, amino or hydroxy! groups in the polypepiide and corresponding groups on the immunogenic carrier material, or alternatively by handing through a conventional bifunctional linking group, or as a fusion protein, 10162] Examples of such immunogenic carriers include: albumirs, such as BSA; globulins; thyroglebudins; hemoglobins; hemocyanins (particularly Keyhole Limpet
Hemocyanin [KLH]); proteins extracted from ascarls, s.¢. ascaris extracts such as those described in J. Immunol. 111 [1973] 260-268, J. Immunol, 122 [1879] 302-
308, J. Immun. 98 [1967] 883-900, a Am. J. Physiol. 189 [1860] 575-578 or purified products thereof; polylysine; polygiutamic acid; lysine-giutamic acid copolymers; copolymers containing lysine or ornithine: etc. 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 - alsc be used in the present invention. The purified protein derivative of tuberculin (PPD) is particularly preferred for utilization in the “active” immunization scheme since (1) it does not induce a T-cell response itself (i.e. itis in affect 2 "T-cell hapten"), and yet behaves as a fully processed antigen and is recognized by T- ( . celisas such; (2) & Is known to be one of the most powerful hapten “carriers” in oo the linked recognition mode; and (3) it can be used in humans without further testing.
[0163] The present invention also relates to polynucleotides encoding the peptides of the present invention, veciors comprising said polynuclectides, and celis harboring said vectors. In addition, active immunization may be achieved by administering the polynucelotides encoding the peplides of the present invention. Vectors suitable for such therapy are known in the art and include, e.g., adenovirus vectors. : :
[0164] “Passive” immunization is achieved by administering anti-lgE antibodies of the present invention, to a patient suffering from lgE-mediated disease or condition.
[0165] These antibodies can be prepared by administering an immunogenic peptide of the present invention to a non-human mammal and collecting the resultant
SU anfiserum, Improved titres can be cbiained by repeated injections over a period of ha time. There is no particular limitation to the spscies of mammals which may ba oo used for sliciting antibodies; it is generally preferred to use rabbits or guinea pigs, but horses, cats, dogs, goats, pigs, rats, cows, sheep, eic., can also be used,
Antibody is recovered by collecting blood from the immunized animal after the passage of 1 io 2 weeks subsequently to the last administration, centrifuging the blood and isolating serum from the blood. Monoclonal antibodies may e.g. be : human or muring.
[6166] When immunizing a subject, an antibody of the present invention can be introduced into the mammal by, e.g., intramuscular injection. However, any form of antibody administration may be used. Any conventional liquid or solid vehicle may be empioyed 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 B.8 tc 7.2, preferably about pH 7.0, may be used zs a vehicle, alone or with a suitable adjuvant, such as an aluminium hydroxide-based adjuvant.
[0167] The following examples are offered by way of illustration and not by way of limitation.
EXAMPLES
Example 1 - Humanization of Anti-igE Murine MAb TES-C21 4. 10168] The sequences of the heavy chain variable region (Vu) and the light chain variable region (Vi) of murine mAb TES-C21 were compared with human antibody dermline sequences available in the public daiabases. Several criteria were used when deciding on a template as described in step 1 above, including overall iength, similar CDR position within the framework, overall homology, size of the
CDR, etc. All of these criteria taken together provided a result for choosing the optimal human template as shown in the sequence alignment between TES-C21
MAb heavy and light chain sequences and the respective human template sequences depicted in Figure 3A and 3B, 0169) In this case, more than one human framework template was used to design this antibody. The human template chosen for the Vy chain was a combination of - DP8s (aa residues 1-95) and JH4b (az residues 103-113) {See Figure 3B). The human template chosen for the Vi chain was a combination of L16 (VK subgroup ho lll, 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 Vu and about 74% for VV.
[0170] Once the template was chosen, 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'%). The overlapping nucleotides encoding partial Vy and V, sequences were synthesized in the range of about 63 to about 76 nucleotides with 18 fo 21 nucieotide overlaps. : [06171] PCR amplification of Vy and Vy gene was performed using a biotinylated forward primer containing the specific sequence to the framework region FR1 and an overhanging sequence annealed io the end of leader sequence (Genelll) and a reverse primer from the conserved constant region (Ck 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.
[6172] 5-Phosphorylation of PCR product was performed using 2ug PCR product, 1ul. of ~~ T4 polynucleotide kinase (10 units/ul), 2ul of 10x PNK buffer, 1uL of 10mM ATP ; ) in a total volume of 20uL adjusted by ddHe0. After incubating at 37°C for 45 minutes, and heat denaturation at 65°C for 10 min, the reaction volume was adjusted to 200uL. by adding adH,O for the next step. : 16173] The 100uL of streptavidin-coated magnetic beads were washed twice with 200uL 2x B&W buffer and resuspended in 200uL 2x B&W buffer. The phosphorylated
PCR product was mixed with beads, and incubated at room temperature (RT) for 16 min with mild shaking.
[0174] The beads were sedimentad and washed twice with 200uL 2x B&W buffer. The non-biotinylated ssDNA (minus strand) was eluted with 300uL freshly prepared 0.450 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. ( ) [0175] The ssDNA was precipitated from the supernatant by adding 1uL glycogen or {(10ma/mL}, 4/10 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 20ul. ddH0, The ssDNA was quantitated by spotiing on an sthidium bromide (EiBr) agarose plate with DNA standards, or by measuring
OBzs0. 44 Co
Example 2 :
Cloning of Vy and VV, info Phage-Expression Vector }
[0176] Vy and Vi were cloned into a phage-expression vector by hybridization mutagenesis. Uridinylated templates were prapared by infecting CJ236 E, coli strain (dut” ung’) with M13-based phage (phage-expression vector TN0OD3). [0177} The following components [200 ng of uridinviated phage vector (8.49 kb); 92 ng phosphorviated single-stranded H chain (489 bases); 100 ng phosphorylated single-stranded L chain (528 bases); 1ul. 10X annealing buffer; adjust volume : with ddH,0 to 10 ull were annealed (at about 8-foid molar ratio of insert to vactor) by PCR holding the temperature at 85°C for 5 min {denaturation} and then i 1 ramping to 55°C over 1 hour. The samples were chilled on ice. ) ol [0178] To the annealed product the following components were added: 1.4uL 10 X oo synthesis buffer; 0.5ul. T4 DNA ligase (1 unit/ul}; 1 pL T4 DNA polymerase {1 unit/ul) followed by incubating on ice for 5 min, and 37°C for 1.5 hours. The product was then ethanol precipitated, and dissolved in 10 pL of ddH20 or TE. 0179] DNA was digested with 1 pL Xbal (10unit/uL) for 2 h, and heat inactivated at 65°C for 20 min. Digested DNA was transfected info 50 ul of electro-competent
DH10B cetls by electroporation. The resulting phage were titered by growing on - Xi.-1Biue bacterial tawn at 37°C overnight. Clones were sequenced to confirm composition, :
Example 3
Deep Well Culture for Library Screening .
A. Plating Phage Library : { ! 10180] The phage library was diluted in LB media to achieve the desired number of je plagues per plate. High titer phage was mixed with 200 pL XL-18 cell culture. aml LB top agar was mixed, poured onto an LB plate, and allowed to sit at room temperature for 10 minutes. The plats was incubated overnight at 37°C. 2. Phage Elution 0181} 100 pl of phags slution buffer (10mM Tris-Cl, pH 7.5, 10mM EDTA, 100mM
NaCl) was added to each well of a sterile U-bottom 98 well plate. A single phage plagus from the overnight library plate was transferred with a filtered pipette tip fo a well. The phage siution 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
[0182] XL1B cells from 50mL culture were added io 2xYT media at a 1:100 dilution. The cells were grown at 37°C in a shaker until the Aggy was between 0.9101.2,
C. Infection with Phage in Deep Well Plates
[6183] When the cells reached the appropriate OD, 1M IPTG {1:2000) was added to the
XL 1B culture. The final concentration of IPTG was 8.5m. 750ul. of cell culture was transferred to each well of a 96 well — deep well plate (Fisher Scientific). : . Each well was inoculated with 2510 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 b ol [0184) Foliowing incubation, the deep well plates were centrifuged at 3,250 rpm for 20 : 3 minutes using the Backman JA-5.3 plate rotor. 50uL of supernatant was withdrawn from each well for ELISA. ) E. Innoculation of 15mL Liquid Cultures of XL- 1 cells 0185) XL-1s wera grown at 37°C in the shaker (250rpm) in 2xXYT containing 10 pa/ml of tetracyoline until Aspe = 0.8 fo 1.2. 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 inoculated with 10 pL of phage from the : high titer stock (liter = ~10"" pfu/mL) and incubated for 1 hour at 37°C. The cells were grown overnight at room temperature with shaking.
F. Isolation of Soluble Fab from Periplasm
[6186] © The cells were pelieted in an [EC centrifuge at 4,500 rpm for 20 minutes. Culture medium was removed the peliat was resuspend in 650uL of resuspension buffer
Fr J (50mM Tris, pH 8.0 containing 1mM EDTA and 500mM sucrose), vortexed, and or 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.
Example 4
Framework Modification
[6187] There were twelve murine/human wobble residues within the framework at the potential key positions described above. Position 73 in Vi was kept as the murine residue threonine in the humanization library beacause this position was
: determined to affect binding. It was noted, however, that threonine at VH 73 is a common human residus in the human germline Vy subgroup 1 and 2.
[0188] 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, 87,88 in Vi, and 1, 3, 4, 49, 60, 85 in Vy, (Kabat number system). (See Figure 4) it was later demonstrated that only positions 27 and 62 significantly affected binding in the Vy region (clone number 1136-2C). rd [0189] The primary screen used was a singie point ELISA (SPE} using culture media - [See description below). The primary screen selected clones that that bind fo the antibody’s target molecule. Clones that gave equal or better signal than the parent molecule were selected for the next round of screening,
[0150] In the second round of screening, individual phage were grown ina 15 mi bacterial culture and periplasmic preparations were used for SPE and ELISA titration assays. The clones that retained higher binding in this assay were further characterized. Once ali the selected primary clones were processed, the top 10- 15% clones were sequenced and the clones arranged according fo sequence.
Representatives from each sequence group were compared against sach other and the best clones seiected. Sequences from these chosen clones were } combined and the effects of various combinations were evaluated. 10191) The constructed library was subjected to an ELISA screen for improved binding to b- : the recombinant human IgE, SE44. Clones with binding affinity greater than or murine TES-C21 Fab were isolated and sequenced. Clone 1D #4, 48, 72, 76, and 138 were further characterized. ELISA titration curves for clone 4, 48, 72,78, and 136 are shown in Figure 5A and 58 indicating that their affinity Is similar to the parent, TES-C21. Thess clones compete with murine TES-C21 for binding to - human IgE indicating that the binding epitope was not changed during the : humanization process. The humanized Fabs did not bind to FeeRI-bound gk suggesting that it is less fikely that the humanized antibodies will crosslink the receptor to cause histamine release when they were sonstructed into divalent ig.
[0192] Humanized clone 1386 retained 5 murine heavy chain framework residues (= 84.3 % human Vy framework homology), with a 100% human light chain framework selected for by affinity maturation. The inhibition of IgE binding to FeeR! by the humanized Fab was demonstrated (Figure 8).
Example 5
Single Point ELISA Protocol for Screening anti IgE : [0193] Plates were coated with 2ug/mb 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 be : 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. 50uljwell of bictinylated SE44 at 0.015ug/ml diluted in 0.5% BSA/PBS and 0.05% TWEEN®was then added. Plates were then incubated for 2 hours at room temperature and washed 4X PBST. 50ul/well
StreptAvidin HRP 1:2000 dilution in 0.5% BSA/PBS and 0.05% TWEEN® was added and the plates incubated 1 hour at room temperature. Plates were washed
Bx with PBST. 50ul/well TMB substrate (sigma) was added to develop and then stopped by adding 50ullwell 0.2M HoS0,,
Example 6 :
ELISA Titration: anti IgE N
[0194] 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 a were blocked with 200uLiwel 3 % BSA/PBS for 1 hour at 37°C,
[0193] The plates were washed 4x with PES/0.1% TWEEN® (PBST). 50uliwell Fab {from 18m periplasmic rep) was added starting with a dilution of 1:2 and diluting 3 fold serially in 0.5% BSA/PBS and .05% TWEEN®20. Plates were incubated for 2 hours at room temperature,
[6156] The plates were washsd 4x with PBST and 50ullwell 1:1000 (0.8ug/mi) allution of biotin-sheep anti human Fd in 0.5% BSA/PBS and 0.05% TWEEN®20 was added.
The plates ware incubated again for 2 hours at room temperature.
[0197] Following a wash 4x with PBST, 50ul/weli Neutra-avidin-AP 1: 2000(0.¢ ug/mi} in 0.5% BSA/PBS and 0.05% TWEEN® 20 was added and the plates were incubated 1 hour at room temperature. : } oo
[0198] The plates were washed 4x with PBST. And developed by adding 50ul/well pNPP substrate. Development was stopped by adding 50ul/well 3M NaCH. The absorbance of each well was read at 405nm or 41Cnm.
Example 7
Protocol for Affinity Purification of M13 phage Expressed Soluble Fab
A. DAY1
[0199] Two 500 mL cultures (2xYT) containing 10 mg/ml tetracycline were innoculaled vr A with 5 mi overnight stock XL1B and grown at 37°C to AB00 = 0.810 1.2. IPTG ’ 3 was added to a concentration of 0.5mM. The call culture was then infected with 200 ul phage per culture and incubated for 1 hour at 37° C with shaking.
Following infection, the cells were grown at 25°C overnight with shaking.
B. DAY 2
[0200] Cells were pelleted at 3500 x g for 30 minutes at 4 °C in 250ml centrifuge tubes.
Culture medium was aspirated and the pellets were resuspended in a total of 12- mi lysis buffer (Buffer A + prolease inhibitor cocktall).
Buffer A: {1 liter) 50mM NaH.POy4 6.9 g NaH2PO4HD {or 8 g NaHzPO.) 300mM Nall 17.54 g NaCl 10mm imidazole 0.68 g imidazole (MW 68.08) adiust pH to 8.0 using NaOH po Lysis buffer: - Mix 25 mL of Buffer A with one tabiet of Complete Protease Inhibitor
E Cocktail (Roche, Basel, Switzerland),
[0201] Resuspended cells were transferred into 2 50m conical tube and lysed with 100pL 100 mg/mL lysozyme by inverting the tube several times until the mixture moves iogether as a blob (due to the lysis}, Cells were sonicated an ica followed by the addition of 10 pL DNase | (about 1000 units) and gently rocked at 4°C for minutes. Debri was pelieted by centrifugation at 12000 x g for 30 minutes at 4 °C, using 50 mL centrifuge tubes. Supernatants were fransferred fo a new conical tube and stored at 4 °C.
[0202] Ni-NT agarose (Qiagen, Valencis, CA} was used to purify the soluble Fabs according to the manufacturer's protocol. The lysate was mixed with Ni-NTA and loaded into & column. The flow through was collected for SDS-PAGE analysis.
The column was washed with 20 mL buffer (50mM NaH;P C4, 300mM NaCl, 15mM imidazole, adjust pH to 8.0 with NaOH) foltowed by a 20 mL wash with 50mm NaksPO,, 300mM NaCl, 20mM imidazole. Fabs were eluted with 6 x 500 : pL eiution buffer (30m NatPO4, 300mM NaCl, 450mM imidazole, adjust pH to 8.0 with NaOH) and analyzed by SDS PAGE. Column fractions were stored af 4 °C. Column fractions wars analyzed by SDS-PAGE and the fraction with the greatest amount of Fab was selected and dialyzed in PBS at 4 °C. (= Example 8 - Soluble Receptor Assay [0203} A 96 well assay plate suitable for ELISA was coated with 0.05 mL 0.5ug/mL FeeRl alpha-chain receptor coating buffer (59 mM carbonate/bicarbonate, pH 9.8) for 12 hours at 4-8°C. The wells ware aspirated and 250ul blocking buffer (PBS, 1%
BSA, pH 7.2) was added and incubated for 1 hour at 37°C. in a separate assay plate the samples and reference TES-C21 MAbs were fitered from 200 to 0.001ug/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 bistinylated igk was added and the plate incubated for 2-3 hours at 25°C. The FoeRl — coated wells were washed three times with PBS and 0.05% TWEEN 20 and 50uL from the sample wells were transferred and incubated with agitation for 30 minutes at 25°C. Fifty pliwell of 1 mg/mL Streptavidin-HRP, diluted 1:2000 in assay buffer, was incubated for 30 er minutes with agitation and then the plate was washed as before. Fifty uliwell of or TMB substrate was added and color was developed. The reaction was stopped by adding an equal volume of 0.2 M M280, and the absorbance measured at 450nm.
Exampie 9
Binding of Antibodies to Igk-loaded FoeRl 16204] Antibody binding to human IgE associated with the alpha-subunit of FeeR] was determined by preincubating with 10 pg/ml human IgE for 30 min at 4°C. Plates were washed three times followed by a one hour incubation with varying concentrations of either muring anti-human gk MAb E-10-10 or the humanized
Fab variant. Binding of Fabs was detacted with a biotin labeled anti human Fd antibody followed by SA-HRP. Murine MAb E-10-10 was detected by Goat anti-murine Ig Fo HRP-conjugated Ab.
Exampie 10
Cione Characterization
[0205] Each candidate was assayad for binding affinity and positive clones were i 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. b 10206] A library of variants was created. The amino acid sequences for the various
CDRs which demonstrated improved affinity are depicted in Table 1. Figure 7 Co presents high affinity candidates having combinations of substitutions,
TABLE 1. reoRLT jeoma:
P RASQSIGTNIM SEQIDNOS | P MYWLE SEQID NO 15 #2 RASQRIGTNIH | SEQ IDNOT #2 OYYWLE | SEQ IDNO 17
CDRLZ: | CDRHZ: i rE TERE | PESTER SE #2 YASESY [SEQ ID NO 10 | #2 EIDPGTFTTNYNEKFKA SEC ID NO 20 #3 VYASESDS | SEQID NO 11 | #3 EISPDTFTTNYNEKFKA | SEQID NG 21 ~ rn wr ET SEE TEE ET
PaCS SEES TH SETS
FRET SEA BG | ESSER RoE
IE CDRHS:
Ir rSENODY[SEBNG ES #1 FBHFSGMNYDYFDY | SEQ ID NO 27
P = Parent
{0207 Nineteen heavy chain variants are presented in Figure 9 and 35 light chain variants are presented in Figure 8. Three candidates were further characterized for binding affinity and these are pressnied in Table 2. ~ TABLE2 Binding Affinity ee [re EE
Binding Affinity
Tee eee]
Exampie 11
Expression and purification of anti-lgE antibodies and HRP-conjugation
[0208] High affinity MAbs candidates were generated. For the generation of intact anti-
IgE MAbs, the heavy and light chains variable regions were PCR amplified from phage vectors templates and subcloned separately info 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 ling N30 using electroporation by techniques well known in the art. See, e.g, Liou etal. J
Immunol. 143(12):3967-75 (1989). Antibodies were purified from the single stable a cell ine supernatants using protein A-sepharose (Pharmacia). The concentration of the antibody was determined using spectrophotometer at 280nm and FCA ~ zssay (IDEXX). 10209] Purified antibodies were conjugated by horseradish peroxidase (HRP) using peroxidase conjugation kit (Zymed Labs, San Francisco, CA) according to the manufacturer's protocol. The fiter of sach conjugated anti-IgE MAb was determined using ELISA with plates coated with a monoclonal human IgE (SE44),
[0210] The following cultures have been deposited with the American Type Culture
Collection, 10804 University Boulevard, Manassas Va, 20110-2208 USA (ATCC):
Thon mee
Anti-igk CL-5A | PTA-5679 | December 3, 2003
Anti-igk CL-5! | PTA-5680 } December 3, 2003 [02113 This deposit was made under the provisions of the Budapest Treaty on the international Recognition of the Deposit of Microorganisms for the Purpose of
Patent Procedure and the Regulations thereunder (Budapest Treaty). This pred assures maintenance of a viable culture for 30 years from the date of deposit. The oo organism will be made available by ATCC under the terms of the Budapest
Treaty, which assures permanent and unrestricted availability of the progeny of the culture to the public upon issuance of the periinent U.S, patent. 0212) The assignae of the present application has agreed that if the culture on deposit should dis or be lost or destroyed when cultivated under suitable conditions, twill be promptly replaced on notification with a viable specimen of the same culture.
Availability of the deposited strain is not to be construed as a license fo practice : the invention in contravention of the rights granted under the authority of any : government in accordance with its patent laws. 10213) The foregoing written specification is considered to be sufficient to enable one skilled in the art to practice the invention. The present invention is not io be limited in scope by the cultures deposited, since the deposited embodiments are intended - as illustration of one aspect of the invention and any culture that are functionally equivalent ars within the scope of this invention, The deposit of material herein does not constitute an admission that the written description herein contained is inadequate to enable the practice of any aspect of the Invention, including the best mode thereof, nor is it to be construed as limiting the scope of the claims fo the specific illustration that it represents. indeed, various modifications of the invention in addition to those shown and described hersin will become apparent fo those skilied in the art from the foregoing description and fall within the scope of the appended claims.
Example 12
Mapping of the High Affinity Binding Epitope of Human IgE
A. Paptide synthesis and anti-IgE binding assay
[0214] Studies have shown that 1gE binds io its receptor through the Cu3 domain. Since the HA Anti-IgE antibodies of the present invention very sfficientty block Igk from binding to its receptor, we mapped the epitope using peptides that encompassed the entire CH3 domain. First, we prepared two V5-4agged peptides, one comprised the entire constant region of human Igk and one comprised just the
Cn2-Cu3 region of human gk. These fwo peptides were expressed by in Vitro transcription-fransiation and used in a Western blot assay to detect HA Anti-igE { mo! MAb binding. Both CL-2C and CL-5! MAbs were abie to bind to the intact human ~ IgE as welt as both the peptides. 0215] To map the epliope more specifically, 73 overlapping peplides were synthesized which encompassed amino acid residues 141 fo 368 of human IgE, which included the entire Cy3 domain. Each peplide consisted of 12 amino acid residues having a 3 amino acid overlap with the 3’ end of the previous peptide,
SPOTs membranas were synthesized with fluorenylmethoxycarboyl (Fmoc) amino acids on celiulose membrane. The membranes were rinsed in methanol and then washad in TBS (pH 7.5) 3X for 10 min. After an overnight incubation in blocking solution (5% milk or 3% BSA in TBS), HRP-labeled anti-igE MAbs diluted in blocking solution were incubated with the membrane for 3 hra. After washing 3X for 15 min in TBS-TWEEN®, using SuperSignal HRP substrate (Parse), IgE reactivity was measured by chemiluminescence exposure of BioMax MS film { (Kodak) for the desired time. - [0216] The results from the experiment indicate that the HA Anti-Igk MAbs bind to two regions in the Cn portion of IgE, which are represented by the following two peptide sequences: NPRGVEAYLSRP (epitope "A") and HPHLPRALMRST : - {Epitope “B"). (See Figure 12.) Binding fo epitope A was several times weaker than to epitopes B.
B. Alanine Scan Mutagenesis
[0217] An alanine scan atong with substitutions of amino acids In the peptides with those that are found in igG1 was carried out io determine which amine aclds are critically involved in MA Anti-igE MADb's binding to these peptides. Amino acids that were determined to be important for HA Anti-igE MAb binding were replaced using an in vitro mutagenesis strategy in the ¢ chain of IgE. Another peptide covering the C2 and Ce3 regions as described earlier was also used in this study, {See Figures 13 and 14). .
[6218] The EU numbering scheme for human gk amino acid residues has been used.
Polymerase chain reaction (PCR) was used to amplify the entire Fc region of IgE, oo and a truncated form of IgE Fc containing only the CH2-CH3 domain. The DNA products were cloned directly into pcDNAZ expression vector {Invitrogene,
Carlsbad CA) using TOPC cloning {invitrogene, Carlsbad, CA). 10219] Mutagenesis in IgE-Fc was performed using overlapping PCR (Ho et al, 1589). : = ! The DNA products were purified by agarose gel electrophoresis, digested with an oo appropriate restriction enzyme(s), and subcioned info the pcDNAS3 expression vector, For each variant construct, PCR amplified regions were completely sequenced using dideoxynucieotide method from both strands of DNA. ] Recombinant human IgE Fc and its mutants were expressed usihg reticulocyte lysate based in-vitro transcription and translation coupled system (Promega,
Madison, Wi)
[0220] Lysates from this in-viiro transcription and translation coupled system (10 ul reaction mix) were subjected to SDS-PAGE {12 %) and then fransferred to nifrocelluialose 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 detecied using & goat anti-human lgG Fe conjugaied io horseradish peroxidase (Jackson Labs, Bar Harbor, Maing) 0 and the immunoreactive bands ware visualized by the SuperSignal Western : blotting detection kit (Pierce), Anti-VE antibodies were usad as a positive contral that detected the V5 tag introduced at the C-terminus of these peptides. The
Western blot with anti-v5 antibodies demonstrated that all the peptides were expressed at almost equal tevel. Interestingly, HA Anti-IgE MAbs were able to bind io the peptide that carred mutations in epilope ‘A’, but, they did not bind to the peptide that carried mutations in epitope 'B’, indicating that this second site was more important for binding. {See Figures 15).
Example 13
Active Immunization of Transgenic Mice Using an immunogenic Peptide of
Epitope B 10221) Transgenic mice that constitutively express human IgE were used to demonstrate the active production of antibodies fo a human immunogenic peplide of Epitope B,
Two fusion peptides, each comprising an immunogenic peptide of the invention, a . cysteine residue and KLH, were chemically synthesized. Tha sequence of . peptide 1 was: . {(KLH-Cys) - Leu Pro Arg Ala Leu Met Arg Sar Thr oo : and the sequence of peptide 2 was: :
Leu Pro Arg Ala Leu Met Arg Ser Thr — {Cys-KLH) : 10222] The transgenic mice were inlected subcutaneousty with 20 vg of the immunogenic peptide in complete Freund's adjuvant (Difco Laboratories, Detroit, Mi) in 200 pL of PBS pH 7.4. At two-week intervals the mice were twice injected : subcutaneously with 20 pg of the peplide immunogen in incomplete Freund's adjuvant. Then, two weeks later and three days prior to sacrifice, the mice were again injected intraperitonsally with 20 ug of the same immunogen in PBS. Serum was collected and tested for the presence of anti-lgE antibodies spadific for
Epitope B. As seen in Figure 168, the peptide elicited anti-IgE antibodies in these fransgenic mice. 10223] Those skilled in the art will recognize, or be abie io ascertain using no more than routing experimentation, many equivalents to the specific embodiments of the invention described herein. Such sguivalenis are intended to be encompassed by the following claims.
SEQUENCE LISTING
<110> TANOX, INC,
STNGH, Sanjaya
HUANG, banyang
FUNG, Sek Chung <120> Identification of Unique, High Affinity IgE Epitopes <130> TNX-1030 <150> PCT/US04/02892 <151> 2004-02-02 <150> PCT/US04/02894 <151> 2004-02-02 <160> 77 pd <170> PatentIn version 3.2 ee 210-1 - «211» 107 ; <Z12> PRT ~ <213> Murine <220> <221> wmisc_Teature <223> TES-CZ1 LIGHT CHAIN <400> 1
Asp Ile Leu teu Thr Gin Ser Pro Ala Ile Leu Ser val ser Pro Gly 1 5 10 15
Glu Arg val Ser Phe Ser Cys Arg Ala Ser 6In Ser Ile &ly Thr Asn
Ile His Trp Tyr Gin Gin Arg Thr Asp Gly Ser Pro Arg Leu Lau Ile 40 45 x
FT Lys Tyr Ala Ser Glu ser Ile ser Gly Ile Pro Ser Arg Phe Ser Gly fe 50 55 50 ser Gly ser Gly Thr Glu phe Thr Leu Asn Ile Asn Ser val Glu ser 65 70 75 80
Glu asp Tle Ala Asp Tyr Tyr Cys Gin Gin Ser Asp Ser Trp Pro Thr 85 80 85 '
Thr phe Gly Gly Gly Thr Lys teu Glu Ile Lys 100 105 <210> 2 <21l> 107 : <212> PRT <213> Human <220> <221> misc_feature
Page 1
<223> L16/-3K4 human light chain consensus sequance template <400> 2
Glu Ile val met Thr Gln Ser Pro Ala Thr Leu Ser val Ser Pro Gly 1 3 10 15
Glu Arg Ala Thr Ley Ser Cys Arg Ala Ser Glin Ser vel ser Ser Ash :
Leu sla Trp Tvr Gin Gin Lys Pro Gly Gln Ala Pro Arg Leu Leu Ile 40 45
Tyr Gly Ala Ser Thr Arg Ala Thr Gly Ile pro Ala Arg phe Ser Gly 50 55 . 680 ( ser Gly ser Gly Thr ¢lu Phe Thr Leu Thr Ile Ser Ser Leu Gin Ser
Tr €5 70 . 75 80
Glu Asp Phe Ale val Tyr Tyr Cys Glin Glin Tyr Asn Asn Trp Pro Leu 85 80 95
Thr phe Gly Gly Gly Thr Lys val Glu Ile Lys 10C 105 <210> 3 «211» 123 «212» PRT <213> MURINE <220> <221> misc feature . <223> TES-CZ1 Heavy Chain <400> 3 ¢In val Gln Leu 6in Gln Ser Gly Ala Glu Leu Met Lys Pro Gly ala ( 1 5 10 15 ser val Lys Ile ser Cys Lys Thr Thr ¢ly Tyr Thr phe Ser Met Tyr 20 25 30
Trp Leu Glu Trp val Lys Gin Arg Pro Gly His Gly Leu Glu Trp val 35 40 45
Gly Glu Tle ser pro Gly Thr rhe Thr Thr Asn Tyr asn Glu Lys Phe 50 55% 60 tys Ala Lys Ala Thr phe Thr Ala Asp Thr Ser Ser asn Thr Ala Tyr 65 70 75 80
Leu Gn Leu Ser Gly Leu Thr Ser Glu Asp ser Ala val Tyr phe Cys 85 a0 es
Ala arg rhe Ser His Phe Ser Gly Ser Asn Tyr Asp Tyr Phe Asp Tyr
Page 2
100 105 110
Trp Gly Gin Gly Thr Ser Leu Thr val Ser ser 115 120 «210 4 «<Z11l> 113 =Z212> PRT <713> Human <220> : <221> misc_Teature <223> DP88/1M4b human heavy chain consensus sequence template <400> 4 :
Gin val Gin Leu val Gln Ser Giy Ala Glu val Lys Lys Pro Gly Ser
Lo 1 5 10 15 ser val Lys val ser Cys Lys ala Ser Gly Gly Thr phe Ser ser Tyr
Ala Ile ser Trp val Arg Gin Ala Pro Gly Gin Gly Leu Glu Trp Met 40 45
Gly Gly Iie Ile pro Ile Phe Gly Thr Ala Asn Tyr Ala Gin Lys Phe 30 55 £0 cln Gly Arg val Thr Ile Thr Ala Asp Lys Ser Thr Ser Thr ala Tyr es 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala val Tyr Tyr Cys 85 90 85 ala arg Tvr Phe Asp Tyr Leu val Gln Gly Thr Ser teu Thr val Ser a 100 105 110 <210> 5 <231> 11 <21id» PRT <213> ARTIFICIAL <220> «223» TES-C21 CDRLI SEQUENCE (TABLE 1) . <400> 5
Arg Ala ser Gln Ser ITe Gly Thr Asn Iie His 1 5 10 <210> 6 <211l> 11 <212> PRT <213> ARTIFICIAL .
Page 3
<220> <273> (CDRLL VARIANT SEQUENCE #1 (TABLE 1) : <400> 6
Arg Ala Ser Arg Ser Ile Gly Thr Asn Ile His 1 5 0 ibs 7 » <2il» 11 <212> PRT ‘ . «213» ARTIFICIAL . <220> <223> CDRLI VARIANT SEQUENCE #2 (TABLE 1) <400> 7 {0 Arg Ala Ser Gin Arg Ile Gly Thr Asn Ile His oo 1 5 10 : <Z210> B «211» 7 <212> PRT <213» ARTIFICIAL <220> «223» TES-C21 CDRLZ SEQUENCE (TABLE 1) ' } <400> 8
Tyr Ala ser Glu Ser Ile Ser 1 5 <210> 9 <211l> 7 «212» PRT <213> ARTIFICIAL <220> } <223> CDRL? VARIANT #1 (TABLE 1)
Fo : fo <400> 9
Tyr Ala Tyr Glu Ser Ile ser 1 5 <Z3i0> 10 <211> 7 <2i2> PRT . <213> ARTIFICIAL <220> <223> CDRLZ VARIANT #2 (TABLE 1) . <400> 10
Tyr Ala Ser Giu Ser Ile Tyr 1 3 <210> 11 ’ <211> 7 <212> PRT . Fage 4
<213> ARTIFICIAL <220> <223> CDRLZ VARIANT #3 (TABLE 1) <400> 11
Tyr Ala Ser Glu Ser Asp ser 1 5 <210> 12 , 231 7 : <212> PRT <213> ARTIFICIAL <220> : <223> CDRLZ VARIANT #4 (TABLE 1) oo <400> 12 { . ’ Tyr Ala Ser Glu Ser Glu Ser 1 5 : <210> 13 <2il> 9 Co «212> PRT <213> ARTIFICIAL <220> «273%» TES-C21 CDRL3 (TABLE 1) } <400> 13
Gln Gln Ser Asp $er Trp Pro Thr Thr oo 1 5 <210> 14 <Zil> 9 <Z1Z2> PRT <213> ARTIFICIAL a «220 {0 <223> CDRL.3 VARIANT {TABLE} - <400> 14 pia ala ser Trp ser Trp Pro Thr Thr <210> 15 : : <211> 5 : : ‘ <212> PRT <213> ARTIFICIAL <220> «223» TES-CZ21 CDRHL ; ] <400> 15
Met Tyr Trp Leu 6Tu 1 5 <210> 16 <211> 5
Page 5
<212> PRT : <213> ARTIFICIAL : <220> <223> CDRH1 VARIANT #1 (TABLE 1) <400> 16
Trp Tyr Trp Leu Glu 1 5 <210> 17 <21l1i> 5 <212> PRT <213> ARTIFICIAL . . ‘ <220> <223> CDRHL #2 (TABLE 1) { <400> 17
Tyr Tyr Trp Led Glu 1 5 <210> 18 <21ll= 17 <212> PRT <213> ARTIFICIAL . <220> : <223»> TES-C21 CDRHZ (TABLE 1D <400> 18
Glu Ile ser Pro Gly Thr Pha Thr Thr Asn Tyr Asn Glu Lys Phe Lys 1 5 10 : 15
Ala
B 210» 19 { <Z11> 17
Fie <212> PRT : - <213> ARTIFICIAL . <220> <223> CDRHZ VARIANT #1 (TABLE 1) i <400= 19
Glu Ile Glu Pro Gly Thr phe Thr Thr Asn Tyr Asn Glu Lys Phe Lys 1 5 10 15
Ala ,<210> 20 . <211l> 17 <212> PRT <213> ARTIFICIAL <220> <223> CDRHZ VARIANT #2 (TABLE 1D ’
Page §
<400> 20
Glu Ile Asp Pro Gly Thr Phe Thr Thr Asn Tyr Asn Glu Lys Phe Lys i 5 10 15
Ala oo <210> 21 «211s 17 <212> PRT <213> ARTIFICIAL <220> <223> CDRMZ VARIANT #3 (TABLE 1) { . <400= 21 - Glu Ile Ser Pro Asp Thr phe Thr Thr Asn Tyr Asn Glu Lys Phe Lys 1 5 10 15
Ala <210> 22 <211s> 17 «212» PRT <213> ARTIFICIAL <220> <223> CDRH2 VARIANT #4 (TABLE 1) : <400> 22 ctu Ile ser Pro Giu Thr Phe Thr Thr Asn Tyr Asn Glu Lys Phe Lys 1 5 10 15
Ala ( <210> 23 «211» 17 <212> PRT <213> ARTIFICIAL <220> ' <223> CDRH? VARIANT #5 (TABLE 1) <40Q> 23
Glu Ile ser pre Gly Thr Phe Glu Thr Asn Tyr Asn Glu Lys Phe Lys 1 5 10 15
Ala <210> 24 oo <21l> i7 «212» PRT <213> ARTIFICIAL
Page 7
<220> <223> CDRH2Z VARIANT #6 (TABLE 1) : <400> 24 :
Glu ze ¢lu Pro Gly Thr phe Glu Thr asa Tyr Asn Glu Lys the Lys 1 5 1
Ala i <210> 25 <211> 17 «212» PRT <213> ARTIFICIAL . { <220>
Te <223> CDRHZ VARIANT #7 (TABLE 1) <400> 25
Glu Ile Asp Pro Gly Thr Phe Glu Thr Asn Tyr Asn Glu Lys Phe Lys i 5 10 15
Ala <210> 26 <211l> 14 <21i2> PRT «213» ARTIFICIAL <220> <Z23> TES-CZ1 CORH3 (TABLE 1) <400> 26
Phe ser His Phe Ser Gly $er Ash Tyr Asp Tyr Phe Asp Tyr 1 5 10 { = <210> 27 <21l> 14 <212> PRT <Z13> ARTIFICIAL <220> <223> CDRH3 VARIANT #1 (TABLE 1) «400» 27
Phe Ser His Phe Ser Gly Met Asn Tyr Asp Tyr Phe Asp Tyr 1 5 10 <210> 28 <211> 14 <212> PRT <213> ARTIFICIAL <220> «223» (CDRH3 VARIANT #2 (TABLE 1) <400> 28
Page §
Phe Ser His Phe Ser Gly GIn Asn Tyr hep Tyr Phe Asp Tyr 1 5 } I <210> 29 «211> 14 <212> PRT <213> ARTIFICIAL <220> <223> CDRH3 VARIANT #3 (TABLE 1) : <400> 29 rhe Ser His Phe hr Gly Ser Asn Tyr hse Tyr Phe asp Tyr 1 oo <210> 30 : <211> 23 <212> PRT . <213> ARTIFICIAL <220> <223> FRLL VARIANT 136 <400> 30 | | -
Glu Ie val met Thr Gin Ser Pro Ala Thr leu Ser val Sar Pro Gly 1 5 10 15
Glu arg Ala Thr Leu Ser Cys : <210> 31 <211> 23 . <212> PRT <213> ARTIFICIAL . <220> <223> FRL1 VARIANT 1 { = <400> 31° oe Asp Ile Leu Met Thr Gin Ser Pro Ala Thr Leu Ser val Ser Pro Gly 1 5 10 15
Glu Arg Ala Thr Leu Ser Cys 20 «210» 32 . <231> 23 : <212> PRT «213» ARTIFICIAL <220> <223> FRLL1 VARIANT 2 <400> 32
Asp Iie val Leu Thr &In Ser Pro Ala Thr Leu Ser val Ser Pro-Gly 1 5 10 15
Page 9 .
Glu Arg Ala Thr Leu Ser Cys <210> 33 «21%» 23 <212> PRT <213> ARTIFICIAL . <220> «223» FRLL VARIANT 4 <400> 33
Asp Ile Leu teu Thr Gin Ser Pro ala Thr teu Ser val Ser Pro Gly 1 5 10 15 . &lu Arg Ala Thr Leu Ser Cys ( 20 <210> 34 <21l> 23 : «212» FRT <213> ARTIFICIAL <Z220> <223> FRL1L VARIANT 13 <400> 34 clu Ile val Leu Thr Gln Ser Pro Ala Thr Leu Ser val Ser pro Gly 1 5 10 5
Glu Arg Ala Thr Leu Ser Cys 20 <210> 35 <21ll> 23 <212> PRT <Z13» ARTIFICIAL ( <220> : <223> FRL1 VARIANT 18 <400> 35
Glu Ile Leu Leu Thr Gin Sar Pro Ala Thr Leu Ser val Ser Pro Gly 1 5 10. 15
Glu Arg Ala Tar Leu Ser Cys 0 . <210> 36 <211l> 23 <212> PRT <Z13> ARTIFICIAL . <220> <223> FRL1 VARIANT 25 <400> 36
Asp Ile val Met Thr Gln Ser Pro Ala Thr Leu Ser val Ser Pro Gly
Page 10
1 5 10 15
Glu Arg Ala Thr Leu Sar Cys ) : : . <210> 37 <21i> 23 <212> PRT : <213> ARTIFICIAL } <220> <223> FRL1 VARIANT 27 . <400> 37
Glu Tle teu Met Thr &ln Ser Pro Ala Thr Leu Ser val ser Pro Gly ~ 1 5 10 15 oo Glu Arg Ala Thr teu Ser Cys «210» 38 <Z2il> 15 <Z12> PRT <Z13» ARTIFICIAL <220> : «223> FERLZ VARIANT 136 <400> 38 :
Trp Tyr Gin Gin Lys Pro Gly 61n Ala Pro Arg Leu Leu Ife Tyr
CX 5 10 15 <210> 30 «211» 15 <212> PRT <213> ARTIFICIAL a <220> [ «223». FRLZ VARIANT 1 i <400> 39
Trp Tyr Gin Gln Lys Pro Gly Gin Ala Pro Arg Leu Leu Ile Lys 1 5 oie 15 <210> 40 <211l> 32 <212» PRT <213> ARTIFICIAL <220> <223> FRL3 VARIANT 136 <400> 40
Gly Ile Pro Ala Arg Phe ser Gly Ser Gly Ser Gly Thr Glu phe Thr 1 5 10 15
Leu Thr Ile Ser Ser Leu Gln Ser Glu Asp Phe Alfa val Tyr Tyr Cys 20 25 30
Page il oo
«<210> 41 <211> 32 <212> PRT . © «213» ARTIFICIAL <220> «223» FRL3 VARIANT 1 <400> 41 ‘ : Gly Ile Pro ser Arg phe Ser Gly Ser Gly Ser Gly Thr Glu phe Thr
SL 5 1.0 15
Ley Thr Ite Ser Ser Leu Gin Ser Glu Asp Phe Ala val Tyr Tyr Cys : {0 <210> 42 <211l> 32 : <212» PRT «213» ARTIFICIAL : <220> <223»> FRL3 VARIANT 13 <400> 42
Gly Ile Pro Ala Arg Phe Ser Gly Ser Gly Ser Gly Thr Glu Phe Thr 1 5 10 15 rau Thr Ile Ser Ser Leu Gin Ser Glu Asp Phe Ala Asp Tyr Tyr Cys 20 25 30 <210> 43 <211> 32 <Z12> PRT <213>. ARTIFICIAL <220> - <223> FRL3 VARIANT 18 : <400> 43 - Gly Ile Pro ser Arg Phe ser Gly ser Gly ser Gly Thr Glu Phe Thr 1 3 10 15
Leu Thr Ile ser Ser Leu Gin Ser Glu Asp phe Ala Asp Tyr Tyr Cys : 20 25 30 <210> 44 ‘ . <211> 10 «212» PRT : <213> ARTIFICIAL <220> «223» FRL4 VARIANT <400> 44 phe Gly Giy &ly Thr Lys val Glu Ile Lys 1 5 : 10
Fage 12
<210> 45 «211» 30 <212> PRT <213> ARTIFICIAL . <220> <223> FRHML VARIANT 136 <400> 45
Gain val 6In Leu val Gln ser Gly Ala Glu val Met Lys Pro Gly Ser } 1 5 : 10 15
Ser val Lys val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Ser 25 . 30 oo <2i0> 48
SA <211> 30 <212> PRT } . «213» ARTIFICIAL <220> <223> FRHL VARIANT 2 <400> 46
Gin val Gin Leu val Gin Ser Gly Ala GTu val Lys Lys Pro Gly Ser i 5 10 13 ser val Lys val Ser Cys Lys Ala Ser Gly Tyr Thr phe Ser 20 25 30 <210> 47 : <211> 14 <212> PRT <213> ARTIFICIAL <220> <Z23> FRH2Z VARIANT 136 ( <400> 47 — Trp val Arg Gin Ala Pro Gly His Gly Leu Glu Trp Met Gly 1 5 10 <210> 48 <211> 14 <212> PRT <213» ARTIFICIAL <220> «223» FRHZ VARIANT Z <400> 48
Trp val Arg Gin Ala Pro Gly Glin Gly Leu Glu Trp Met Gly 1 5 1 <210> 48 <211> 14 : <212» PRT <213» ARTIFICIAL
Page 13
<220> Co <223> FRHZ VARIANT 8 : <400> 48 : )
Trp val Arg Gin Ala Pro Gly Gin Gly Leu &lu Trp val Gly 1 5 10 <210> 50 <211> 14 <212> PRT <Z13> ARTIFICIAL <220> : : <Z23> + FRHZ VARIANT 21 } 3 <400> 50
Lo Trp val Arg Gin Ala Pro Gly His Gly Leu Glu Trp val Gly 1 5 io : <210> 351 <211> 32 «212» PRT «213» ARTIFICIAL <220> <223> FRH3 VARIANT 136 <400> 51
Arg val Thr Phe Thr ala asp Thr Ser Thr Ser Thr Ala Tyr Met Glu : 1 5 10 15
Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala val Tyr Tyr Cys Ala Arg <210> 52 <211> 32 <212> PRT ( <213> ARTIFICIAL : - <220> <223> FRH3 VARIANT 1 <400> 52 Co arg Ala Thr Phe Thr ala asp Thr Ser Thr Ser Thr Ala Tyr Met Glu 1 5 10 15
Leu Ser ser Leu Arg Ser Glu Asp Thr Ala val Tyr Tyr Cys Ala Arg 20 23 30 <210> 53 ) <211l> 32 : «212» PRT : «213» ARTIFICIAL . ’ <220> <223> FRH3 VARIANT 43 <400> 53
Page 14
Arg val Thr le Thr Ala Asp Thr ser Thr ser Thr ala Tyr Met Glu 1 5 10 15
Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala val Tyr “Tyr Cys Ala arg <210> 54 <211> 32 <212> PRT : <213> ARTIFICIAL <220> «223> FRH3 VARIANT 103 . <400> 54
SU arg Ala Thr Ile Thr Ala asp Thr Ser Thr Ser Thr Ala Tyr Met Glu
Wo 1 5 10 15
Lel Ser ser Leu Arg Ser Giu Asp Thr Ala val Tyr Tyr Cys Ala Arg 20 25 30 <210> 55 : <21l> 11 ‘ <Z12> PRT <213» ARTIFICIAL : <220> <223> FRH4 VARIANT 136 : <400> 355
Trp Gly Gla Gly Thr teu val Thr val Ser ser 1 5 10 «210» 56 <21l> 19 <212> PRT ee «213» Bacteriophage M13mpl8 ! bo — <22{> <221> misc_feature «223» Gene III signal Seguence <400> 56
Met Glu Trp Ser Gly val Phe Met Phe teu Ley Ser val Thr ala Gly 1 5 10 15 val His ser <210> 57 <211> 107 <Z1lZ» PRT <213» ARTIFICIAL <220> <723> LIGHT CHAIN VARIABLE REGION OF CLONE 136 page 15
: <400> 57
Giu Ile val Met Thr Gln Ser Pro ala Thr Leu Ser val Ser Pro Gly 1 5 10 Co 15
Glu Arg Ala Thr Leu Ser Cys Arg Ala ser Gln Ser Ile Gly Thr Asn
Ile His Trp Tyr Gln Gin Lys Pro &ly Gln Ala Pro Arg Leu Ley Ile © 40 45
Tyr Tyr Ale Ser Glu Ser Ile Ser Gly Ile Pro ala Arg Phe Ser Gly 50 55 60 ser Gly Ser Gly Thr Glu Phe Thr Leu Thr Ile Ser Ser Leu Gin Ser so &3 70 75 BO glu asp phe ala val Tyr Tyr Cys Gin Gin Ser Asp Ser Trp Pro Thr 85 8c 95
Thr phe Gly Gly Gly Thr Lys val Glu Ile Lys 100 105 <210> 38 <211> 105 <212> PRT <213> ARTIFICIAL <220> . «223» LIGHT CHAIN CONSTANT REGION OF CLONE 136 0 <400> 58
Thr val ala Ala Pro Ser val Phe Ile Phe Pro Pro Ser Asp Glu Gin 1 5 10 15 i. Leu Lys ser Gly Thr Ala Ser val val Cys Leu Leu Asn Asn Phe Tyr ( . 20 25 30 - Pro Arg Glu Ala.Lys val Gln Trp Lys val Asp Asn Ala Leu ¢lIn Ser 35 40 45
Gly Asn ser Gin Glu Ser val Thr Glu Gin Asp Ser Lys Asp Ser Thr 50 55 60
Tyr Ser Leu Ser Ser Thr Leu The Leu Ser Lys Ala Asp Tyr Glu Lys : £5 70 75 80
His Lys val Tyr Ala Cys Glu val Thr His Gin Gly Leu Ser Ser Pro 85 5 95 val Thr Lys Ser Phe Asn Arg Gly Glu Cys 100 105 <210> 58 : rage 16
<211> 123 <212> PRT : <213> ARTIFICIAL <220> . «223» HEAVY CHAIN VARIABLE REGION OF CLONE 136 ) <400> 59 ;
Gin val an Leu val Gin Ser Gly Ala Glu val Met Lys Pro Gly Ser : i 5 10 15 - ser val Lys val Ser Cys Lys Ala ser Gly Tyr Thr Phe Ser Met Tyr
Trp Leu Glu Trp val Arg Gin Ala Pro Gly His Gly Leu Glu Trp Met } 35 40 45 - Gly Glu Ile Ser Pro Gly Thr Phe Thr Thr Asn Tyr Asn Glu Lys Phe 50 55 60
Lys Ala Arg val Thr phe Thr Ala Asp Thr Ser Thr ser Thr Ala Tyr 65 70 75 80 met Glu Leb Ser Ser Leu Arg Ser Glu Asp Thr Ala val Tyr Tyr Cys 85 90 93 tla Arg Phe Ser His phe Ser Gly Ser Asn Tyr Asp Tyr Phe Asp Tyr 100 185 110
Trp Gly Gln Gly Thr teu val Thr val Ser ser 115 120 ’ «210s 60 . <211l> 3230 <212> PRT . <213> ARTIFICIAL (= or <220> - «2723» CONSTANT REGION OF HUMAN IgGl <400> 60 £la ser Thr Lys Gly Pro Ser val Phe Pro Leu Ala Pro Ser Ser Lys } 1 5 10 15 ser Thr ser Gly Gly Thr Ala Ala Leu Gly Cys Leu val Lys Asp Tyr 20 25 30 pha Pro ¢lu Pro vel Thr val Ser Trp Asn Ser ¢ly Ala Leu Thr Ser ‘ 40 ’ 45
Gly val Mis Thr rhe Pro Ala val Leu Gln Ser Ser Gly teu Tyr Ser 50 55 60
Ley ser ser val val Thr val Pro Ser Ser Ser Let Gly Thr &lin Thr . 65 70 75 80 rage 17
Tyr Ile Cys Asn val Asn His Lys Pro Ser Asn Thr Lys val Asp Lys 85 90 95
Lys val Glu Pro Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys 100 105 110 pro Ala Pro Glu Leu Leu Gly Gly Pro Ser val Phe Leu Phe Pro Pro 115 120 125
Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu val Thr Cvs 130 1.35 140 val val val asp val Ser His Glu Asp Pro Glu val tys Phe Asn Trp
Fa 145 150 155 160
Tyr val Asp Gly val Glu val His Asn Ala Lys Thr Lys Pro Arg Glu 165 17G 175 .
Glu Gin Tyr Asn Ser Thr Tyr Arg val val Ser val Leu Thr val Leu 180 185 190
His Tn Asp Trp Leu Ash Gly Lys Glu Tyr Lys Cys Lys val Ser Asn 185 200 205
Lys aa Leu Pro Ala Pro Ile Glu Lys Thr Ile Ser Lys Ala Lys ¢ly 210 215 220 '
GIn Pro Arg Glu Pro GIn val Tyr Thr Leu Pro Pro Ser Arg Asp Glu 225 © 230 235 ST 240
Leu Thr Lys Asn Gln val Ser Leu Thr Cys Leu val Lys Gly Phe Tyr 245 250 255 . i . te Pro Ser Asp Ile Ala val glu Trp Glu Ser Asn Gly Glin Pro Glu Asn 260 265 270
Asn Tyr Lys Thr Thr Pro Pro val teu Asp Ser Asp Gly Ser Phe Phe 275 280 285
Leu Tyr ser Lys Lau Thr val Asp Lys Ser Arg Trp Gln Gin Gly ASN 290 295 300 val phe Ser Cys Ser val Met Wis Glu Ala Leu His Asn His Tyr Thr 305 310 315 320
Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys 325 550 <210> 61 . «21%» 107 <212» PRT page 18
<213> ARTIFICIAL <220> Co <223> LIGHT CHAIN VARIABLE REGION OF CLONE CL-2C <400> 61
Glu Ile val met Thr Gln Ser Pro Ala Thr Leu Ser val Ser Pro Gly 1 5 10 15
Glu Arg Ala Thr teu Ser Cys Arg Ala Ser Gin Ser Ile Gly Thr Asn
Ile His Trp Tyr Gn Gln Lys Pro Gly Gin Ala Pro Arg Leu Leu Ile 40 45
JE Tyr Tyr Ala Ser Glu Ser Ile ser Gly Ile Pro Ala Arg Phe Ser Gly ( 50 55 60 ser Gly Ser Gly Thr Glu Phe Thr Leu Thr Ile ser ser Lau Gln Ser 65 70 75 80
Glu Asp phe Ala val Tyr Tyr Cys Gin Gin ser Trp Ser Trp Pro Thr : 85 90 95
Thr phe Gly Gly Gly Thr Lys val Glu Ile Lys 100 105 <210> 62 «211» 123 . <212> PRT <Z13> ARTIFICIAL <220> J <223> HEAVY CHAIN VARIABLE REGION OF CLONE CL-~2C <400> 62 ¢ gTn val ¢ln Leu val Gln sar Gly Ala Glu val Met Lys Pro Gly ser
Ss 1 5 1G 15 ser val Lys val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Ser Trp Tyr 20 25 30
Trp Leu Glu Trp val Arg Gin Ala Pro Gly His Gly Leu Glu Trp Met 35 40 45
Gly Glu Ie asp pro Gly Thr Phe Thr Thr Asn Tyr Asn Glu Lys Phe 50 55 80
Lys ala Arg val Thr phe Thr Ala Asp Thr Ser Thr Ser Thr Ala Tyr 65 70 75 - 80
Met Glu Leu Ser Ser Leu Arg Ser Giu Asp Thr ala val Tyr Tyr Cys : 85 ad 95
Page 19
Ala Arg Phe Ser His Phe Ser Gly Ser Asn Tyr Asp Tyr Phe Asp Tyr 100 i05 110 i
Trp Gly 61n Gly Thr Leu val Thr val Ser Ser 115 120 <Z210> 63 <211> 107 . <212> PRT : <Zi3> ARTIFICIAL <220> <223> LIGHT CHAIN VARIABLE REGION OF CLONE CL=-5T <400> 83
Glu Ile val Met Thr Gln Ser Pro Ala Thr Leu Ser val Ser Pro Gly { 1 5 10 15
Glu Arg Ala Thr Leu Ser Cys Arg Ala Ser Glin Ser Ile Gly Thr Ash tle His Trp Tyr Gin 61n Lys Pro Gly ¢In Ala Pro Arg Leu Leu Ile 40 45
Tyr Tyr Ala Ser Glu Ser Ile Ser Gly Ile Pro Ala Arg Phe ser Gly 50 55 60
Ser Gly ser Gly Thr iu Phe Thr Leu Thr Ile Ser Ser Leu Gln Ser 65 70 75 - BO
Glu Asp Phe Ala val Tyr Tyr Cys Gin &ln Ser Trp Ser Trp Pro Thr 85 af a5
Thr phe Gly Gly Gly Thr Lys val Glu Ile Lys 100 105 . a <210> 64 :
Ce <211> 123 <212> PRT <213> ARTIFICIAL . 220» <223> HEAVY CHAIN VARIABLE REGION OF CLONE CL-5T <400> 64 :
Gln val 61n Ley val &ln ser Gly Ala Glu val Met Lys Pro Gly Ser 1 5 10 15 ser val Lys val Ser Cys Lys Ala Ser Gly Tyr Thr phe Ser Met Tyr 20 23 30
Trp Leu Glu Trp val Arg Gin Ala Pro Gly His Gly Leu Glu Trp Met : . 35 40 45
Gly Glu Ile Asp Pro Gly Thr phe Glu Thr Asn Tyr Asn Glu Lys Phe :
Page 20
50 55 60
Lys Ala Arg val Thr Phe Thr Ala Asp Thr Ser Thr Ser Thr Ala Tyr 65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Giu Asp Thr Ala val Tyr Tyr Cys © 85 50 a5 : Alz Arg Phe ser Ris Phe Ser Gly Ser Asn Tyr Asp Tyr Phe Asp Tyr : 100 105 110
Trp Gly Gin Gly Thr Leu val Thr val Ser ser 113 120 0 <2i0> £5
Lo <211> 107 <21l2> PRT <213> ARTIFICIAL <220> <223> LIGHT CHAIN VARIABLE REGION OF CLONE ClL-5A <400> 65 clu The val met Thr Gln Ser Pro Ala Thr Leu Ser val Ser Pro Gly : 1 5 io 15
Glu Arg Ala Thr Lau Ser Cys Arg Ala Ser Gln Ser Ile Gly Thr Asn 23 30
Tle His Trp Tyr ¢1n Gln Lys Pro Gly Gin Ala Pro Arg teu Leu Ile 40 45 :
Tyr Tyr Ala ser Glu.Ser Ile Ser Gly Ile Pro Ala Arg Phe Ser Gly 50 55 60 ‘ : ser gly Ser Gly Thr Glu phe Thr Leu Thr ITe Ser ser teu Glin Ser i 65 70 75 80
Glu Asp Phe ala val Tyr Tyr Cys Gn Gln Ser Trp Ser Trp Pro Thr g5 90 gs thr phe Gly Gly Gly Thr Lys val ¢lu Ile Lys 1006 105 <210> 66 <211> 123 : <212> PRT <213> ARTIFICIAL <220> <Z23 HEAVY CHAIN VARIABLE REGION OF CLONE CL-5A <400> 66
Gin val Gln teu val Gn ser Gly Ala alu val Met Lys Pro Gly Ser 1 5 is rage 21
Ser val Lys val Ser Cys Lys Ala Sar Gly Tyr Thr Phe Ser Trp Tyr
Trp Leu Glu Trp val arg Gn Ala Pro Gly His Gly Leu Glu Trp met 33 40 45
Gly Glu Ile Glu Pro Gly Thr Glu Thr Thr Asn Tyr Asn Glu Lys Phe 50 55 60
Lys Ala Arg val Thr phe Thr ala Asp Thr Ser Thr Sar Thr ala Tyr 63 70 75 80 :
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala val Tyr Tyr Cys pe 85 50 as
Ala Arg Phe Ser His phe Ser Gly Ser Asn Tyr Asp Tyr Phe Asp Tyr 100 105 110
Trp Gly GIn Gly Thr Leu val Thr val Ser Ser : 115 120 <210> 67 <211> 107 <21l2> PRT <213> ARTIFICIAL <22{(> ’ «223» LIGHT CHAIN VARIABLE REGION OF CLONE CL-2B : <400> 67
Glu Ile val Met Thr &In Ser Pro Ala Thr Leu Ser val ser Pro Gly 1 5 10 15
Glu Arg Ala Thr teu Ser Cys Arg Ala Ser Gln Ser Ile &ly Thr Asn i 20 25 30
Ile His Trp Tyr Gin Gin Lvs Pro Giy Gln Ala Pro Arg Leu Ley Ile 40 45
Tyr Tyr ala ser Glu Ser Ile Ser Gly Ile Pro Ala Arg Phe Ser Gly 50 55 60 + Ser &ly Ser Gly Thr Glu phe Thr Leu Thr Ile Ser Ser Leu Glin Ser 65 70 75. 80
Glu Asp Phe Ala val Tyr Tyr cys Gin Gin Ser Trp Ser Trp Pro Thr 85 a0 a5
Thr phe Gly Gly Gly Thr Lvs val clu Ie Lys 100 105 <210> 68 oo
Page 22 Co
<211l> 123 <212> PRT <213> ARTIFICIAL . <220> . <223> HEAVY CHAIN VARIABLE REGION OF CLONE CL-2B . } <400> 68
Gln val Gin Leu val Gln ser aly Ala Glu val Met Lys Pro Gly ser 1 5 10 is ser val Lys val Ser Cys Lys Ala Ser &ly Tyr Thr Phe Ser Tyr Tyr : 20 25 30
Trp Leu Glu Trp val Arg Gln Ala Pro Gly His Gly Leu Glu Trp Met 35 40 45
Co
Gly Glu Ile Asp Pro Gly Thr Phe Thr Thr asn Tyr Asn Glu Lys phe 50 55 60
Lys Ala arg val Thr phe Thr Ala Asp Thr Ser Thr ser Thr Ala Tyr £5 7G 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala val Tyr Tyr Cys 85 80 a5
Ale Arg Phe Ser His Phe Ser Gly Ser Asn Tyr Asp Tyr Phe Asp Tyr 100 105 110
Tre Gly Gla Gly Thr Leu val thr val ser Ser 115 120 «210» 68 <li> 107 <21l2> PRT
N <213> ARTIFICIAL { <Z220> si «223 LIGHT CHAIN VARIABLE REGION OF CLONE CL-1136-2C <400= G2
Glu Tle val met Thr Gin Ser Pro Ala Thr Leu Ser val Ser Pro Gly 1 5 10 15
Glu arg Ala Thr Leu Ser Cys Arg Ala Ser Gin Ser Ile Gly Thr Asn
Ile Wis Trp Tyr Glin Gln Lys Pro Gly Gin Ala Pro Arg Leu Leu Ile 40 45
Tyr Tyr Ala Ser Glu ser Ile Ser Gly Ile Pro Ala Arg Phe ser ely 50 55 50 sar Gly ser Gly Thr Glu Phe Thr Leu Thr Ile Ser Ser Leu Gin Ser 65 70 75 80 page 23
Glu Asp Phe Ala val Tyr Tyr Cys Gin Gin Ser Trp Ser Trp Pro Thr 85 9G a5
Thr Phe Gly Gly Gly Thr Lys val Glu Ile Lys 106 Taos «21> 70 «211s 123 : : «212» PRT <213> ARTIFICIAL <220> <223> HEAVY CHAIN VARIABLE REGION OF CLONE CL-1136-2C <400> 70 : o Gln val &in Leu val Glin ser Gly Ala Glu val Lys Lys Pro Gly Ser oe 1 5 it 15 ser val Lys val Ser Cys Lys ala Ser Gly Tyr Thr Phe Ser Met Tyr
Trp Leu Glu Trp val Arg Gln Ala Pro Gly Gln GIy Leu Glu Trp Met 40 45
Gly Glu Ile ser Pro Gly Thr Phe Thr Thr asn Tyr asn Glu Lys Phe 50 55 60
Lys ala Arg val Thr Phe Thr Ala Asp Thr Ser Thr Ser Thr Ala Tyr 65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala val Tyr Tyr Cys : 85 80 C85
Ala Arg Phe Sar His Phe Ser Gly Sar Asn Tyr Asp Tyr Phe Asp Tyr
FU 100 105 110 or Trp G1y Gin Gly Thr Leu val Thr val ser ser 115 120 <2L0> 71 <211> 108 «212» PRT <213> Homo sapiens «400» 71
Asp Ser Asn Pro Arg Gly val ser Ala Tyr Leu Ser Arg Pro Ser Pro 1 5 - 30 15
Phe Asp Leu Phe Ile Arg Lys Ser pro Thr Ile Thr cys Leu val val 20 oo 25 30
Asp Leu Ala Pro Ser Lys Gly Thr val Asn Leu Thr Trp Sar Arg Ala 35 40 45
Page 24 .
ser Gly Lys Pro val Asn His Ser Thr Arg Lys Glu Glu Lys Gin Arg : 50 55 80 asn Gly Thr Leu Thr val Thr ser Thr Leu Pro val Gly Thr Arg Asp
CEs - 70 75 BO
Trp Ile ¢lu Gly Glu Thr Tyr in Cys Arg val Thr #is Pro His Leu 85 20 85
Pro Arg Ala Leu Met Arg Ser Thr Thr Lys Thr Ser 100 105 <210- 72 a <21l> 12 { <212» PRT
See” <213> ARTIFICIAL ’ ‘ <220> «223» PEPTIDE DERIVED FROM CH3 DOMAIN OF IGE <220> } <221> misc_feature <222> (73..(7) . <223> Xaa can be any naturally occurring amino acid <220> ) <221> misc_ feature «222» (9)..(10) , . <273» Xaa can be any naturally occurring amine acid . <220> <221> wmisc_feature <222> (12)..(12) } . i <273> Maa can be any naturally occurring amine acid <400> 72 ash Pro Arg Gly val Ser Xaa Tyr Xaa Xaa Arg Xaa a 1 5 10 or <210> 73 «211s 12 «212» PRT <213> ARTIFICIAL : <220> <223> PEPTIDE DERIVED FROM CH3 DOMAIN OF IGE <4Q0> 73
Asn Pro Arg Gly val Ser Ala Tyr Leu Ser Arg Pro : 1 5 10 <210> 74 <211> © <212» PRT <213> ARTIFICIAL <220> <273> PEPTIDE DERIVED FROM CHI DOMAIN OF IGE
Page 25
<220> . <221> ‘misc_Teature : - <222> {(B)..(&) . . . <7» Xas can be any naturally occurring amino acid «220 . . <221> misc.feature <222> (82..(9) , . . <733» Xaz can be any naturally occurring amine acid <400> 74
Leu Pro Arg Ala Leu Xaa Arg ser Xaa 1 5 <210> 75 2 <211> 8 :
Lo <212> PRT «213% ARTIFICIAL : <220> <273> PEPTIDE DERIVED FROM CHZ DOMAIN OF IGE <400> 75 =
Leu Pro Arg Ala Leu Met Arg Ser Thr 1 . . <210> 76 «211» 12 . <212> PRT <213> ARTIFICIAL : <220> ‘ <223> PEPTIDE DERIVED FROM CHI DOMAIN OF IGE <400> 76 wis Pro His Leu Pro Arg Ala Leu Mel Arg Ser Thr . i 5 10 be 210» 77 = «211s 12 «212» PRT <Z13> ARTIFICIAL } : <220> <223> PEPTIDE DERIVED FROM CH? DOMAIN OF IGE . <4Q0> 77 oC
Ley pro Arg Ala Leu Met Arg Ser Thr Tor Lys Thr i 2 4 . page 26

Claims (21)

We Claim:
1. An isolated peptide consisting essentially of the amine acid sequence: Asn Pro Arg Gly Val Ser Xaa Tyr Xaa Xaa Arg Xaa (SEQ ID NO. 72).
2. The amino acid of claim 1, wherein the amino acid sequence is: Asn Pro Arg Gly Val Ser Ala Tyr Leu Ser Arg Pro (SEQ ID NO. 73).
3. An isolated peptide consisting essentially of the amino acid sequence: Leu Pro Arg Ala Leu Xaa Arg Ser Xaa (SEQ ID NO: 74).
4, The peptide of claim 3, wherein the amino acid sequence is: a) Leu Pro Arg Ala Leu Met Arg Ser Thr (SEQ ID NO: 75) Bb) Hi Pro His Leu Pro Arg Ala Leu Met Arg Ser Thr (SEQ ID NO: 76); or C) Leu Pro Arg Ala Leu Met Arg Ser Thr Thr Lys Thr (SEQ ID NO:77).
5. A composition comprising the peptide of claim 1 or claim 3 and a physiologically acceptable carrier, diluent, stabilizer or excipient.
B. The composition of claim 5, further comprising an immunogenic carrier.
7. The composition of claim 8, wherein the immunogenic carrier is selected from the group consisting of BSA, KLH, tetanus toxoid, and diphtheria toxoid.
:
8. An isolated antibody that specifically binds to the peptide of any one of claims 1 to 4.
9, The antibody of claim 8, further comprising a label.
10. The antibody of claim 8, wherein the antibody is: a) a chimeric antibody, b) a single chain antibody, Cc) a Fab fragment, d) a F(ab"), fragment, e) a human antibody, or f) a humanized antibody.
11. A composition comprising an antibody of claim 8 and an acceptable cartier.
12. A method preparing a polyclonal antibody, the method comprising: a) immunizing an animal with a polypeptide consisting of an amino acid sequence of SEQ ID NO:72 or SEQ ID NO:74 under conditions to elicit an . antibody response, hb) isolating antibodies from the animal, and BE ¢) screening the isolated antibodies with the polypeptide, thereby identifying a polyclonal antibody which specifically binds with high affinity to a polypeptide comprising an amino acid sequence of SEQ 1D NO:72 or SEQ 1D NO:74.
13. A polyclonal antibody produced by a method of claim 12.
14. A composition comprising the polyclonal antibody of claim 13 and a suitable carrier.
15. A method of making a monocional antibody, the method comprising: a) immunizing an animal with a polypeptide consisting essentially of an amino acid sequence of SEQ ID NO:72 or SEQ ID NO:74 under conditions to elicit an antibody response, ’ b) isolating antibody producing cells from the animal, c) fusing the antibody producing cells with immortalized cells to form monoclonal antibody-producing hybridoma cells, d) culturing the hybridoma cells, and e) isolating from the culture a monoclonal antibody which specifically binds with high affinity to a polypeptide comprising an amino acid sequence of SEQ ID NO:72 or SEQ ID NO:74.
16. A monoclonal antibody produced by a method of claim 15.
17. A composition comprising the monoclonal antibody of claim 16 and a suitable carrier.
18. The antibody of claim 8, wherein the antibody is produced by screening a Fab expression library.
19. The antibody of claim 8, wherein the antibody is produced by screening a combinatorial immunogiobulin library.
20. A kit comprising the antibody of any one of claims 8-19.
21. A method of inducing an immunological response to IgE in a mammal comprising administering the peptide or the composition of any one of claims 1-7, in an amount sufficient fo induce a response in said mammal.
SG2012057212A 2004-02-02 2004-07-29 Identification of novel ige epitopes SG183683A1 (en)

Applications Claiming Priority (2)

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

Publications (1)

Publication Number Publication Date
SG183683A1 true SG183683A1 (en) 2012-09-27

Family

ID=34841291

Family Applications (2)

Application Number Title Priority Date Filing Date
SG2012057212A SG183683A1 (en) 2004-02-02 2004-07-29 Identification of novel ige epitopes
SG200900706-3A SG149892A1 (en) 2004-02-02 2004-07-29 Identification of novel ige epitopes

Family Applications After (1)

Application Number Title Priority Date Filing Date
SG200900706-3A SG149892A1 (en) 2004-02-02 2004-07-29 Identification of novel ige epitopes

Country Status (8)

Country Link
EP (1) EP1718669A4 (en)
JP (1) JP2008507474A (en)
KR (4) KR101365375B1 (en)
CN (1) CN102702359A (en)
AU (2) AU2004315197B2 (en)
CA (1) CA2552999A1 (en)
SG (2) SG183683A1 (en)
WO (1) WO2005075504A1 (en)

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008019131A2 (en) * 2006-08-04 2008-02-14 The Trustees Of The University Of Pennsylvania Methods and compositions for treating ige-mediated diseases
US7767206B2 (en) * 2006-10-02 2010-08-03 Amgen Inc. Neutralizing determinants of IL-17 Receptor A and antibodies that bind thereto
JP2010519194A (en) * 2007-02-15 2010-06-03 アストラゼネカ・アクチエボラーグ Binding elements for IgE molecules
AR065368A1 (en) * 2007-02-15 2009-06-03 Astrazeneca Ab ANTIBODIES FOR IGE MOLECULES
EP2012122A1 (en) * 2007-07-06 2009-01-07 Medigene AG Mutated parvovirus structural proteins as vaccines
CL2008003808A1 (en) * 2007-12-21 2011-03-11 Glaxosmithkline Biologicals Sa Malaria vaccine component comprising an rts, s and / or an immunogenic particle that is derived from the cs protein of one or more strains of p. vivax and hepatitis b antigen s or an immunogenic particle comprising rts, csv-s and optionally the unbound antigen s and a stabilizing agent; and vaccine.
SG10201400388QA (en) 2008-12-09 2014-05-29 Pfizer Vaccines Llc IgE CH3 peptide vaccine
EP2942061A3 (en) 2010-06-07 2016-01-13 Pfizer Vaccines LLC Ige ch3 peptide vaccine
WO2018129248A1 (en) 2017-01-06 2018-07-12 The Regents Of The University Of California Therapeutic anti-ige antibodies and methods and compositions thereof
CN112203724A (en) * 2018-03-26 2021-01-08 诺华股份有限公司 Methods of treating chronic idiopathic urticaria using Rigeuzumab
EP4355362A1 (en) 2021-06-14 2024-04-24 Novartis AG Pharmaceutical formulation containing an anti-ige antibody

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS63225397A (en) * 1986-10-03 1988-09-20 Dainippon Pharmaceut Co Ltd Novel peptide having antiallergic action
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
DE69333484T2 (en) * 1992-09-24 2005-03-24 Novartis Ag Reshaped monoclonal antibodies against an immunoglobulin isotype
US6172213B1 (en) * 1997-07-02 2001-01-09 Genentech, Inc. Anti-IgE antibodies and method of improving polypeptides
EP0957111B1 (en) * 1998-04-09 2004-12-29 Idexx Laboratories, Inc. Specific binding proteins for treating canine allergy
US7393529B2 (en) * 1998-04-09 2008-07-01 Idexx Laboratories, Inc. Methods and compositions for inhibiting binding of IgE to a high affinity receptor
TWI227241B (en) * 1998-06-20 2005-02-01 United Biomedical Inc IgE-CH3 domain antigen peptide, peptide conjugate containing the same, and pharmaceutical composition for treating allergies containing the peptide conjugate
GB9822763D0 (en) * 1998-10-20 1998-12-16 Univ Sheffield Immunoglobin variant
KR20020007314A (en) * 1999-02-25 2002-01-26 장 스테판느 Epitopes or mimotopes derived from the c-epsilon-3 or c-epsilon-4 domains of ige, antagonists thereof, and their therapeutic uses
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 (en) * 2001-05-22 2003-02-18 Pfizer Prod Inc IgE VACCINE FREE FROM ANAPHYLAXIS-INDUCING PROPERTY
GB0209878D0 (en) * 2002-04-30 2002-06-05 Glaxosmithkline Biolog Sa Vaccine
ES2566778T3 (en) * 2003-02-01 2016-04-15 Tanox, Inc. High affinity anti-human IgE antibodies

Also Published As

Publication number Publication date
AU2004315197A1 (en) 2005-08-18
AU2009202408A1 (en) 2009-07-09
AU2004315197B2 (en) 2009-06-04
KR20070008578A (en) 2007-01-17
KR101581659B1 (en) 2015-12-31
EP1718669A4 (en) 2007-10-10
WO2005075504A1 (en) 2005-08-18
KR20130088198A (en) 2013-08-07
KR101365375B1 (en) 2014-02-19
JP2008507474A (en) 2008-03-13
SG149892A1 (en) 2009-02-27
KR101562114B1 (en) 2015-11-20
CA2552999A1 (en) 2005-08-18
KR20140021058A (en) 2014-02-19
CN102702359A (en) 2012-10-03
EP1718669A1 (en) 2006-11-08
KR20120056297A (en) 2012-06-01

Similar Documents

Publication Publication Date Title
AU2004209638B2 (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 (en) IDENTIFICATION OF NOVEL IgE EPITOPES