US20040096956A1 - Method for generating libraries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules - Google Patents

Method for generating libraries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules Download PDF

Info

Publication number
US20040096956A1
US20040096956A1 US10/690,396 US69039603A US2004096956A1 US 20040096956 A1 US20040096956 A1 US 20040096956A1 US 69039603 A US69039603 A US 69039603A US 2004096956 A1 US2004096956 A1 US 2004096956A1
Authority
US
United States
Prior art keywords
antibody
antigen
dna
cells
cell
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US10/690,396
Inventor
Michael Wigler
Joseph Sorge
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Stratagene California
Catalyst Assets LLC
Original Assignee
Stratagene California
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 US07/919,730 external-priority patent/US5284555A/en
Priority claimed from US08/315,269 external-priority patent/US5780225A/en
Application filed by Stratagene California filed Critical Stratagene California
Priority to US10/690,396 priority Critical patent/US20040096956A1/en
Assigned to STRATAGENE reassignment STRATAGENE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SORGE, JOSEPH A., WIGLER, MICHAEL H.
Publication of US20040096956A1 publication Critical patent/US20040096956A1/en
Assigned to CATALYST ASSETS LLC reassignment CATALYST ASSETS LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: AGILENT TECHNOLOGIES RESEARCH CORP., AGILENT TECHNOLOGIES, INC., MOLECULAR AFFINITIES CORP., SORGE, JOSEPH A., WIGLER, MICHAEL H.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/102Mutagenizing nucleic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • Monoclonal and polyclonal antibodies are useful for a variety of purposes.
  • the precise antigen specificity of antibodies makes them powerful tools that can be used for the detection, quantitation, purification and neutralization of antigens.
  • Polyclonal antibodies are produced in vivo by immunizing animals, such as rabbits and goats, with antigens, bleeding the animals and isolating polyclonal antibody molecules from the blood.
  • Monoclonal antibodies are produced by hybridoma cells, which are made by fusing, in vitro, immortal plasmacytoma cells with antibody producing cells (Kohler, G. and C. Milstein, Nature, 256:495 (1975)) obtained from animals immunized in vivo with antigen.
  • a method of producing antibodies which avoids the limitations of presently-available methods, such as the requirement for immunization of an animal and in vivo steps, would be very useful, particularly if it made it possible to produce a wider range of antibody types than can be made using presently-available techniques and if it made it possible to produce human antibody types.
  • the present invention relates to a method of producing libraries of genes encoding antigen-combining molecules or antibodies; a method of producing-antigen-combining molecules, also referred to as antibodies, which does not require an in vivo procedure, as is required by presently-available methods; a method of obtaining antigen-combining molecules (antibodies) of selected or defined specificity which does not require an in vivo procedure; vectors useful in the present method and antibodies produced or obtained by the method.
  • the present invention relates to an in vitro process for synthesizing DNA encoding families of antigen-combining molecules or proteins.
  • DNA containing genes encoding antigen-combining molecules is obtained and combined with oligonucleotides which are homologous to regions of the genes which are conserved.
  • Sequence-specific gene amplification is then carried out using the DNA containing genes encoding antigen-combining proteins as template and the homologous oligonucleotides as primers.
  • This invention also relates to a method of creating is diverse libraries of DNAs encoding families of antigen-combining proteins by cloning the product of the in vitro process for synthesizing DNA, described in the preceeding paragraph, into an appropriate vector (e.g., a plasmid, viral or retroviral vector).
  • an appropriate vector e.g., a plasmid, viral or retroviral vector.
  • the subject invention provides an alternative method for the production of antigen-combining molecules, which are useful affinity reagents for the detection and neutralization of antigens and the delivery of molecules to antigenic sites.
  • the claimed method differs from production of polyclonal antibody molecules derived by immunization of live animals and from production of mono-clonal antibody molecules through the use of hybridoma cell lines in that it does not require an in vivo immunization step, as do presently available methods. Rather, diverse libraries of genes which encode-antigen-combining sites comprising a significant proportion of an animal's repertoire of antibody combining sites are made, as described in detail herein. These genes are expressed in living cells, from which molecules of desired antigenic selectivity can be isolated and purified for various uses.
  • Antigen-combining molecules are produced by the present method in the following manner, which is described in greater deail below. Initially, a library of antibody genes which includes a set of variable regions encoding a large, diverse and random group of specificities derived from animal or human immunoglobulins is produced by amplifying or cloning diverse genomic fragments or cDNAs of antibody mRNAs found in antibody-producing tissue.
  • the diversity of the resulting libraries can be increased by means of random mutagenesis.
  • the gene libraries are introduced into cultured host cells, which may be eukaryotic or prokaryotic, in which they are expressed. Genes encoding antibodies of desired antigenic specificity are identified, using a method described herein or known techniques, isolated and expressed in quantities in appropriate host cells, from which the encoded antibody can be purified.
  • a library of genes encoding immunoglobulin heavy chain regions and a library of genes encoding immunoglobulin light chain regions are constructed. This is carried out by obtaining antibody-encoding DNA, which is either genomic fragments or cDNAs of antibody mRNAs, amplfying or cloning the fragments or cDNAs; and introducing them into a standard framework antibody gene vector, which is used to introduce the antibody-encoding DNA into cells in which the DNA is expressed.
  • the vector includes a framework gene encoding a protein, such as a gene encoding an antibody heavy chain or an antibody light chain which can be of any origin (human, non-human) and can be derived from any of a number of existing DNAs encoding heavy chain immuno-globulins or light chain immunoglobulins.
  • a framework gene encoding a protein, such as a gene encoding an antibody heavy chain or an antibody light chain which can be of any origin (human, non-human) and can be derived from any of a number of existing DNAs encoding heavy chain immuno-globulins or light chain immunoglobulins.
  • Such vectors are also a subject of the present invention and are described in greater detail in a subsequent section.
  • Genes from one or both of the libraries are introduced into appropriate host cells, in which the genes are expressed, resulting in production of a wide variety of antigen-combining molecules.
  • Genes encoding antigen-combining molecules of desired specificity are identified by identifying cells producing antigen-combining molecules which react with a selected antigen and then obtaining the genes of interest.
  • the genes of interest can subsequently be introduced into an appropriate host cell (or can be further modified and then introduced into an appropriate host cell) for further production of antigen-combining molecules, which can be purified and used for the same purposes for which conventionally-produced antibodies are used.
  • antigen-combining molecules which are of wider diversity than are antibodies available as a result of known methods; novel antigen-combining molecules with a diverse range of specificities and affinities and antigen-combining molecules which are predominantly human in origin.
  • antigen-combining molecules are a subject of the present invention and can be used clinically for diagnostic, therapeutic and prophylactic purposes, as well as in research contexts, and for other purposes.
  • FIG. 1 is a schematic representation of the method of the present invention by which antigen-combining molecules, or antibodies, are produced.
  • FIG. 2 is a schematic representation of amplification or cloning of IgM heavy chain variable region DNA from mRNA, using the polymerase chain reaction.
  • Panel A shows the relevant regions of the poly adenylated mRNA encoding the secreted form of the IgM heavy chain.
  • S denotes the sequences encoding the signal peptide which causes the nascent peptide to cross the plasma membrane.
  • V, D and J together comprise the variable region.
  • C H 1, C H 2, and C H 3 are the three constant domains of C ⁇ . Hinge encodes the hinge region.
  • C, B and Z are oligonucleotide PCR primers (discussed below).
  • Panel B shows the reverse transcript DNA product of the mRNA primed by oligonucleotide Z, with the addition of poly-dC by terminal transferase at the 3′ end.
  • Panel C is a schematic representation of the annealing of primer A to the reverse transcript DNA.
  • Panel D shows the final double stranded DNA PCR product made utilizing primers A and B.
  • Panel E shows the product of PCR annealed to primer C.
  • Panel F is a blowup of Panel E, showing in greater detail the structure of primer C.
  • Primer C consists of two parts: a 3′ part complementary to IgM heavy chain mRNA as shown, and a 5′ part which contains restriction site RE2 and spacer.
  • Panel G shows the final double stranded DNA PCR product made utilizing primers A and C and the product of the previous PCR (depicted in D) as template. The S, V, D, J regions are again depicted.
  • FIG. 3 is a schematic representation of the heavy chain framework vector pFHC.
  • the circular plasmid (above) is depicted linearized (below) and its relevant components are shown: animal cell antibiotic resistance marker; bacterial replication origin; bacterial cell antibiotic resistance marker; C ⁇ enhancer; LTR containing the viral promoter from the Moloney MLV retrovirus DNA; PCR primer (D); cDNA cloning site containing restriction endonuclease sites, RE1 and RE2, separated by spacer DNA; C ⁇ exons; and poly A addition and termination sequences derived from the C ⁇ gene or having the same sequence as the C ⁇ gene.
  • FIG. 4 depicts a nucleotide sequence of the C H 1 exon of the C ⁇ gene, and its encoded amino acid sequence (Panel A). The nucleotide coordinate numbers are listed above the line of nucleotide sequences. Panel B depicts the N-doped sequence, as defined in the text.
  • the present invention provides a method of producing antigen-combining molecules (or antibodies) which does not require an in vivo immunization procedure and which makes it possible to produce antigen-combining molecules with far greater diversity than is shown by antibodies produced by currently-available techniques.
  • the present invention relates to a method of producing libraries of genes encoding antigen-combining molecules (antibody proteins) with diverse antigen-combining specificities; a method of producing such antigen-combining molecules, antigen-combining molecules produced by the method and vectors useful in the method.
  • the following is a description of generation of such libraries, of the present method of producing antigen-combining molecules of selected specificity and of vectors useful in producing antigen-combining molecules of the present invention.
  • the process makes use of techniques which are known to those of skill in the art and can be applied as described herein to produce and identify antigen-combining molecules of desired antigenic specificity: the polymerase chain reaction (PCR), to amplify and clone diverse cDNAs encoding antibody mRNAs found in antibody-producing tissue; mutagenesis protocols to further increase the diversity of these cDNAs; gene transfer protocols to introduce antibody genes into cultured (prokaryotic and eukaryotic) cells for the purpose of expressing them; and screening protocols to detect genes encoding antibodies of the desired antigenic specificity.
  • PCR polymerase chain reaction
  • FIG. 1 A general outline of the present method is represented in FIG. 1.
  • a key step in the production of antigen-combining molecules by the present method is the construction of a “library” of antibody genes which include “variable” regions encoding a large, diverse, but random set of specificities.
  • the library can be of human or non-human origin and is constructed as follows:
  • genomic DNA encoding antibodies or cDNAs of antibody mRNA is obtained.
  • This DNA can be obtained from any source of antibody-producing cells, such as spleen cells, peripheral blood cells, lymph nodes, inflammatory tissue cells and bone marrow cells. It can also be obtained from a genomic library or cDNA library of B cells.
  • the antibody-producing cells can be of human or non-human origin; genomic DNA or mRNA can be obtained directly from the tissue (i.e., without previous treatment to remove cells which do not produce antibody) or can be obtained after the tissue has been treated to increase concentration of antibody-producing cells or to select a particular type(s) of antibody-producing cells (i.e., treated to enrich the content of antibody-producing cells).
  • Antibody-producing cells can be stimulated by an agent which stimulates antibody mRNA production (e.g., lipopolysaccharide) before DNA is obtained.
  • Antibody-encoding DNA is amplified and cloned using a known technique, such as the PCR using appropriately-selected primers, in order to produce sufficient quantities of the DNA and to modify the DNA in such a manner (e.g., by addition of appropriate restriction sites) that it can be introduced as an insert into an E. coli cloning vector.
  • This cloning vector can serve as the expression vector or the inserts can later be introduced into an expression vector, such as the framework antibody gene vector described below.
  • Amplified and cloned DNA can be further diversified, using mutagenesis, such as PCR, in order to produce a greater diversity or wider repertoire of antigen-binding molecules, as well as novel antigen-binding molecules.
  • Cloned antibody-encoding DNA is introduced into an expression vector, such as the framework antibody gene vector of the present invention, which can be a plasmid, viral or retroviral vector. Cloned antibody-encoding DNA is inserted into the vector in such a manner that the cloned DNA will be expressed as protein in appropriate host cells. It is essential that the expression vector used make it possible for the DNA insert to be expressed as a protein in the host cell.
  • One expression vector useful in the present method is referred to as the framework antibody gene vector.
  • Vectors useful in the present method contain antibody constant region or portions thereof in such a manner that when amplified DNA is inserted, the vector expresses a chimeric gene product comprising a variable region and a constant region in proper register. The two regions present in the chimeric gene product can be from the same type of immunoglobulin molecule or from two different types of immunoglobulin molecules.
  • These libraries of antibody-encoding genes are then expressed in cultured cells, which can be eukaryotic or prokaryotic.
  • the libraries can be introduced into host cells separately or together. Introduction of the antibody-encoding DNA in vitro into host cells (by infection, transformation or transfection) is carried out using known techniques, such as electroporation, protoplast fusion or calcium phosphate co-precipitation. If only one library is introduced into a host cell, the host cell will generally be one which makes the other antibody chain, thus making it possible to produce complete/functional antigen-binding molecules. For example, if a heavy chain library produced by the present method is introduced into host cells, the host cells will generally be cultured cells, such as myeloma cells or E. coli, which naturally produce the other (i.e., light) chain of the immunoglobulin or are engineered to do so. Alternatively, both libraries can be introduced into appropriate host cells, either simultaneously or sequentially.
  • Host cells in which the antibody-encoding DNA is expressed can be eukaryotic or prokaryotic. They can be immortalized cultured animal cells, such as a myeloma cell line which has been shown to efficiently express and secrete introduced immunoglobulin genes (Morrison, S. L. et al., Ann. N.Y. Acad. Sci., 507:187 (1987); Kohler, G. and C. Milstein, Eur. J. Immunol., 6:511 (1976); Oi, V. T., et al., Immunoglobulin Gene Expression in Transformed Lymphoid Cells, 80:825 (1983); Davis, A. C. and M. J. Shulman, Immunol. Today, 10:119 (1989)).
  • One host cell which can be used to express the antibody-encoding DNA is the J558L cell line or the SP2/0 cell line.
  • Cells expressing antigen-combining molecules with a desired specificity for a given antigen can then be selected by a variety of means, such as testing for reactivity with a selected antigen using nitrocellulose layering.
  • the antibodies identified thereby can be of human origin, nonhuman origin or a combination of both. That is, all or some of the components (e.g., heavy chain, light chain, variable regions, constant regions) can be encoded by DNA of human or nonhuman origin, which, when expressed produces the encoded chimeric protein which, in turn, may be human, nonhuman or a combination of both.
  • all or some of the regions are referred to as being of human origin or of nonhuman origin, based on the source of the DNA encoding the antigen-combining molecule region in question.
  • the resulting antigen-combining molecule has a heavy chain variable region of mouse origin.
  • Antibodies produced may be used for such purposes as drug delivery, tumor imaging and other therapeutic, diagnostic and prophylactic uses.
  • antibodies of a desired binding specificity are obtained, their genes may be isolated and further mutagenized to create additional antigen combining diversity or antibodies of higher affinity for antigen.
  • Cells expressing antibody reactive to antigen are identified by a nitrocellulose filter overlay and antibody is prepared from cells identified as expressing it.
  • nitrocellulose filter overlay As described in a subsequent section, there are alternative methods of library construction, other expression systems which can be used, and alternative selection systems for identifying antibody-producing cells or viruses.
  • Step 1 in this specific protocol is construction of libraries of genes in E. coli which encode immunoglobulin heavy chains. This is followed by the use of random mutagenesis to increase the diversity of the library, which is an optional procedure.
  • Step 2 is introduction of the library, by transfection, into myeloma cells.
  • Step 3 is identification of myeloma cells expressing antibody with the desired specificity, using the nitrocellulose filter overlay technique or techniques known to those of skill in the art.
  • Step 4 is isolation of the gene(s) encoding the antibody with the desired specificity and their expression in appropriate host cells, to produce antigen-combining fragments useful for a variety of purposes.
  • pFHC E. coli plasmid vector
  • pFHC contains a “framework” gene, which can be any antibody heavy chain and serves as a site into which the amplified cloned gene product (genomic DNA or cDNA of antibody mRNAs) is introduced.
  • pFHC is useful as a vector for this purpose because it contains RE1 and RE2 cloning sites.
  • Other vectors which include a framework gene and other cloning sites can be used for this purpose as well.
  • the framework gene includes a transcriptional promoter (e.g., a powerful promoter, such as a Moloney LTR (Mulligan, R. C., In Experimental Manipulation of Gene Expression, New York Adacemic Press, p. 155 (1983)) and a C ⁇ chain transcriptional enhancer to increase the level of transcriptions from the promoter (Gillies, S. D., et al., Cell, 33:717 (1983), a cloning site containing RE1 and RE2; part of the C ⁇ heavy chain gene encoding secreted protein; and poly A addition and termination sequences (FIG. 3).
  • a transcriptional promoter e.g., a powerful promoter, such as a Moloney LTR (Mulligan, R. C., In Experimental Manipulation of Gene Expression, New York Adacemic Press, p. 155 (1983)
  • a C ⁇ chain transcriptional enhancer to increase the level of transcriptions from the promoter
  • the framework antibody gene vector of the present invention also includes a selectable marker (e.g., an antibiotic resistance gene such as the neomycin resistance gene, neo R ) for animal cells; sequences for bacterial replication (ori); and a selectable marker (e.g., the ampicillin resistance gene, Amp R ) for bacterial cells.
  • the framework gene can be of any origin (human, non-human), and can derive from any one of a number of existing DNAs encoding heavy chain immunoglobulins (Tucker, P. W., et al., Science, 206:1299 (1979); Honjo, T., et al., Cell, 18:559 (1979); Bothwell, A. L.
  • the vector retains the introns between the C H 1, hinge, C H 2 and C H 3 exons.
  • the “variable region” of the gene which includes the V, D and J regions of the antibody heavy chain and which encodes the antigen binding site, is deleted and replaced with two consecutive restriction endonuclease cloning sites, RE1 and RE2.
  • the restriction endonuclease site RE1 occurs just 3′ to the LTR promoter and the restriction endonuclease site RE2 occurs within the constant region just 3′ to the J region (see FIG. 3).
  • Another key step in the production of antigen-combining molecules in this embodiment of the present invention is construction in an E. coli vector of a library of cDNAs encoding the variable region of mouse immunoglobulin genes.
  • the pFHC vector which includes cloning sites designated RE1 and RE2, is used for cloning heavy chain variable regions, although any cloning vector with cloning sites having the same or similar characteristics (described below) can be used.
  • a light chain vector can be designed, using the above described procedures and procedures known to a person of ordinary skill in the art.
  • non-immune mouse spleens are used as the starting material.
  • mRNA is prepared directly from the spleen or from spleen processed in such a manner that it is enriched for resting B cells. Enrichment of tissue results in a more uniform representation of antibody diversity in the starting materials.
  • Lymphocytes can be purified from spleen using ficoll gradients (Boyum, A., Scand. J. of Clinical Invest., 21:77 (1968)). B cells are separated from other cells (e.g., T cells) by panning with anti-IgM coated dishes (Wysocki , L. J. and V. L. Sato, Proc. Natl. Acad. Sci.
  • Poly A+ mRNA from total mouse spleen is prepared according to published methods (Sambrook, J., et al., Molecular Cloning: A Laboratory Manual, 2d Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)). Production of antibody mRNA can first be stimulated by lipopolysaccharide (LPS) (Andersson, J. A., et al., J. Exp. Med., 145:1511 (1977)). First strand cDNA is prepared to this mRNA population using as primer an oligonucleotide, Z, which is complementary to C ⁇ in the C H 1 region 3′ to J. This primer is designated Z in FIG. 2.
  • LPS lipopolysaccharide
  • First strand cDNA is then elongated by the terminal transferase reaction with dCTP to form a poly dC tail (Sambrook, J., et al., Molecular Cloning: A Laboratory Manual, 2d Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)).
  • PCR polymerase chain reaction
  • Primer B is complementary to all C ⁇ genes, which encode the heavy chain of molecules of the IgM class, the Ig class expressed by all B cell clones prior to class switching (Schimizu, A. and T. Honjo, Cell, 36:801-803 (1984)) and present in resting B cells.
  • the resultant PCR product includes a significant proportion of cDNAs encompassing the various V H regions expressed as IgM in the mouse. (The use of other primers complementary to the cDNA genes encoding the constant regions of other immunoglobulin heavy chains can be used in parallel reactions to obtain the variable regions expressed on these molecules, but for simplicity these are not described).
  • Primer C like primer B, is complementary to the C ⁇ gene 3′ to J and just 5′ to primer B (see FIG. 2).
  • Primer C contains the RE2 site at its 5′ end.
  • the RE2 sequence is chosen in such a manner that when it is incorporated into the framework vector, no alteration of coding sequence of the C ⁇ chain occurs (See FIGS. 2 and 3).
  • This method of amplifying C ⁇ cDNAs incorporates the idea of nested primers for cloning a gene when the nucleotide sequence of only one region of the gene is known (Ohara, O., et al., Proc. Natl. Acad. Sci. USA, 86:5673 (1989)).
  • the PCR product is then cleaved with restriction enzymes RE1 and RE2 and cloned into the RE1 and RE2 sites of the pFHC vector (described below).
  • the sequence of primers and of RE1 and RE2 sites are selected so that when the PCR product is cloned into these sites, the sites are recreated and the cloned antibody gene fragments are brought back into the proper frame with respect to the framework immunoglobulin gene present in pFHC. This results in creation of a C ⁇ minigene which lacks the intron normally present between J and the C H 1 region of C ⁇ (See FIG. 3). These procedures result in production of the heavy chain library used to produce antigen-binding molecules of the present invention, as described further below.
  • diversity of the heavy chain variable region is increased by random mutagenesis, using techniques known to those of skill in the art.
  • the library produced as described above is amplified again, using PCR under conditions of limiting nucleotide concentration. Such conditions are known to increase the infidelity of the polymerization and result in production of mutant products.
  • Primers useful for this reaction are Primers C and D, as represented in FIGS. 2 and 3. Primer D derives from pFHC just 5′ to RE1.
  • the PCR product, after cleavage with RE1 and RE2 is recloned into the framework vector pFHC. To the extent that mutation affects codons of the antigen binding region, this procedure increases the diversity of the binding domains.
  • the starter library has a complexity of 10 6 elements, and an average of one mutation is introduced per complementarity determining region, and it is assumed that the complementarity determining region is 40 amino acids in size and that any of six amino acid substitutions can occur at a mutated codon
  • the diversity of the library can be increased by a factor of about 40 ⁇ 6, or 240, for single amino acid changes and 240 ⁇ 240, or about 6 ⁇ 10 4 , for double amino acid changes, yielding a final diversity of approximately 10 11 . This is considered to be in the range of the diversity of antibodies which animals produce (Tonegawa, S., Nature, 302:575 (1983)).
  • the framework vector for the light chain library includes components similar to those in the vector for the heavy chain library: the enhancer, promoter, a bacterial selectable marker, an animal selectable marker, bacterial origin of replication and light chain exons encoding the constant regions.
  • the animal selectable marker should differ from the animal selectable in pFHC. For example, if pFHC contains neo R , pFLC can contain Eco gpt.
  • a light chain library which contains diverse light chain fragments, is prepared as described above for construction of the heavy chain library.
  • the primers used are different from those described above for heavy chain library construction.
  • the primers are complementary to light chain mRNA encoding constant regions.
  • the framework vector contains the light chain constant region exons.
  • the library of immunoglobulin chain genes produced as described is subsequently introduced into a line of immortalized cultured animal cells, referred to as the “host” cells, in which the genes in the library are expressed.
  • the host cells particularly useful for this purpose are plasmacytoma cell lines or myeloma cell lines which have been shown to efficiently express and secrete introduced immunoglobulin genes (Morrison, S. L., et al., Ann. N.Y. Acad. Sci., 507:187 (1987); Kohler, G. and C. Milstein, Eur. J. Immunol., 6:511 (1976); Galfre and C. Milstein, Methods Enzymol., 73:3 (1981); Davis, A. C. and M. J.
  • the J558L cell line can be cotransfected using electro-poration or protoplast fusion (Morrison, S. L., et al., Ann. N.Y. Acad. Sci., 507:187 (1987)) and transfected cells selected on the basis of auxotrophic markers present on light and heavy chain libraries.
  • chimeric antibodies antibodies encoded by all or part of two or more genes
  • These chimeric antibodies are of two types: those in which one chain is encoded by a host cell gene and the other chain is encoded by an exogenously introduced antibody gene and those in which both the light and the heavy chain are encoded by an exogenous antibody gene. Both types of antibodies will be secreted.
  • a library of cells producing antibodies of diverse specificities is produced as a result.
  • the library of cells can be stored and maintained indefinitely by continuous culture and/or by freezing. A virtually unlimited number of cells can be obtained by this process.
  • Cells producing antigen-binding molecules of selected specificity can be identified and isolated using nitrocellulose filter layering or known techniques.
  • the same methods employed to identify and isolate hybridoma cells producing a desired antibody can be used:—cells are pooled and the supernatants tested for reactivity with antigen (Harlow, E. and D. Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, N.Y., p. 283 (1988). Subsequently, individual clones of cells are identified, using known techniques.
  • a preferred method for identification and isolation of cells makes use of nitrocellulose filter overlays, which allow the screening of a large number of cells.
  • Cells from the library of transfected myeloma cells are seeded in 10 cm 2 petri dishes in soft agar (Cook, W. D. and M. D. Scharff, PNAS, 74:5687 (1977); Paige, C. J., et al., Methods in Enzymol., 150:257 (1987)) at a density of 10 4 colony forming units, and allowed to form small colonies (approximately 300 cells). A large number of dishes (>100) may be so seeded. Cells are then overlayed with a thin film of agarose ( ⁇ 1 mm) and the agarose is allowed to harden. The agarose contains culture medium without serum.
  • Nitrocellulose filters (or other protein-binding filters) are layered on top of the agarose, and the dishes are incubated overnight. During this time, antibodies secreted by the cells will diffuse through the agarose and adhere to the nitrocellulose filters. The nitrocellulose filters are keyed to the underlying plate and removed for processing.
  • the method for processing nitrocellulose filters is identical to the methods used for Western blotting (Harlow, E. and D. Lane, Antibodies: Laboratory Manual, Cold Spring Harbor, N.Y., p. 283 (1988)).
  • the antibody molecules are adsorbed to the nitrocellulose filter.
  • the filters, as prepared above, are then blocked.
  • the desired antigen for example, keyhole lymphet hemocyanin (KLH), which has been iodinated with radioactive 125 I, is then applied in Western blotting buffers to the filters. (Other, non radiographic methods can be used for detection). After incubation, the filters are washed and dried and used to expose autoradiography film according to standard procedures.
  • KLH keyhole lymphet hemocyanin
  • the autoradiography film will be exposed.
  • Cells expressing the KLH reactive antibody can be identified by determining the location on the dish corresponding to an exposed filter; cells identified in this manner can be isolated using known techniques. Cells which are isolated from a region of the dish can then be rescreened, to insure the isolation of the clone of antigen-binding molecule-producing cells.
  • the gene(s) encoding an antigen-binding molecule of selected specificity can be isolated. This can be carried out, for example, as follows: primers D and C (see FIGS. 2 and 3) are used in a polymerase chain reaction, to produce all the heavy chain variable region genes introduced into the candidate host cell from the library. These genes are cloned again in the framework vector pFHC at the RE1 and RE2 sites. Similarly, all the light chain regions introduced into the host cell from the library are cloned into the light chain vector, pFLC. Members of the family of vectors so obtained are then transformed pairwise into myeloma cells, which are tested for the ability to produce and secrete the antibody with the desired selectivity.
  • antigen-binding molecules whose affinity for a selected antigen is altered (e.g., different from the affinity of a corresponding antigen-binding molecule produced by the present method). This can be carried out, for example, to increase the affinity of an antigen-binding molecule by randomly mutagenizing the genes isolated as described above using previously-described mutagenesis methods.
  • the variable region of antigen-binding molecule-encoding genes can be sequenced and site directed mutagenesis performed to mutate the complementarity determining regions (CDR) (Kabat, E. A., J. Immunol., 141:S 25-36 (1988)). Both processes result in production of a sublibrary of genes which can be screened for antigen-binding molecules of higher affinity or of altered affinity after the genes are expressed in myeloma cells.
  • variable regions Only following heavy-chain class switching are these variable regions expressed with a heavy chain of a different class (Shimizu, A. and T. Honjo, Cell, 36:801-803 (1984)).
  • the predominant population of B cells in nonimmune spleen cells is IgM + -cells (Cooper, M. D. and P. Burrows, In Immunoglobulin Genes, Academic Press, N.Y. p. 1 (1989)).
  • unidirectional nested PCR amplification is described above, other PCR procedures, as well as other DNA amplification techniques can be used to amplify DNA as needed in the present invention.
  • bidirectional PCR amplification of antibody variable regions can be carried out.
  • Framework vectors other than one using a mouse C ⁇ heavy chain constant region, which contains the C ⁇ enhancer and introns and a viral promoter can be used for inserting the products of PCR.
  • the vectors described were chosen for their subsequent use in the expression of the antibody genes, but any eukaryotic or prokaryotic cloning vector could be used to create a library of diverse cDNA genes encoding variable regions of antibody molecules.
  • the inserts from this vector could be transferred to any number of expression vectors.
  • other framework vectors which include intronless genes can be constructed, as can other heavy chain constant regions.
  • viral vectors or retroviral vectors can be used to introduce genes into myeloma cells.
  • the source for antibody molecule mRNAs can also be varied. Purified resting B lymphocytes from mouse nonimmunized spleen are described above as such a source. However, total spleens (immunized or not) from other animals, including humans, can be used, as can any source of antibody-producing cells (e.g., peripheral blood, lymph nodes, inflammatory tissue, bone marrow).
  • antibody-producing cells e.g., peripheral blood, lymph nodes, inflammatory tissue, bone marrow.
  • H and L chain gene DNA into myeloma cells using cotransformation by electroporation or protoplast fusion methods is described above (Morrison, S. L. and V. T. Oi, Adv. Immunol., 44:65 (1989)).
  • any means by which DNA can be introduced into living cells in vivo can be used, provided that it does not significantly interfere with the ability of the transformed cells to express the introduced DNA.
  • a method other than cotransformation can be used.
  • Cotransfection was chosen for its simplicity, and because both the H and L chains can be introduced into myeloma cells. It may be possible to introduce only the H chain into myeloma cells.
  • retroviral infection may be used.
  • Replication-incompetent retroviral vectors can be readily constructed which can be packaged into infective particles by helper cells (Mann, R., et al., Cell, 33:153-159 (1903)).
  • Viral titers of 10 5 infectious units per ml. can be achieved, making possible the transfer of very large numbers of genes, into myeloma cells.
  • Methods of identifying antigen-binding molecule-expressing cells expressing an antigen-binding molecule of selected specificity other than the nitrocellulose filter overlay technique described above can be used.
  • An important characteristic of any method is that it be useful to screen large numbers of different antibodies.
  • the nitrocellulose filter overlay technique for example, if 300 dishes are prepared and 10 4 independent transformed host cells per dish are screened, and if, on average, each cell produces ten different antibody molecules, then 300 ⁇ 10 4 ⁇ 3, or about 10 7 different antibodies can be screened at once. However, if the antibody molecules can be displayed on the cell surface, still larger numbers of cells can be screened using affinity matrices to pre-enrich for antigen-binding cells.
  • BCL 1 B 1 immortal B cell lines, such as BCL 1 B 1 , which will express IgM both on the cell surface and as a secreted form (Granowicz, E. S., et al., J. Immunol., 125:976 (1980)). If such cells are infected by retroviral vectors containing the terminal C ⁇ exons, the infected cells will likely produce both secreted and membrane bond forms of IgM (Webb, C. F., et al., J. Immunol., 143:3934-3939 (1989)). Still other methods can be used to detect antibody production. If the host cell is E. coli, a nitrocellulose overlay is possible, and such methods have been frequently used to detect E.
  • Viral coating can be used as a means of identifying viruses encoding antigen-combining molecules.
  • a viral vector is used to direct the synthesis of diverse antibody molecules.
  • the virus Upon lytic infection of host cells, and subsequent cell lysis, the virus becomes “coated” with the antibody product it directs. That is, the antibody molecule becomes physically linked to the outside of a mature virus particle, which can direct its synthesis.
  • Methods for viral coating are described below.
  • Viruses coated by antibody can be physically selected on the basis of their affinity to antigen which is attached to a solid support. The number of particles which can be screened using this approach is well in excess of 10 9 and it is possible that 10 11 different antibody genes could be screened in this manner.
  • an affinity matrix containing antigen used to purify those viruses encoding antibody molecules with affinity to antigen and which coat the surface of the virus which encodes those antibodies is used.
  • One method of viral coating is as follows—: A diverse library of bacteriophage ⁇ encoding parts of antibody molecules that are expressed in infected E. coli and which retain the ability to bind antigens is created, using known techniques (Orlandi, R., et al., Proc. Natl. Acad. Sci. USA, 86:3833 (1989); Huse, W. D., et al., Science, 246:1275 (1989); Better, M., et al., Science, 240:1041 (1988); Skerra, A. and A. Pluckthon, Science, 240:1038 (1988)). Bacteria infected with phage are embedded in a thin film of semisolid agar.
  • Greater than 10 7 infected bacteria may be plated in the presence of an excess of uninfected bacteria in a volume of 1 ml of agar and spread over a 10 cm 2 surface.
  • the agar contains monovalent antibody “A” (Parham, P., In Handbook of Experimental Immunology: Immunochem., Blackwell Scientific Publishers, Cambridge, Mass., pp. 14.1-14.23 (1986)), which can bind the ⁇ coat proteins and which has been chemically coupled to monovalent antibody “B”, which can bind an epitope on all viral directed antibody molecules.
  • Monovalent antibodies are used to prevent the crosslinking of viral particles. Upon lytic burst, progeny phage particles become effectively cross linked to the antibody molecule they encode.
  • a nitrocellulose filter (or other protein binding filter) is prepared as an affinity matrix by adsorbing the desired antigen. The filter is then blocked so that no other proteins bind nonspecifically. The filter is overlayed upon the agar, and coated phage are allowed to bind to the antigen by way of their adherent antibody molecules. Filters are washed to remove nonspecifically bound phage. Specifically bound phage therefore represent phage encoding antibodies with the desired specificity. These can now be propagated by reinfection of bacteria.
  • the present invention makes it possible to produce antigen-binding molecules which, like antibodies produced by presently-available techniques, bind to a selected antigen (i.e., having binding specifity).
  • Antibodies produced as described can be used, for example, to detect and neutralize antigens and deliver molecules to antigenic sites.
  • IgM heavy chain variable DNA is amplified from mRNA by the procedure represented schematically in FIG. 2.
  • Panel A depicts the relevant regions of the poly adenylated mRNA encoding the secreted form of the IgM heavy chain.
  • S denotes the sequences encoding the signal peptide which causes the nascent peptide to cross the plasma membrane, a necessary step in the processing and secretion of the antibody.
  • V, D and J derive from separate exons and together comprise the variable region.
  • C H 1, C H 2, and C H 3 are the three constant domains of C ⁇ . “Hinge” encodes the hinge region.
  • Primer C, B and Z are oligonucleotide PCR primers used in the amplification process.
  • Primers B and Z are complementary to the mRNA, and occur in the order shown relative to C.
  • Primer C in addition to being complementary to mRNA, has an extra bit of sequence at its 5′ end which allows the cloning of its PCR product. This is described below.
  • Panel B depicts the reverse transcript DNA product of the mRNA primed by oligonucleotide Z, with the addition of poly-dC by terminal transferase at the 3′ end of the product.
  • Panel C depicts the annealing of primer A to the reverse transcript DNA represented in Panel B.
  • Primer A contains the restriction endonuclease site RE1, with additional DNA at its 5′ end.
  • the constraints on the RE1 site are described in Example 2.
  • Panel D depicts the final double stranded DNA PCR product made utilizing primers A and B.
  • Panel E depicts the PCR product shown in Panel D annealed to Primer C.
  • Panel F is a blow up of panel E showing the structure of primer C.
  • Primer C consists of two parts: a 3′ part complementary to IgM heavy chain mRNA as shown, and a 5′ part which contains restriction site RE2. and spacer. Constraints on RE2 are described in Example 2.
  • Panel G depicts the final double stranded DNA PCR product utilizing Primers A and C and the product of the previous PCR (depicted in Panel D) as template. The S, V, D, J regions are again depicted.
  • a heavy chain framework vector designated pFHC, is constructed, using known techniques (See FIG. 3). It is useful for introducing antibody-encoding DNA into host cells, in which the DNA is expressed, resulting in antibody production.
  • the circular plasmid (above) is depicted linearized (below) and its relevant components are shown.
  • the neomycin antibiotic resistance gene (neo R ) is useful for selecting transformed animal cells (Sambrook, J., et al., Molecular Cloning: A Laboratory Manual, 2d Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)).
  • the bacterial replication origin and ampicillin antibiotic resistance genes useful respectively, for replication in E. coli and rendering E.
  • coli resistant to ampicillin can derive from any number of bacterial plasmids, including PBR322 (Sambrook, J., et al., Molecular Cloning: A Laborator Manual, 2d Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)).
  • the C ⁇ enhancer which derives from the intron between exons J and C H 1 of the C ⁇ gene, derives from any one of the cloned C ⁇ genes (Kawakami, T., et al., Nucleic Acids Research, 8:3933 (1980); Honjo, T., Ann. Rev. Immunol., 1:499 (1983)) and increases levels of transcription from antibody genes.
  • LTR contains the viral promoter from the Moloney MLV retrovirus DNA (Mulligan, R. C., Experimental Manipulation of Gene Expression, New York Academic Press, p. 155 (1983)).
  • D represents the PCR primer described in the text, depicted in its 5′ to 3′ orientation. The only constraints on D are its orientation, its complementarity to pFHC and its order relative to the RE1 and RE2 cloning sites. Preferably, D is within 100 nucleotides of RE1.
  • the cDNA cloning site contains restriction endonuclease sites RE1 and RE2, separated by spacer DNA which allows their efficient cleavage. The constraints on RE1 and RE2 are described below.
  • C H 1 The C ⁇ exons, as described in the text and literature, direct the synthesis of IgM heavy chain. Only part of C H 1 is present, as described below. C H 3 is chosen to contain the C ⁇ s region which specifies a secreted form of the heavy chain ((Kawakami, T., et al., Nucleic Acids Research, 8:3933 (1980); Honjo, T., Ann. Rev. Immunol., 1:499 (1983)). Finally, pFHC contains poly A addition and termination sequences which can be derived from the C ⁇ gene itself (Honjo, T., Ann. Rev.
  • the plasmid can be produced by combining the individual components, or nucleic acid segments, depicted in FIG. 3, using PCR cassette assembly (See below). Because the entire nucleotide sequence of each component is defined, the entire nucleotide sequence of the plasma is defined.
  • RE1 The constraints on RE1 are simple. It should be the sole cleavage site on the plasmid for its restriction endonuclease.
  • the choice of RE1 can be made by computer based sequence analysis (Intelligenetics Suite, Release 5:35, Intelligenetics).
  • RE2 The constraints on RE2 are more complex. First, it must be the sole cleavage site on the plasmid for its restriction endonuclease, as described for RE1. Moreover, the RE2 site must be such that when the PCR product is inserted, a gene is thereby created which is capable of directing the synthesis of a complete IgM heavy chain. This limits the choices for RE2, but the choices available can be determined by computer based sequence analysis. The choices can be determined as follows. First, a list of restriction endonucleases that do not cleave pFHC is compiled (see Table 1).
  • the rare non-cutters are surveyed by computer analysis for those which will cleave the N-doped sequence.
  • the search program will show a possible restriction endonuclease site, assuming a match between N and the restriction endonuclease cutting site. For example, with 39 rare non-cutters, 22 will cleave the N-doped sequence of C ⁇ C H 1, many of them several times (see Table 2).
  • Def means a definite cut site, of which there are none, because of the Ns.
  • Pos means a possible cleavage site at the indicated nucleotide position if N is chosen appropriately.
  • Y indicates any pyrimidine, “R” indicates any purine and “N” indicates any nucleotide.
  • the nucleotide positions refer to coordinates represented in FIG. 4.
  • Line 1 represents part of the actual amino acid sequence specified by the mouse C ⁇ C H 1 gene region
  • line 2 is the actual nucleotide sequence.
  • a cleavage site for the rare non-cutter BssHII is created.
  • the new sequence (containing the BssHII site) GCG CGC still encodes the identical amino acid sequence. Therefore, the sequence of the primer C is chosen to be the complement of line 3, and RE2 is the BssHII site.
  • a primer will function in the PCR and vector construction as desired. Other examples are possible, and the same process can be used in designing vectors and primers for cloning light chain variable regions.
  • the process of PCR cassette assembly is a method of constructing plasmid molecules (in this case the plasmid pFHC) from fragments of DNA of known nucleotide sequence.
  • Each fragment is then individually PCR amplified using synthesized oligonucleotide primers complementary to the terminal sequences of the fragment. These primers are synthesized to contain on their 5′ ends restriction endonuclease cleavage sites from the compiled list.
  • each PCR product can be so designed that each fragment can be assembled one by one into a larger plasmid structure by cleavage and ligation and transformation into E. coli.

Abstract

A method of producing libraries of genes encoding antigen-combining molecules or antibodies is described. In addition, a method of producing antigen-combining molecules which does not require an in vivo procedure is described. Vectors useful in the present method and antigen-combining molecules produced by the method are discussed. The antigen-combining molecules are useful for the detection, quantitation, purification and neutralization of antigens, as well as for diagnostic, therapeutic and prophylactic purposes.

Description

    RELATED APPLICATIONS
  • This application is a continuation of, and hereby claims priority to and incorporates by reference in their entirety, co-pending U.S. patent application Ser. No. 09/798,720, filed on Mar. 2, 2001 which is a continuation of U.S. patent application Ser. No. 09/439,732, filed Nov. 12, 1999, now U.S. Pat. No. 6,303,313; which is a continuation of abandoned U.S. patent application Ser. No. 08/997,195, filed Dec. 23, 1997; which is a continuation of U.S. patent application Ser. No. 08/315,269, filed Sep. 29, 1994, now U.S. Pat. No. 5,780,225; which is a continuation of U.S. patent application Ser. No. 07/919,730, filed Jul. 24, 1992, now U.S. Pat. No. 5,284,555; which is a continuation of abandoned U.S. application Ser. No. 07/464,350, filed Jan. 11, 1990. This application also claims priority to and incorporates by reference PCT Application No. PCT/US91/00209, filed Jan. 10, 1991.[0001]
  • BACKGROUND OF THE INVENTION
  • Monoclonal and polyclonal antibodies are useful for a variety of purposes. The precise antigen specificity of antibodies makes them powerful tools that can be used for the detection, quantitation, purification and neutralization of antigens. [0002]
  • Polyclonal antibodies are produced in vivo by immunizing animals, such as rabbits and goats, with antigens, bleeding the animals and isolating polyclonal antibody molecules from the blood. Monoclonal antibodies are produced by hybridoma cells, which are made by fusing, in vitro, immortal plasmacytoma cells with antibody producing cells (Kohler, G. and C. Milstein, Nature, 256:495 (1975)) obtained from animals immunized in vivo with antigen. [0003]
  • Current methods for producing polyclonal and monoclonal antibodies are limited by several factors. First, methods for producing either polyclonal or monoclonal antibodies require an in vivo immunization step. This can be time consuming and require large amounts of antigen. Second, the repertoire of antibodies expressed in vivo is restricted by physiological processes, such as those which mediate self-tolerance that disable auto-reactive B cells (Goodnow, C. C., et al., Nature, 334:676 (1988); Goodnow, J. W., [0004] Basic and Clinical Immunology, Ed. 5, Los Altos, Calif., Large Medical Publications (1984); Young, C. R., Molecular Immunology, New York, Marcel Dekker (1984)). Third, although antibodies can exist in millions of different forms, each with its own unique binding site for antigen, antibody diversity is restricted by genetic mechanisms for generating antibody diversity (Honjo, T., Ann. Rev. Immunol., 1:499 (1983); Tonegawa, S., Nature: 302:575 (1983)). Fourth, not all the antibody molecules which can be generated will be generated in a given animal. As a result, raising high affinity antibodies to a given antigen can be very time consuming and can often fail. Fifth, the production of human antibodies of desired specificity is very problematical.
  • A method of producing antibodies which avoids the limitations of presently-available methods, such as the requirement for immunization of an animal and in vivo steps, would be very useful, particularly if it made it possible to produce a wider range of antibody types than can be made using presently-available techniques and if it made it possible to produce human antibody types. [0005]
  • DISCLOSURE OF THE INVENTION
  • The present invention relates to a method of producing libraries of genes encoding antigen-combining molecules or antibodies; a method of producing-antigen-combining molecules, also referred to as antibodies, which does not require an in vivo procedure, as is required by presently-available methods; a method of obtaining antigen-combining molecules (antibodies) of selected or defined specificity which does not require an in vivo procedure; vectors useful in the present method and antibodies produced or obtained by the method. [0006]
  • The present invention relates to an in vitro process for synthesizing DNA encoding families of antigen-combining molecules or proteins. In this process, DNA containing genes encoding antigen-combining molecules is obtained and combined with oligonucleotides which are homologous to regions of the genes which are conserved. Sequence-specific gene amplification is then carried out using the DNA containing genes encoding antigen-combining proteins as template and the homologous oligonucleotides as primers. [0007]
  • This invention also relates to a method of creating is diverse libraries of DNAs encoding families of antigen-combining proteins by cloning the product of the in vitro process for synthesizing DNA, described in the preceeding paragraph, into an appropriate vector (e.g., a plasmid, viral or retroviral vector). [0008]
  • The subject invention provides an alternative method for the production of antigen-combining molecules, which are useful affinity reagents for the detection and neutralization of antigens and the delivery of molecules to antigenic sites. The claimed method differs from production of polyclonal antibody molecules derived by immunization of live animals and from production of mono-clonal antibody molecules through the use of hybridoma cell lines in that it does not require an in vivo immunization step, as do presently available methods. Rather, diverse libraries of genes which encode-antigen-combining sites comprising a significant proportion of an animal's repertoire of antibody combining sites are made, as described in detail herein. These genes are expressed in living cells, from which molecules of desired antigenic selectivity can be isolated and purified for various uses. [0009]
  • Antigen-combining molecules are produced by the present method in the following manner, which is described in greater deail below. Initially, a library of antibody genes which includes a set of variable regions encoding a large, diverse and random group of specificities derived from animal or human immunoglobulins is produced by amplifying or cloning diverse genomic fragments or cDNAs of antibody mRNAs found in antibody-producing tissue. [0010]
  • In an optional step, the diversity of the resulting libraries can be increased by means of random mutagenesis. The gene libraries are introduced into cultured host cells, which may be eukaryotic or prokaryotic, in which they are expressed. Genes encoding antibodies of desired antigenic specificity are identified, using a method described herein or known techniques, isolated and expressed in quantities in appropriate host cells, from which the encoded antibody can be purified. [0011]
  • Specifically, a library of genes encoding immunoglobulin heavy chain regions and a library of genes encoding immunoglobulin light chain regions are constructed. This is carried out by obtaining antibody-encoding DNA, which is either genomic fragments or cDNAs of antibody mRNAs, amplfying or cloning the fragments or cDNAs; and introducing them into a standard framework antibody gene vector, which is used to introduce the antibody-encoding DNA into cells in which the DNA is expressed. The vector includes a framework gene encoding a protein, such as a gene encoding an antibody heavy chain or an antibody light chain which can be of any origin (human, non-human) and can be derived from any of a number of existing DNAs encoding heavy chain immuno-globulins or light chain immunoglobulins. Such vectors are also a subject of the present invention and are described in greater detail in a subsequent section. Genes from one or both of the libraries are introduced into appropriate host cells, in which the genes are expressed, resulting in production of a wide variety of antigen-combining molecules. [0012]
  • Genes encoding antigen-combining molecules of desired specificity are identified by identifying cells producing antigen-combining molecules which react with a selected antigen and then obtaining the genes of interest. The genes of interest can subsequently be introduced into an appropriate host cell (or can be further modified and then introduced into an appropriate host cell) for further production of antigen-combining molecules, which can be purified and used for the same purposes for which conventionally-produced antibodies are used. [0013]
  • Through use of the method described, it is possible to produce antigen-combining molecules which are of wider diversity than are antibodies available as a result of known methods; novel antigen-combining molecules with a diverse range of specificities and affinities and antigen-combining molecules which are predominantly human in origin. Such antigen-combining molecules are a subject of the present invention and can be used clinically for diagnostic, therapeutic and prophylactic purposes, as well as in research contexts, and for other purposes.[0014]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a schematic representation of the method of the present invention by which antigen-combining molecules, or antibodies, are produced. [0015]
  • FIG. 2 is a schematic representation of amplification or cloning of IgM heavy chain variable region DNA from mRNA, using the polymerase chain reaction. [0016]
  • Panel A shows the relevant regions of the poly adenylated mRNA encoding the secreted form of the IgM heavy chain. S denotes the sequences encoding the signal peptide which causes the nascent peptide to cross the plasma membrane. V, D and J together comprise the variable region. C[0017] H1, CH2, and C H3 are the three constant domains of Cμ. Hinge encodes the hinge region. C, B and Z are oligonucleotide PCR primers (discussed below).
  • Panel B shows the reverse transcript DNA product of the mRNA primed by oligonucleotide Z, with the addition of poly-dC by terminal transferase at the 3′ end. [0018]
  • Panel C is a schematic representation of the annealing of primer A to the reverse transcript DNA. [0019]
  • Panel D shows the final double stranded DNA PCR product made utilizing primers A and B. [0020]
  • Panel E shows the product of PCR annealed to primer C. [0021]
  • Panel F is a blowup of Panel E, showing in greater detail the structure of primer C. Primer C consists of two parts: a 3′ part complementary to IgM heavy chain mRNA as shown, and a 5′ part which contains restriction site RE2 and spacer. [0022]
  • Panel G shows the final double stranded DNA PCR product made utilizing primers A and C and the product of the previous PCR (depicted in D) as template. The S, V, D, J regions are again depicted. [0023]
  • FIG. 3 is a schematic representation of the heavy chain framework vector pFHC. The circular plasmid (above) is depicted linearized (below) and its relevant components are shown: animal cell antibiotic resistance marker; bacterial replication origin; bacterial cell antibiotic resistance marker; Cμ enhancer; LTR containing the viral promoter from the Moloney MLV retrovirus DNA; PCR primer (D); cDNA cloning site containing restriction endonuclease sites, RE1 and RE2, separated by spacer DNA; Cμ exons; and poly A addition and termination sequences derived from the Cμ gene or having the same sequence as the Cμ gene. [0024]
  • FIG. 4 depicts a nucleotide sequence of the C[0025] H1 exon of the Cμ gene, and its encoded amino acid sequence (Panel A). The nucleotide coordinate numbers are listed above the line of nucleotide sequences. Panel B depicts the N-doped sequence, as defined in the text.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention provides a method of producing antigen-combining molecules (or antibodies) which does not require an in vivo immunization procedure and which makes it possible to produce antigen-combining molecules with far greater diversity than is shown by antibodies produced by currently-available techniques. [0026]
  • The present invention relates to a method of producing libraries of genes encoding antigen-combining molecules (antibody proteins) with diverse antigen-combining specificities; a method of producing such antigen-combining molecules, antigen-combining molecules produced by the method and vectors useful in the method. The following is a description of generation of such libraries, of the present method of producing antigen-combining molecules of selected specificity and of vectors useful in producing antigen-combining molecules of the present invention. [0027]
  • As described below, the process makes use of techniques which are known to those of skill in the art and can be applied as described herein to produce and identify antigen-combining molecules of desired antigenic specificity: the polymerase chain reaction (PCR), to amplify and clone diverse cDNAs encoding antibody mRNAs found in antibody-producing tissue; mutagenesis protocols to further increase the diversity of these cDNAs; gene transfer protocols to introduce antibody genes into cultured (prokaryotic and eukaryotic) cells for the purpose of expressing them; and screening protocols to detect genes encoding antibodies of the desired antigenic specificity. A general outline of the present method is represented in FIG. 1. [0028]
  • Construction of Library of Genes Encoding Antigen-Combining Molecules
  • A key step in the production of antigen-combining molecules by the present method is the construction of a “library” of antibody genes which include “variable” regions encoding a large, diverse, but random set of specificities. The library can be of human or non-human origin and is constructed as follows: [0029]
  • Initially, genomic DNA encoding antibodies or cDNAs of antibody mRNA (referred to as antibody-encoding DNA) is obtained. This DNA can be obtained from any source of antibody-producing cells, such as spleen cells, peripheral blood cells, lymph nodes, inflammatory tissue cells and bone marrow cells. It can also be obtained from a genomic library or cDNA library of B cells. The antibody-producing cells can be of human or non-human origin; genomic DNA or mRNA can be obtained directly from the tissue (i.e., without previous treatment to remove cells which do not produce antibody) or can be obtained after the tissue has been treated to increase concentration of antibody-producing cells or to select a particular type(s) of antibody-producing cells (i.e., treated to enrich the content of antibody-producing cells). Antibody-producing cells can be stimulated by an agent which stimulates antibody mRNA production (e.g., lipopolysaccharide) before DNA is obtained. [0030]
  • Antibody-encoding DNA is amplified and cloned using a known technique, such as the PCR using appropriately-selected primers, in order to produce sufficient quantities of the DNA and to modify the DNA in such a manner (e.g., by addition of appropriate restriction sites) that it can be introduced as an insert into an [0031] E. coli cloning vector. This cloning vector can serve as the expression vector or the inserts can later be introduced into an expression vector, such as the framework antibody gene vector described below. Amplified and cloned DNA can be further diversified, using mutagenesis, such as PCR, in order to produce a greater diversity or wider repertoire of antigen-binding molecules, as well as novel antigen-binding molecules.
  • Cloned antibody-encoding DNA is introduced into an expression vector, such as the framework antibody gene vector of the present invention, which can be a plasmid, viral or retroviral vector. Cloned antibody-encoding DNA is inserted into the vector in such a manner that the cloned DNA will be expressed as protein in appropriate host cells. It is essential that the expression vector used make it possible for the DNA insert to be expressed as a protein in the host cell. One expression vector useful in the present method is referred to as the framework antibody gene vector. Vectors useful in the present method contain antibody constant region or portions thereof in such a manner that when amplified DNA is inserted, the vector expresses a chimeric gene product comprising a variable region and a constant region in proper register. The two regions present in the chimeric gene product can be from the same type of immunoglobulin molecule or from two different types of immunoglobulin molecules. [0032]
  • These libraries of antibody-encoding genes are then expressed in cultured cells, which can be eukaryotic or prokaryotic. The libraries can be introduced into host cells separately or together. Introduction of the antibody-encoding DNA in vitro into host cells (by infection, transformation or transfection) is carried out using known techniques, such as electroporation, protoplast fusion or calcium phosphate co-precipitation. If only one library is introduced into a host cell, the host cell will generally be one which makes the other antibody chain, thus making it possible to produce complete/functional antigen-binding molecules. For example, if a heavy chain library produced by the present method is introduced into host cells, the host cells will generally be cultured cells, such as myeloma cells or [0033] E. coli, which naturally produce the other (i.e., light) chain of the immunoglobulin or are engineered to do so. Alternatively, both libraries can be introduced into appropriate host cells, either simultaneously or sequentially.
  • Host cells in which the antibody-encoding DNA is expressed can be eukaryotic or prokaryotic. They can be immortalized cultured animal cells, such as a myeloma cell line which has been shown to efficiently express and secrete introduced immunoglobulin genes (Morrison, S. L. et al., [0034] Ann. N.Y. Acad. Sci., 507:187 (1987); Kohler, G. and C. Milstein, Eur. J. Immunol., 6:511 (1976); Oi, V. T., et al., Immunoglobulin Gene Expression in Transformed Lymphoid Cells, 80:825 (1983); Davis, A. C. and M. J. Shulman, Immunol. Today, 10:119 (1989)). One host cell which can be used to express the antibody-encoding DNA is the J558L cell line or the SP2/0 cell line.
  • Cells expressing antigen-combining molecules with a desired specificity for a given antigen can then be selected by a variety of means, such as testing for reactivity with a selected antigen using nitrocellulose layering. The antibodies identified thereby can be of human origin, nonhuman origin or a combination of both. That is, all or some of the components (e.g., heavy chain, light chain, variable regions, constant regions) can be encoded by DNA of human or nonhuman origin, which, when expressed produces the encoded chimeric protein which, in turn, may be human, nonhuman or a combination of both. In such antigen-combining molecules, all or some of the regions (e.g., heavy and light chain variable and constant regions) are referred to as being of human origin or of nonhuman origin, based on the source of the DNA encoding the antigen-combining molecule region in question. For example, in the case in which DNA encoding mouse heavy chain variable region is expressed in host cells, the resulting antigen-combining molecule has a heavy chain variable region of mouse origin. Antibodies produced may be used for such purposes as drug delivery, tumor imaging and other therapeutic, diagnostic and prophylactic uses. [0035]
  • Once antibodies of a desired binding specificity are obtained, their genes may be isolated and further mutagenized to create additional antigen combining diversity or antibodies of higher affinity for antigen. [0036]
  • Construction of Immunoglobulin Heavy Chain Gene Library and Production of Encoded Antigen-Binding Molecules
  • The following is a detailed description of a specific experimental protocol which embodies the concepts described above. Although the following is a description of one particular embodiment, the same procedures can be used to produce libraries in which the immunoglobulin and the heavy chain class are different or in which light chain genes are amplified and cloned. The present invention is not intended to be limited to this example. In the embodiment presented below, a diverse heavy chain gene library is constructed. Using the principles described in relation to the heavy chain gene library, a diverse light chain gene library is also constructed. These are co-expressed in an immortal tumor cell capable of producing antibodies, such as plasma-cytoma cells or myeloma cells. Cells expressing antibody reactive to antigen are identified by a nitrocellulose filter overlay and antibody is prepared from cells identified as expressing it. As described in a subsequent section, there are alternative methods of library construction, other expression systems which can be used, and alternative selection systems for identifying antibody-producing cells or viruses. [0037]
  • Step 1 in this specific protocol is construction of libraries of genes in [0038] E. coli which encode immunoglobulin heavy chains. This is followed by the use of random mutagenesis to increase the diversity of the library, which is an optional procedure. Step 2 is introduction of the library, by transfection, into myeloma cells. Step 3 is identification of myeloma cells expressing antibody with the desired specificity, using the nitrocellulose filter overlay technique or techniques known to those of skill in the art. Step 4 is isolation of the gene(s) encoding the antibody with the desired specificity and their expression in appropriate host cells, to produce antigen-combining fragments useful for a variety of purposes.
  • Construction
  • One key step in construction of the library of cDNAs encoding the variable region of mouse heavy chain genes is construction of an [0039] E. coli plasmid vector, designated pFHC. pFHC contains a “framework” gene, which can be any antibody heavy chain and serves as a site into which the amplified cloned gene product (genomic DNA or cDNA of antibody mRNAs) is introduced. pFHC is useful as a vector for this purpose because it contains RE1 and RE2 cloning sites. Other vectors which include a framework gene and other cloning sites can be used for this purpose as well. The framework gene includes a transcriptional promoter (e.g., a powerful promoter, such as a Moloney LTR (Mulligan, R. C., In Experimental Manipulation of Gene Expression, New York Adacemic Press, p. 155 (1983)) and a Cμ chain transcriptional enhancer to increase the level of transcriptions from the promoter (Gillies, S. D., et al., Cell, 33:717 (1983), a cloning site containing RE1 and RE2; part of the Cμ heavy chain gene encoding secreted protein; and poly A addition and termination sequences (FIG. 3). The framework antibody gene vector of the present invention (pFHC) also includes a selectable marker (e.g., an antibiotic resistance gene such as the neomycin resistance gene, neoR) for animal cells; sequences for bacterial replication (ori); and a selectable marker (e.g., the ampicillin resistance gene, AmpR) for bacterial cells. The framework gene can be of any origin (human, non-human), and can derive from any one of a number of existing DNAs encoding heavy chain immunoglobulins (Tucker, P. W., et al., Science, 206:1299 (1979); Honjo, T., et al., Cell, 18:559 (1979); Bothwell, A. L. M., et al., Cell, 24:625 (1981); Liu, A.Y, et al., Gene, 54:33 (1987); Kawakami, T., et al., Nuc. Acids. Res., 8:3933 (1980)). In this embodiment, the vector retains the introns between the CH1, hinge, CH2 and C H3 exons. The “variable region” of the gene, which includes the V, D and J regions of the antibody heavy chain and which encodes the antigen binding site, is deleted and replaced with two consecutive restriction endonuclease cloning sites, RE1 and RE2. The restriction endonuclease site RE1 occurs just 3′ to the LTR promoter and the restriction endonuclease site RE2 occurs within the constant region just 3′ to the J region (see FIG. 3).
  • Another key step in the production of antigen-combining molecules in this embodiment of the present invention is construction in an [0040] E. coli vector of a library of cDNAs encoding the variable region of mouse immunoglobulin genes. In this embodiment, the pFHC vector, which includes cloning sites designated RE1 and RE2, is used for cloning heavy chain variable regions, although any cloning vector with cloning sites having the same or similar characteristics (described below) can be used. Similarly, a light chain vector can be designed, using the above described procedures and procedures known to a person of ordinary skill in the art.
  • In this embodiment, non-immune mouse spleens are used as the starting material. mRNA is prepared directly from the spleen or from spleen processed in such a manner that it is enriched for resting B cells. Enrichment of tissue results in a more uniform representation of antibody diversity in the starting materials. Lymphocytes can be purified from spleen using ficoll gradients (Boyum, A., [0041] Scand. J. of Clinical Invest., 21:77 (1968)). B cells are separated from other cells (e.g., T cells) by panning with anti-IgM coated dishes (Wysocki , L. J. and V. L. Sato, Proc. Natl. Acad. Sci. 75:2844 (1978)). Because activated cells express the IL-2 receptor but resting B cells do not, resting B cells can be separated yet further from activated cells by panning. Further purification by size fractionation on a Cell Sorter results in a fairly homogeneous population of resting B cells.
  • Poly A+ mRNA from total mouse spleen is prepared according to published methods (Sambrook, J., et al., [0042] Molecular Cloning: A Laboratory Manual, 2d Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)). Production of antibody mRNA can first be stimulated by lipopolysaccharide (LPS) (Andersson, J. A., et al., J. Exp. Med., 145:1511 (1977)). First strand cDNA is prepared to this mRNA population using as primer an oligonucleotide, Z, which is complementary to Cμ in the CH1 region 3′ to J. This primer is designated Z in FIG. 2. First strand cDNA is then elongated by the terminal transferase reaction with dCTP to form a poly dC tail (Sambrook, J., et al., Molecular Cloning: A Laboratory Manual, 2d Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)).
  • This DNA product is then used as template in a polymerase chain reaction (PCR) to amplify cDNAs encoding antibody variable regions (Saiki, R. K., et al., [0043] Science, 239:487 (1988); Ohara, O., et al., Proc. Natl. Acad. Sci. USA, 86:5673 (1989)). Initially, PCR is carried out with two primers: primer A and primer B, as represented in FIG. 2. Primer A contains the RE1 site at its 5′ end, followed by poly dG. Primer B is complementary to the constant (CH1) region of the Cμ gene, 3′ to the J region and 5′ to primer Z (see FIG. 2). Primer B is complementary to all Cμ genes, which encode the heavy chain of molecules of the IgM class, the Ig class expressed by all B cell clones prior to class switching (Schimizu, A. and T. Honjo, Cell, 36:801-803 (1984)) and present in resting B cells. The resultant PCR product includes a significant proportion of cDNAs encompassing the various VH regions expressed as IgM in the mouse. (The use of other primers complementary to the cDNA genes encoding the constant regions of other immunoglobulin heavy chains can be used in parallel reactions to obtain the variable regions expressed on these molecules, but for simplicity these are not described).
  • Next, the product of the first PCR procedure is used again for PCR with primer A and primer C. Primer C, like primer B, is complementary to the [0044] Cμ gene 3′ to J and just 5′ to primer B (see FIG. 2). Primer C contains the RE2 site at its 5′ end. The RE2 sequence is chosen in such a manner that when it is incorporated into the framework vector, no alteration of coding sequence of the Cμ chain occurs (See FIGS. 2 and 3). This method of amplifying Cμ cDNAs, referred to as unidirectional nested PCR, incorporates the idea of nested primers for cloning a gene when the nucleotide sequence of only one region of the gene is known (Ohara, O., et al., Proc. Natl. Acad. Sci. USA, 86:5673 (1989)). The PCR product is then cleaved with restriction enzymes RE1 and RE2 and cloned into the RE1 and RE2 sites of the pFHC vector (described below). The sequence of primers and of RE1 and RE2 sites are selected so that when the PCR product is cloned into these sites, the sites are recreated and the cloned antibody gene fragments are brought back into the proper frame with respect to the framework immunoglobulin gene present in pFHC. This results in creation of a Cμ minigene which lacks the intron normally present between J and the CH1 region of Cμ (See FIG. 3). These procedures result in production of the heavy chain library used to produce antigen-binding molecules of the present invention, as described further below.
  • Optionally, diversity of the heavy chain variable region is increased by random mutagenesis, using techniques known to those of skill in the art. [0045]
  • For example, the library produced as described above is amplified again, using PCR under conditions of limiting nucleotide concentration. Such conditions are known to increase the infidelity of the polymerization and result in production of mutant products. Primers useful for this reaction are Primers C and D, as represented in FIGS. 2 and 3. Primer D derives from pFHC just 5′ to RE1. The PCR product, after cleavage with RE1 and RE2, is recloned into the framework vector pFHC. To the extent that mutation affects codons of the antigen binding region, this procedure increases the diversity of the binding domains. For example, if the starter library has a complexity of 10[0046] 6 elements, and an average of one mutation is introduced per complementarity determining region, and it is assumed that the complementarity determining region is 40 amino acids in size and that any of six amino acid substitutions can occur at a mutated codon, the diversity of the library can be increased by a factor of about 40×6, or 240, for single amino acid changes and 240×240, or about 6×104, for double amino acid changes, yielding a final diversity of approximately 1011. This is considered to be in the range of the diversity of antibodies which animals produce (Tonegawa, S., Nature, 302:575 (1983)). Even greater diversity can be generated by the random combination of H and L chains, the result of co-expression in host cells (see below). It is, thus, theoretically possible to generate a more diverse antibody library in vitro than can be generated in vivo. This library of genes is called the “high diversity” heavy chain library. It may be propagated indefinitely in E. coli. A high diversity light chain library can be prepared similarly.
  • The framework vector for the light chain library, designated pFLC, includes components similar to those in the vector for the heavy chain library: the enhancer, promoter, a bacterial selectable marker, an animal selectable marker, bacterial origin of replication and light chain exons encoding the constant regions. For pFLC, the animal selectable marker should differ from the animal selectable in pFHC. For example, if pFHC contains neo[0047] R, pFLC can contain Eco gpt.
  • A light chain library, which contains diverse light chain fragments, is prepared as described above for construction of the heavy chain library. In constructing the light chain library, the primers used are different from those described above for heavy chain library construction. In this instance, the primers are complementary to light chain mRNA encoding constant regions. The framework vector contains the light chain constant region exons. [0048]
  • Introduction of the Library of Immunoglobulin Chain Genes into Immortalized Animal Cells
  • The library of immunoglobulin chain genes produced as described is subsequently introduced into a line of immortalized cultured animal cells, referred to as the “host” cells, in which the genes in the library are expressed. Particularly useful for this purpose are plasmacytoma cell lines or myeloma cell lines which have been shown to efficiently express and secrete introduced immunoglobulin genes (Morrison, S. L., et al., [0049] Ann. N.Y. Acad. Sci., 507:187 (1987); Kohler, G. and C. Milstein, Eur. J. Immunol., 6:511 (1976); Galfre and C. Milstein, Methods Enzymol., 73:3 (1981); Davis, A. C. and M. J. Shulman, Immunol. Today, 10:119 (1989)). For example, the J558L cell line can be cotransfected using electro-poration or protoplast fusion (Morrison, S. L., et al., Ann. N.Y. Acad. Sci., 507:187 (1987)) and transfected cells selected on the basis of auxotrophic markers present on light and heavy chain libraries.
  • As a result of cotransformation and selection for markers on both light chain and heavy chain vectors, most transformed host cells will express several copies of immunoglobulin heavy and light chains from the diverse library, and will express chimeric antibodies (antibodies encoded by all or part of two or more genes) (Nisonoff, A., et al., [0050] In The Antibody Molecule, Academic Press, NY p. 238 (1975)). These chimeric antibodies are of two types: those in which one chain is encoded by a host cell gene and the other chain is encoded by an exogenously introduced antibody gene and those in which both the light and the heavy chain are encoded by an exogenous antibody gene. Both types of antibodies will be secreted. A library of cells producing antibodies of diverse specificities is produced as a result. The library of cells can be stored and maintained indefinitely by continuous culture and/or by freezing. A virtually unlimited number of cells can be obtained by this process.
  • Isolation of Cells Producing Antigen-Binding Molecules of Selected Specificity
  • Cells producing antigen-binding molecules of selected specificity (i.e., which bind to a selected antigen) can be identified and isolated using nitrocellulose filter layering or known techniques. The same methods employed to identify and isolate hybridoma cells producing a desired antibody can be used:—cells are pooled and the supernatants tested for reactivity with antigen (Harlow, E. and D. Lane, [0051] Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, N.Y., p. 283 (1988). Subsequently, individual clones of cells are identified, using known techniques. A preferred method for identification and isolation of cells makes use of nitrocellulose filter overlays, which allow the screening of a large number of cells. Cells from the library of transfected myeloma cells are seeded in 10 cm2 petri dishes in soft agar (Cook, W. D. and M. D. Scharff, PNAS, 74:5687 (1977); Paige, C. J., et al., Methods in Enzymol., 150:257 (1987)) at a density of 104 colony forming units, and allowed to form small colonies (approximately 300 cells). A large number of dishes (>100) may be so seeded. Cells are then overlayed with a thin film of agarose (<1 mm) and the agarose is allowed to harden. The agarose contains culture medium without serum. Nitrocellulose filters (or other protein-binding filters) are layered on top of the agarose, and the dishes are incubated overnight. During this time, antibodies secreted by the cells will diffuse through the agarose and adhere to the nitrocellulose filters. The nitrocellulose filters are keyed to the underlying plate and removed for processing.
  • The method for processing nitrocellulose filters is identical to the methods used for Western blotting (Harlow, E. and D. Lane, [0052] Antibodies: Laboratory Manual, Cold Spring Harbor, N.Y., p. 283 (1988)). The antibody molecules are adsorbed to the nitrocellulose filter. The filters, as prepared above, are then blocked. The desired antigen, for example, keyhole lymphet hemocyanin (KLH), which has been iodinated with radioactive 125I, is then applied in Western blotting buffers to the filters. (Other, non radiographic methods can be used for detection). After incubation, the filters are washed and dried and used to expose autoradiography film according to standard procedures. Where the filters have adsorbed antibody molecules which are capable of binding KLH, the autoradiography film will be exposed. Cells expressing the KLH reactive antibody can be identified by determining the location on the dish corresponding to an exposed filter; cells identified in this manner can be isolated using known techniques. Cells which are isolated from a region of the dish can then be rescreened, to insure the isolation of the clone of antigen-binding molecule-producing cells.
  • Isolation of Genes Encoding Antigen-Binding Molecules of Selected Specificity and Purification of Encoded Antigen-Binding Molecules
  • The gene(s) encoding an antigen-binding molecule of selected specificity can be isolated. This can be carried out, for example, as follows: primers D and C (see FIGS. 2 and 3) are used in a polymerase chain reaction, to produce all the heavy chain variable region genes introduced into the candidate host cell from the library. These genes are cloned again in the framework vector pFHC at the RE1 and RE2 sites. Similarly, all the light chain regions introduced into the host cell from the library are cloned into the light chain vector, pFLC. Members of the family of vectors so obtained are then transformed pairwise into myeloma cells, which are tested for the ability to produce and secrete the antibody with the desired selectivity. Purification of the antibody from these cells can then be accomplished using standard procedures (Johnstone, A. and R. Thorpe, [0053] Immunochem. in Practice, Blackwell Scientific, Oxford, p. 27 (1982); Harlow, E. and D. Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, N.Y., p. 283 (1988)).
  • Alteration of Affinity of Antigen-Binding Molecules
  • It is also possible to produce antigen-binding molecules whose affinity for a selected antigen is altered (e.g., different from the affinity of a corresponding antigen-binding molecule produced by the present method). This can be carried out, for example, to increase the affinity of an antigen-binding molecule by randomly mutagenizing the genes isolated as described above using previously-described mutagenesis methods. Alternatively, the variable region of antigen-binding molecule-encoding genes can be sequenced and site directed mutagenesis performed to mutate the complementarity determining regions (CDR) (Kabat, E. A., [0054] J. Immunol., 141:S 25-36 (1988)). Both processes result in production of a sublibrary of genes which can be screened for antigen-binding molecules of higher affinity or of altered affinity after the genes are expressed in myeloma cells.
  • Alternative Materials and Procedures for Use in the Present Method
  • In addition to those described above for use in the method of the present invention, other materials (e.g., starting materials, primers) and procedures can be used in carrying out the method. For example, use of PCR technology to clone a large collection of cDNA genes encoding variable regions of heavy chains has been described above. Although primers from the Cμ class were described as being used in unidirectional nested PCR, the present invention is not limited to these conditions. For example, primers from any of the other heavy chain classes (Cγ[0055] 3, Cγ1, Cγ2b, Cα for example) or from light chains can be used. Cμ was described as of particular use because of the fact that the entire repertoire of heavy chain variable regions are initially expressed as IgM. Only following heavy-chain class switching are these variable regions expressed with a heavy chain of a different class (Shimizu, A. and T. Honjo, Cell, 36:801-803 (1984)). In addition, the predominant population of B cells in nonimmune spleen cells is IgM+-cells (Cooper, M. D. and P. Burrows, In Immunoglobulin Genes, Academic Press, N.Y. p. 1 (1989)). Although unidirectional nested PCR amplification is described above, other PCR procedures, as well as other DNA amplification techniques can be used to amplify DNA as needed in the present invention. For example, bidirectional PCR amplification of antibody variable regions can be carried out. This approach requires use of multiple degenerate 5′ primers (Orlandi, R., et al., Proc. Natl. Acad. Sci. USA, 86:3833 (1989); Sastry, L., et al., Proc. Natl. Acad. Sci. USA, 86:5728 (1989)). Bidirectional amplification may not pick up the same full diversity of genes as can be expected from unidirectional PCR.
  • In addition, methods of introducing further diversity into the antibody library other than the method for random mutagenesis utilizing PCR described above can be used. Other methods of random mutagenesis, such as that described by Sambrook, et al. (Sambrook, J., et al., [0056] Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N. Y. (1989)) can be used, as can direct mutagenesis of the complementarity determining regions (CDRs).
  • Framework vectors other than one using a mouse Cμ heavy chain constant region, which contains the Cμ enhancer and introns and a viral promoter (described previously) can be used for inserting the products of PCR. The vectors described were chosen for their subsequent use in the expression of the antibody genes, but any eukaryotic or prokaryotic cloning vector could be used to create a library of diverse cDNA genes encoding variable regions of antibody molecules. The inserts from this vector could be transferred to any number of expression vectors. For example, other framework vectors which include intronless genes can be constructed, as can other heavy chain constant regions. In addition to plasmid vectors, viral vectors or retroviral vectors can be used to introduce genes into myeloma cells. [0057]
  • The source for antibody molecule mRNAs can also be varied. Purified resting B lymphocytes from mouse nonimmunized spleen are described above as such a source. However, total spleens (immunized or not) from other animals, including humans, can be used, as can any source of antibody-producing cells (e.g., peripheral blood, lymph nodes, inflammatory tissue, bone marrow). [0058]
  • Introduction of H and L chain gene DNA into myeloma cells using cotransformation by electroporation or protoplast fusion methods is described above (Morrison, S. L. and V. T. Oi, [0059] Adv. Immunol., 44:65 (1989)). However, any means by which DNA can be introduced into living cells in vivo can be used, provided that it does not significantly interfere with the ability of the transformed cells to express the introduced DNA. In fact, a method other than cotransformation, can be used. Cotransfection was chosen for its simplicity, and because both the H and L chains can be introduced into myeloma cells. It may be possible to introduce only the H chain into myeloma cells. Moreover, the H chain itself in many cases carries sufficient binding affinity for antigen. However, other methods can also be used. For example, retroviral infection may be used. Replication-incompetent retroviral vectors can be readily constructed which can be packaged into infective particles by helper cells (Mann, R., et al., Cell, 33:153-159 (1903)). Viral titers of 105 infectious units per ml. can be achieved, making possible the transfer of very large numbers of genes, into myeloma cells.
  • Further increases in the diversity of antibody-producing cells than results from the method described above can be generated if light and heavy chain genes are introduced separately into myeloma cells. Light chain genes can be introduced into one set of myeloma cells with one selectable marker, and heavy chains into another set of cells with a different selectable marker. Myeloma cells containing and expressing both H and L chains could then be generated by the highly efficient process of polyethylene glycol mediated cell fusion (Pontecorvo, G., [0060] Somatic Cell Genetics, 1:397 (1975)). Thus, a method of screening diverse libraries of antibody genes using animal cells is not limited by the number of cells which can be generated, but by the number of cells which can be screened.
  • Methods of identifying antigen-binding molecule-expressing cells expressing an antigen-binding molecule of selected specificity other than the nitrocellulose filter overlay technique described above can be used. An important characteristic of any method is that it be useful to screen large numbers of different antibodies. With the nitrocellulose filter overlay technique, for example, if 300 dishes are prepared and 10[0061] 4 independent transformed host cells per dish are screened, and if, on average, each cell produces ten different antibody molecules, then 300×104×3, or about 107 different antibodies can be screened at once. However, if the antibody molecules can be displayed on the cell surface, still larger numbers of cells can be screened using affinity matrices to pre-enrich for antigen-binding cells. There are immortal B cell lines, such as BCL1B1, which will express IgM both on the cell surface and as a secreted form (Granowicz, E. S., et al., J. Immunol., 125:976 (1980)). If such cells are infected by retroviral vectors containing the terminal Cμ exons, the infected cells will likely produce both secreted and membrane bond forms of IgM (Webb, C. F., et al., J. Immunol., 143:3934-3939 (1989)). Still other methods can be used to detect antibody production. If the host cell is E. coli, a nitrocellulose overlay is possible, and such methods have been frequently used to detect E. coli producing particular proteins (Sambrook, J., et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)). Other methods of detection are possible and one in particular, which involves the concept of “viral coating”, is discussed below.
  • Viral coating can be used as a means of identifying viruses encoding antigen-combining molecules. In this method, a viral vector is used to direct the synthesis of diverse antibody molecules. Upon lytic infection of host cells, and subsequent cell lysis, the virus becomes “coated” with the antibody product it directs. That is, the antibody molecule becomes physically linked to the outside of a mature virus particle, which can direct its synthesis. Methods for viral coating are described below. Viruses coated by antibody can be physically selected on the basis of their affinity to antigen which is attached to a solid support. The number of particles which can be screened using this approach is well in excess of 10[0062] 9 and it is possible that 1011 different antibody genes could be screened in this manner. In one embodiment, an affinity matrix containing antigen used to purify those viruses encoding antibody molecules with affinity to antigen and which coat the surface of the virus which encodes those antibodies is used.
  • One method of viral coating is as follows—: A diverse library of bacteriophage λ encoding parts of antibody molecules that are expressed in infected [0063] E. coli and which retain the ability to bind antigens is created, using known techniques (Orlandi, R., et al., Proc. Natl. Acad. Sci. USA, 86:3833 (1989); Huse, W. D., et al., Science, 246:1275 (1989); Better, M., et al., Science, 240:1041 (1988); Skerra, A. and A. Pluckthon, Science, 240:1038 (1988)). Bacteria infected with phage are embedded in a thin film of semisolid agar. Greater than 107 infected bacteria may be plated in the presence of an excess of uninfected bacteria in a volume of 1 ml of agar and spread over a 10 cm2 surface. The agar contains monovalent antibody “A” (Parham, P., In Handbook of Experimental Immunology: Immunochem., Blackwell Scientific Publishers, Cambridge, Mass., pp. 14.1-14.23 (1986)), which can bind the λ coat proteins and which has been chemically coupled to monovalent antibody “B”, which can bind an epitope on all viral directed antibody molecules. Monovalent antibodies are used to prevent the crosslinking of viral particles. Upon lytic burst, progeny phage particles become effectively cross linked to the antibody molecule they encode. Because lysis occurs in semisolid medium, in which diffusion is slow, cross linking between a given phage and the antibody encoded by another phage is minimized. A nitrocellulose filter (or other protein binding filter) is prepared as an affinity matrix by adsorbing the desired antigen. The filter is then blocked so that no other proteins bind nonspecifically. The filter is overlayed upon the agar, and coated phage are allowed to bind to the antigen by way of their adherent antibody molecules. Filters are washed to remove nonspecifically bound phage. Specifically bound phage therefore represent phage encoding antibodies with the desired specificity. These can now be propagated by reinfection of bacteria.
  • Thus the present invention makes it possible to produce antigen-binding molecules which, like antibodies produced by presently-available techniques, bind to a selected antigen (i.e., having binding specifity). Antibodies produced as described can be used, for example, to detect and neutralize antigens and deliver molecules to antigenic sites. [0064]
  • EXAMPLE I Amplification of IgM Heavy Chain Variable Region DNA from mRNA
  • IgM heavy chain variable DNA is amplified from mRNA by the procedure represented schematically in FIG. 2. In FIG. 2, Panel A depicts the relevant regions of the poly adenylated mRNA encoding the secreted form of the IgM heavy chain. In Panel A, S denotes the sequences encoding the signal peptide which causes the nascent peptide to cross the plasma membrane, a necessary step in the processing and secretion of the antibody. V, D and J derive from separate exons and together comprise the variable region. C[0065] H1, CH2, and C H3 are the three constant domains of Cμ. “Hinge” encodes the hinge region. C, B and Z are oligonucleotide PCR primers used in the amplification process. The only constraints on Primers B and Z are that they are complementary to the mRNA, and occur in the order shown relative to C. Primer C, in addition to being complementary to mRNA, has an extra bit of sequence at its 5′ end which allows the cloning of its PCR product. This is described below. Panel B depicts the reverse transcript DNA product of the mRNA primed by oligonucleotide Z, with the addition of poly-dC by terminal transferase at the 3′ end of the product. Panel C depicts the annealing of primer A to the reverse transcript DNA represented in Panel B. Primer A contains the restriction endonuclease site RE1, with additional DNA at its 5′ end. The constraints on the RE1 site are described in Example 2. Panel D depicts the final double stranded DNA PCR product made utilizing primers A and B. Panel E depicts the PCR product shown in Panel D annealed to Primer C. Panel F is a blow up of panel E showing the structure of primer C. Primer C consists of two parts: a 3′ part complementary to IgM heavy chain mRNA as shown, and a 5′ part which contains restriction site RE2. and spacer. Constraints on RE2 are described in Example 2. Panel G depicts the final double stranded DNA PCR product utilizing Primers A and C and the product of the previous PCR (depicted in Panel D) as template. The S, V, D, J regions are again depicted.
  • EXAMPLE 2 Construction of Heavy Chain Framework Vector pFHC
  • A heavy chain framework vector, designated pFHC, is constructed, using known techniques (See FIG. 3). It is useful for introducing antibody-encoding DNA into host cells, in which the DNA is expressed, resulting in antibody production. The circular plasmid (above) is depicted linearized (below) and its relevant components are shown. The neomycin antibiotic resistance gene (neo[0066] R) is useful for selecting transformed animal cells (Sambrook, J., et al., Molecular Cloning: A Laboratory Manual, 2d Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)). The bacterial replication origin and ampicillin antibiotic resistance genes, useful respectively, for replication in E. coli and rendering E. coli resistant to ampicillin, can derive from any number of bacterial plasmids, including PBR322 (Sambrook, J., et al., Molecular Cloning: A Laborator Manual, 2d Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)). The Cμ enhancer, which derives from the intron between exons J and CH1 of the Cμ gene, derives from any one of the cloned Cμ genes (Kawakami, T., et al., Nucleic Acids Research, 8:3933 (1980); Honjo, T., Ann. Rev. Immunol., 1:499 (1983)) and increases levels of transcription from antibody genes. LTR contains the viral promoter from the Moloney MLV retrovirus DNA (Mulligan, R. C., Experimental Manipulation of Gene Expression, New York Academic Press, p. 155 (1983)). D represents the PCR primer described in the text, depicted in its 5′ to 3′ orientation. The only constraints on D are its orientation, its complementarity to pFHC and its order relative to the RE1 and RE2 cloning sites. Preferably, D is within 100 nucleotides of RE1. The cDNA cloning site contains restriction endonuclease sites RE1 and RE2, separated by spacer DNA which allows their efficient cleavage. The constraints on RE1 and RE2 are described below. The Cμ exons, as described in the text and literature, direct the synthesis of IgM heavy chain. Only part of CH1 is present, as described below. C H3 is chosen to contain the Cμs region which specifies a secreted form of the heavy chain ((Kawakami, T., et al., Nucleic Acids Research, 8:3933 (1980); Honjo, T., Ann. Rev. Immunol., 1:499 (1983)). Finally, pFHC contains poly A addition and termination sequences which can be derived from the Cμ gene itself (Honjo, T., Ann. Rev. Immunol., 1:499 (1983); Kawakami, T., et al., Nucleic Acids Research, 8:3933 (1980)). One potential advantage of using the entire Cμ gene is that in some host cell systems, a membrane bound and secreted form of IgM may be expressed (Granowicz, E. S., et al., J. Immunol, 125:976 (1980)).
  • The plasmid can be produced by combining the individual components, or nucleic acid segments, depicted in FIG. 3, using PCR cassette assembly (See below). Because the entire nucleotide sequence of each component is defined, the entire nucleotide sequence of the plasma is defined. [0067]
  • The constraints on RE1 are simple. It should be the sole cleavage site on the plasmid for its restriction endonuclease. The choice of RE1 can be made by computer based sequence analysis (Intelligenetics Suite, Release 5:35, Intelligenetics). [0068]
  • The constraints on RE2 are more complex. First, it must be the sole cleavage site on the plasmid for its restriction endonuclease, as described for RE1. Moreover, the RE2 site must be such that when the PCR product is inserted, a gene is thereby created which is capable of directing the synthesis of a complete IgM heavy chain. This limits the choices for RE2, but the choices available can be determined by computer based sequence analysis. The choices can be determined as follows. First, a list of restriction endonucleases that do not cleave pFHC is compiled (see Table 1). [0069]
    TABLE 1
    Non-Cutting Enzymes for the Mouse Cμ Gene
    AatII AhaII AseI
    AvrII BgII BspHI
    BssHII BstBI ClaI
    DraI EagI EcoRI
    EcoRV FspI HgaI
    HincII HpaI KpnI
    MluI NaeI NarI
    NdeI NotI NruI
    PaeR7I PvuI RsrII
    SacII SaII ScaI
    SfII SnaBI SpeI
    SphI SspI StuI
    TthlllI XbaI XhoI
  • These are called the “rare non-cutters.” Next, the sequence of C[0070] H1 is rewritten with “N” at the third position of each codon and entered into the computer. This is called the “N-doped sequence” (See FIG. 4). Next, the rare non-cutters are surveyed by computer analysis for those which will cleave the N-doped sequence. The search program will show a possible restriction endonuclease site, assuming a match between N and the restriction endonuclease cutting site. For example, with 39 rare non-cutters, 22 will cleave the N-doped sequence of Cμ CH1, many of them several times (see Table 2). In this table, “Def” means a definite cut site, of which there are none, because of the Ns. “Pos” means a possible cleavage site at the indicated nucleotide position if N is chosen appropriately. “Y” indicates any pyrimidine, “R” indicates any purine and “N” indicates any nucleotide. The nucleotide positions refer to coordinates represented in FIG. 4.
    TABLE 2
    ENZYME RECOGNITION CUT SITE
    AatII (GACGTC) Def none
    Pos 250 309
    AhaII (GRCGYC) Def none
    Pos 247 306
    AvrII (CCTAGG) Def none
    Pos 204
    BspHI (TCATGA) Def none
    Pos 138
    BsshII (GCGCGC) Def none
    Pos 189
    EcoRI (GAATTC) Def none
    Pos 195 334
    EcoRV (GATATC) Def none
    Pos 214
    HgaI (GACGCNNNNN) Def none
    (NNNNNNNNNNGCGTC) Pos 284
    HincII (GTYRAC) Def none
    Pos 183 220
    HpaI (GTTAAC) Def none
    Pos 220
    KpnI (GGTACC) Def none
    Pos 408
    NruI (TCGCGA) Def none
    Pos 174 193 303
    PaeR7 (CTCGAG) Def none
    Pos
    190 339
    PvuI (CGATCG) Def none
    Pos 178
    ScaI (AGTACT) Def none
    Pos 209 266 284
    SpeI (ACTAGT) Def none
    Pos 131 167 359
    SphI (GCATGC) Def none
    Pos 338
    SspI (AATATT) Def none
    Pos 371
    StuI (AGGCCT) Def none
    Pos 149
    TthlllI (GACNNNGTC) Def none
    Pos 212
    XbaI (TCTAGA) Def none
    Pos 338
    XhoI (CTCGAG) Def none
    Pos
    190 339
  • Most of these cleavage sites (about 60%) are compatible with the amino acids specified by C[0071] H1. Therefore, it is possible to mutate CH1 to create a unique site for such an enzyme without altering the amino acid sequence incoded by CH1. One sequence which illustrates this is shown below:
    1) . . . ala met gly cys leu ala arg asp . . .
    2) . . . GCC ATG GGC TGC CTA GCC CGG GAC . . .
    3) . . . GCC ATG GGC TGC CTA GCG CGC GAC . . .
                              ---------
                                BssHII
  • Line 1 represents part of the actual amino acid sequence specified by the mouse Cμ C[0072] H1 gene region, and line 2 is the actual nucleotide sequence. By changing the sequence to the indicated nucleotides underlined on line 3, a cleavage site for the rare non-cutter BssHII is created. The new sequence (containing the BssHII site) GCG CGC still encodes the identical amino acid sequence. Therefore, the sequence of the primer C is chosen to be the complement of line 3, and RE2 is the BssHII site. Such a primer will function in the PCR and vector construction as desired. Other examples are possible, and the same process can be used in designing vectors and primers for cloning light chain variable regions.
  • The choice for primer C puts a constraint on pFHC. In the example shown, the C[0073] H1 region contained on pFHC must begin at its 5′ nd with the mutant sequence GCG CGC. Such mutant fragments can b readily made by the process of PCR cassette assembly described below.
  • The process of PCR cassette assembly is a method of constructing plasmid molecules (in this case the plasmid pFHC) from fragments of DNA of known nucleotide sequence. One first compiles a list of restriction endonucleases that do not cleave any of the fragments. Each fragment is then individually PCR amplified using synthesized oligonucleotide primers complementary to the terminal sequences of the fragment. These primers are synthesized to contain on their 5′ ends restriction endonuclease cleavage sites from the compiled list. Thus, each PCR product can be so designed that each fragment can be assembled one by one into a larger plasmid structure by cleavage and ligation and transformation into [0074] E. coli. Using this method, it is also possible to make minor modifications to modify the terminal sequence of the fragment being amplified. This is done by altering the PCR primer slightly so that a mismatch occurs. In this way it is possible to amplify the Cμ gene starting precisely from the desired point in CH1 (as determined by oligo C above) and creating the RE2 endonuclease cleavage site.
  • Equivalents
  • Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described specifically herein. Such equivalents are intended to be encompassed in the scope of the following claims. [0075]
  • 1 6 1 313 DNA Artificial Sequence Synthesized Nucleotide Sequence 1 agt cag tcc ttc cca aat gtc ttc ccc ctc gtc tcc tgc gag agc ccc 48 Ser Gln Ser Phe Pro Asn Val Phe Pro Leu Val Ser Cys Glu Ser Pro 1 5 10 15 ctg tct gat aag aat ctg gtg gcc atg ggc tgc cta gcc cgg gac ttc 96 Leu Ser Asp Lys Asn Leu Val Ala Met Gly Cys Leu Ala Arg Asp Phe 20 25 30 ctg ccc agc acc att tcc ttc acc tgg aac tac cag aac aac act gaa 144 Leu Pro Ser Thr Ile Ser Phe Thr Trp Asn Tyr Gln Asn Asn Thr Glu 35 40 45 gtc atc cag ggt atc aga acc ttc cca aca ctg agg aca ggg ggc aag 192 Val Ile Gln Gly Ile Arg Thr Phe Pro Thr Leu Arg Thr Gly Gly Lys 50 55 60 tac cta gcc acc tcg cag gtg ttg ctg tct ccc aag agc atc ctt gaa 240 Tyr Leu Ala Thr Ser Gln Val Leu Leu Ser Pro Lys Ser Ile Leu Glu 65 70 75 80 ggt tca gat gaa tac ctg gta tgc aaa atc cac tac gga ggc aaa aac 288 Gly Ser Asp Glu Tyr Leu Val Cys Lys Ile His Tyr Gly Gly Lys Asn 85 90 95 aga gat ctg cat gtg ccc att cca g 313 Arg Asp Leu His Val Pro Ile Pro 100 2 104 PRT Artificial Sequence Synthesized Peptide 2 Ser Gln Ser Phe Pro Asn Val Phe Pro Leu Val Ser Cys Glu Ser Pro 1 5 10 15 Leu Ser Asp Lys Asn Leu Val Ala Met Gly Cys Leu Ala Arg Asp Phe 20 25 30 Leu Pro Ser Thr Ile Ser Phe Thr Trp Asn Tyr Gln Asn Asn Thr Glu 35 40 45 Val Ile Gln Gly Ile Arg Thr Phe Pro Thr Leu Arg Thr Gly Gly Lys 50 55 60 Tyr Leu Ala Thr Ser Gln Val Leu Leu Ser Pro Lys Ser Ile Leu Glu 65 70 75 80 Gly Ser Asp Glu Tyr Leu Val Cys Lys Ile His Tyr Gly Gly Lys Asn 85 90 95 Arg Asp Leu His Val Pro Ile Pro 100 3 313 DNA Artificial Sequence Synthesized Nucleotide Sequence 3 agncantcnt tnccnaangt nttnccnctn gtntcntgng anagnccnct ntcnganaan 60 aanctngtng cnatngcntg nctngcncgn ganttnctnc cnagnacnat ntcnttnacn 120 tgnaantanc anaanaanac ngangtnatn canggnatna gnacnttncc nacnctnagn 180 acnggnggna antanctngc nacntcncan gtnttnctnt cnccnaanag natnctngan 240 ggntcngang antanctngt ntgnaanatn cantanggng gnaanaanag nganctncan 300 gtnccnatnc cng 313 4 8 PRT Artificial Sequence Synthesized Nucleotide Sequence 4 Ala Met Gly Cys Leu Ala Arg Asp 1 5 5 24 DNA Artificial Sequence Synthesized Nucleotide Sequence 5 gccatgggct gcctagcccg ggac 24 6 24 DNA Artificial Sequence Synthesized Nucleotide Sequence 6 gccatgggct gcctagcgcg cgac 24

Claims (17)

We claim:
1. A host cell producing a DNA encoding an antigen-combining protein produced by the method of:
providing DNA containing genes encoding antigen-combining proteins;
inserting said DNA into a framework antibody vector; and
introducing said DNA and antibody framework vector into a host cell.
2. The host cell of claim 1, wherein the host cell is prokaryotic.
3. The host cell of claim 1, wherein the host cell is eukaryotic.
4. The host cell of claim 3, wherein the host cell is an immortalized cultured mammalian cell.
5. The host cell of claim 4, wherein the immortalized cultured mammalian cell is a myeloma or plasmacytoma cell.
6. The host cell of Claim 1 wherein said DNA is introduced into the host cell by a method selected from the group consisting of: electroporation, calcium phosphate coprecipitation, protoplast fusion, viral infection, and cell fusion.
7. The host cell of claim 1, wherein the DNA containing genes encoding antigen-combining proteins encodes an antigen-combining protein selected from the group consisting of:
an immunoglobulin heavy chain variable region and an immunoglobulin light chain variable region.
8. The host cell of claim 1, wherein the antigen-combining proteins comprise antibodies.
9. Host cells producing antibodies, produced by a method comprising:
providing DNA comprising genes encoding antibodies;
inserting the DNA into framework antibody vectors; and
introducing said framework antibody vectors into host cells.
10. The host cells of claim 9, wherein said DNA comprises a vector.
11. The host cells of claim 10, wherein said vector is an expression vector.
12. The host cells of claim 9, wherein the host cells are prokaryotic.
13. The host cells of claim 9, wherein the host cells are eukaryotic.
14. The host cells of claim 13, wherein the host cells are an immortalized cultured mammalian cell line.
15. The host cells of claim 14, wherein the immortalized cultured mammalian cell line is a myeloma or plasmacytoma cell line.
16. The host cells of claim 9, wherein said DNA is introduced into the host cells by a method selected from the group consisting of: electroporation, calcium phosphate coprecipitation, protoplast fusion, viral infection, and cell fusion.
17. The host cells of claim 9, wherein the DNA containing genes encoding antibodies encodes immunoglobulin heavy chain variable regions or immunoglobulin light chain variable regions.
US10/690,396 1990-01-11 2003-10-20 Method for generating libraries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules Abandoned US20040096956A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/690,396 US20040096956A1 (en) 1990-01-11 2003-10-20 Method for generating libraries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US46435090A 1990-01-11 1990-01-11
WOPCT/US91/00209 1991-01-10
PCT/US1991/000209 WO1991010737A1 (en) 1990-01-11 1991-01-10 Production of antibodies using gene libraries
US07/919,730 US5284555A (en) 1991-08-15 1992-07-24 Process for preparing organophosphines
US08/315,269 US5780225A (en) 1990-01-12 1994-09-29 Method for generating libaries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules
US99719597A 1997-12-23 1997-12-23
US09/439,732 US6303313B1 (en) 1990-01-11 1999-11-12 Method for generating libraries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules
US09/798,720 US6635424B2 (en) 1990-01-11 2001-03-02 Method for generating libraries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules
US10/690,396 US20040096956A1 (en) 1990-01-11 2003-10-20 Method for generating libraries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/798,720 Continuation US6635424B2 (en) 1990-01-11 2001-03-02 Method for generating libraries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules

Publications (1)

Publication Number Publication Date
US20040096956A1 true US20040096956A1 (en) 2004-05-20

Family

ID=23843589

Family Applications (4)

Application Number Title Priority Date Filing Date
US09/439,732 Expired - Fee Related US6303313B1 (en) 1990-01-11 1999-11-12 Method for generating libraries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules
US09/798,720 Expired - Fee Related US6635424B2 (en) 1990-01-11 2001-03-02 Method for generating libraries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules
US09/800,229 Expired - Fee Related US6479243B1 (en) 1990-01-11 2001-03-05 Method for generating libraries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules
US10/690,396 Abandoned US20040096956A1 (en) 1990-01-11 2003-10-20 Method for generating libraries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US09/439,732 Expired - Fee Related US6303313B1 (en) 1990-01-11 1999-11-12 Method for generating libraries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules
US09/798,720 Expired - Fee Related US6635424B2 (en) 1990-01-11 2001-03-02 Method for generating libraries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules
US09/800,229 Expired - Fee Related US6479243B1 (en) 1990-01-11 2001-03-05 Method for generating libraries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules

Country Status (3)

Country Link
US (4) US6303313B1 (en)
AU (1) AU7247191A (en)
WO (1) WO1991010737A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090318308A1 (en) * 2006-05-30 2009-12-24 Millegen Highly diversified antibody libraries

Families Citing this family (509)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991010737A1 (en) 1990-01-11 1991-07-25 Molecular Affinities Corporation Production of antibodies using gene libraries
ATE449853T1 (en) 1990-02-01 2009-12-15 Siemens Healthcare Diagnostics PRODUCTION AND USE OF HUMAN ANTIBODIES GENE BANKS (ßHUMAN ANTIBODIES LIBRARIESß)
US5427908A (en) * 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
GB9015198D0 (en) * 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US7063943B1 (en) 1990-07-10 2006-06-20 Cambridge Antibody Technology Methods for producing members of specific binding pairs
US6916605B1 (en) 1990-07-10 2005-07-12 Medical Research Council Methods for producing members of specific binding pairs
US6172197B1 (en) 1991-07-10 2001-01-09 Medical Research Council Methods for producing members of specific binding pairs
GB9206318D0 (en) * 1992-03-24 1992-05-06 Cambridge Antibody Tech Binding substances
US5962255A (en) * 1992-03-24 1999-10-05 Cambridge Antibody Technology Limited Methods for producing recombinant vectors
US6225447B1 (en) 1991-05-15 2001-05-01 Cambridge Antibody Technology Ltd. Methods for producing members of specific binding pairs
DE69230142T2 (en) 1991-05-15 2000-03-09 Cambridge Antibody Tech METHOD FOR PRODUCING SPECIFIC BINDING PAIRS
US6492160B1 (en) 1991-05-15 2002-12-10 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
US5858657A (en) * 1992-05-15 1999-01-12 Medical Research Council Methods for producing members of specific binding pairs
US5270170A (en) * 1991-10-16 1993-12-14 Affymax Technologies N.V. Peptide library and screening method
US5733731A (en) * 1991-10-16 1998-03-31 Affymax Technologies N.V. Peptide library and screening method
PT1696031E (en) 1991-12-02 2010-06-25 Medical Res Council Production of anti-self antibodies from antibody segment repertoires and displayed on phage
US5733743A (en) * 1992-03-24 1998-03-31 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
DK0744958T3 (en) 1994-01-31 2003-10-20 Univ Boston Polyclonal antibody libraries
DK1143006T3 (en) * 1995-08-18 2008-07-14 Morphosys Ip Gmbh Vectors / DNA sequences from human combinatorial antibody libraries
US6828422B1 (en) 1995-08-18 2004-12-07 Morphosys Ag Protein/(poly)peptide libraries
US7368111B2 (en) 1995-10-06 2008-05-06 Cambridge Antibody Technology Limited Human antibodies specific for TGFβ2
US7888466B2 (en) 1996-01-11 2011-02-15 Human Genome Sciences, Inc. Human G-protein chemokine receptor HSATU68
DE19739685A1 (en) 1997-09-10 1999-03-11 Eichel Streiber Christoph Von Monoclonal antibodies for the therapy and prophylaxis of diseases caused by Clostridium difficile
CA2323776C (en) 1998-03-19 2010-04-27 Human Genome Sciences, Inc. Cytokine receptor common gamma chain like
CA2363779A1 (en) 1999-02-26 2000-08-31 Human Genome Sciences, Inc. Human endokine alpha and methods of use
US6492497B1 (en) 1999-04-30 2002-12-10 Cambridge Antibody Technology Limited Specific binding members for TGFbeta1
DK1488806T3 (en) * 1999-06-01 2016-02-08 Biogen Ma Inc Blocking monoclonal antibody to VLA-1 and its use in the treatment of vascular disorders
US7291714B1 (en) 1999-06-30 2007-11-06 Millennium Pharmaceuticals, Inc. Glycoprotein VI and uses thereof
US20040001826A1 (en) 1999-06-30 2004-01-01 Millennium Pharmaceuticals, Inc. Glycoprotein VI and uses thereof
JP4656478B2 (en) * 2000-02-22 2011-03-23 株式会社医学生物学研究所 Antibody library
EP1274720A4 (en) 2000-04-12 2004-08-18 Human Genome Sciences Inc Albumin fusion proteins
US20030031675A1 (en) 2000-06-06 2003-02-13 Mikesell Glen E. B7-related nucleic acids and polypeptides useful for immunomodulation
EP1294949A4 (en) 2000-06-15 2004-08-25 Human Genome Sciences Inc Human tumor necrosis factor delta and epsilon
ATE494304T1 (en) 2000-06-16 2011-01-15 Human Genome Sciences Inc IMMUNE-SPECIFIC BINDING ANTIBODIES AGAINST BLYS
GB0022978D0 (en) 2000-09-19 2000-11-01 Oxford Glycosciences Uk Ltd Detection of peptides
CA2429544C (en) * 2000-11-17 2010-10-19 University Of Rochester In vitro methods of producing and identifying immunoglobulin molecules in eukaryotic cells
US20050196755A1 (en) * 2000-11-17 2005-09-08 Maurice Zauderer In vitro methods of producing and identifying immunoglobulin molecules in eukaryotic cells
TWI327599B (en) 2000-11-28 2010-07-21 Medimmune Llc Methods of administering/dosing anti-rsv antibodies for prophylaxis and treatment
PT1355919E (en) 2000-12-12 2011-03-02 Medimmune Llc Molecules with extended half-lives, compositions and uses thereof
WO2002064612A2 (en) 2001-02-09 2002-08-22 Human Genome Sciences, Inc. Human g-protein chemokine receptor (ccr5) hdgnr10
US8981061B2 (en) 2001-03-20 2015-03-17 Novo Nordisk A/S Receptor TREM (triggering receptor expressed on myeloid cells) and uses thereof
US8231878B2 (en) 2001-03-20 2012-07-31 Cosmo Research & Development S.P.A. Receptor trem (triggering receptor expressed on myeloid cells) and uses thereof
JP4303475B2 (en) * 2001-04-13 2009-07-29 バイオジェン・アイデック・エムエイ・インコーポレイテッド Antibody against VLA-1
CA2444632A1 (en) 2001-04-13 2002-10-24 Human Genome Sciences, Inc. Vascular endothelial growth factor 2
JP4309758B2 (en) 2001-05-25 2009-08-05 ヒューマン ジノーム サイエンシーズ, インコーポレイテッド Antibodies that immunospecifically bind to TRAIL receptors
US6867189B2 (en) 2001-07-26 2005-03-15 Genset S.A. Use of adipsin/complement factor D in the treatment of metabolic related disorders
EP2277888A3 (en) 2001-12-21 2011-04-27 Human Genome Sciences, Inc. Fusion proteins of albumin and erythropoietin
US7244592B2 (en) 2002-03-07 2007-07-17 Dyax Corp. Ligand screening and discovery
CN1646160A (en) 2002-03-13 2005-07-27 拜奥根Idec马萨诸塞公司 Anti-alpha V beta 6 antibodies
GB0207533D0 (en) 2002-04-02 2002-05-08 Oxford Glycosciences Uk Ltd Protein
EP2270049A3 (en) 2002-04-12 2011-03-09 Medimmune, Inc. Recombinant anti-interleukin-9-antibody
CN100418981C (en) 2002-06-10 2008-09-17 瓦西尼斯公司 Gene differentially expressed in breast and bladder cancer and encoded polypeptides
US7425618B2 (en) 2002-06-14 2008-09-16 Medimmune, Inc. Stabilized anti-respiratory syncytial virus (RSV) antibody formulations
WO2004016750A2 (en) 2002-08-14 2004-02-26 Macrogenics, Inc. FcϜRIIB-SPECIFIC ANTIBODIES AND METHODS OF USE THEREOF
HUE034378T2 (en) 2002-10-16 2018-02-28 Purdue Pharma Lp Antibodies that bind cell-associated CA 125/O722P and methods of use thereof
DE10303974A1 (en) 2003-01-31 2004-08-05 Abbott Gmbh & Co. Kg Amyloid β (1-42) oligomers, process for their preparation and their use
EP2289559B1 (en) 2003-02-20 2014-02-12 Seattle Genetics, Inc. Anit-CD70 antibody-drug conjugates and their use for the treatment of cancer and immune disorders
GEP20094629B (en) 2003-03-19 2009-03-10 Biogen Idec Inc Nogo receptor binding protein
KR20110094361A (en) 2003-04-11 2011-08-23 메디뮨 엘엘씨 Recombinant il-9 antibodies and uses thereof
US7393534B2 (en) 2003-07-15 2008-07-01 Barros Research Institute Compositions and methods for immunotherapy of cancer and infectious diseases
WO2005014795A2 (en) 2003-08-08 2005-02-17 Genenews Inc. Osteoarthritis biomarkers and uses thereof
ES2458636T3 (en) 2003-08-18 2014-05-06 Medimmune, Llc Humanization of antibodies
IL158287A0 (en) 2003-10-07 2004-05-12 Yeda Res & Dev Antibodies to nik, their preparation and use
HUE026260T2 (en) 2003-11-21 2016-06-28 Ucb Biopharma Sprl Method for the treatment of multiple sclerosis by inhibiting IL-17 activity
GB0329825D0 (en) 2003-12-23 2004-01-28 Celltech R&D Ltd Biological products
KR101184391B1 (en) 2004-02-09 2013-03-14 휴먼 게놈 사이언시즈, 인코포레이티드 Albumin fusion proteins
EP1786463A4 (en) 2004-03-26 2009-05-20 Human Genome Sciences Inc Antibodies against nogo receptor
CA2955027A1 (en) 2004-04-15 2005-11-10 University Of Florida Research Foundation, Inc. Neural proteins as biomarkers for nervous system injury and other neural disorders
ES2442386T3 (en) 2004-04-23 2014-02-11 Bundesrepublik Deutschland Letztvertreten Durch Das Robert Koch-Institut Vertreten Durch Seinen Pr Method for the treatment of conditions mediated by T cells by the decrease of positive ICOS cells in vivo.
WO2006002437A2 (en) 2004-06-24 2006-01-05 Biogen Idec Ma Inc. Treatment of conditions involving demyelination
EP2329714A1 (en) 2004-08-03 2011-06-08 Biogen Idec MA Inc. Influence of TAJ in the neuronal functions
US7700720B2 (en) 2004-09-21 2010-04-20 Medimmune, Llc Antibodies against and methods for producing vaccines for respiratory syncytial virus
WO2006047417A2 (en) 2004-10-21 2006-05-04 University Of Florida Research Foundation, Inc. Detection of cannabinoid receptor biomarkers and uses thereof
AU2005299355A1 (en) 2004-10-27 2006-05-04 Medimmune, Llc Modulation of antibody specificity by tailoring the affinity to cognate antigens
GB0426146D0 (en) 2004-11-29 2004-12-29 Bioxell Spa Therapeutic peptides and method
CN103497993A (en) 2005-02-07 2014-01-08 基因信息公司 Mild osteoarthritis biomarkers and uses thereof
WO2006089133A2 (en) 2005-02-15 2006-08-24 Duke University Anti-cd19 antibodies and uses in oncology
JP2008531730A (en) 2005-03-04 2008-08-14 キュアーディーエム、インク. Methods and pharmaceutical compositions for treating type I diabetes mellitus and other conditions
JP5153613B2 (en) 2005-03-18 2013-02-27 メディミューン,エルエルシー Antibody framework shuffle
GB0506912D0 (en) 2005-04-05 2005-05-11 Celltech R&D Ltd Biological products
EP3479844B1 (en) 2005-04-15 2023-11-22 MacroGenics, Inc. Covalent diabodies and uses thereof
AU2006236225C1 (en) 2005-04-19 2013-05-02 Seagen Inc. Humanized anti-CD70 binding agents and uses thereof
EP2221316A1 (en) 2005-05-05 2010-08-25 Duke University Anti-CD19 antibody therapy for autoimmune disease
WO2006128083A2 (en) 2005-05-25 2006-11-30 Curedm, Inc. Human proislet peptide, derivatives and analogs thereof, and methods of using same
KR20080025174A (en) 2005-06-23 2008-03-19 메디뮨 인코포레이티드 Antibody formulations having optimized aggregation and fragmentation profiles
EP1907421A4 (en) 2005-06-30 2012-03-28 Abbott Lab Il-12/p40 binding proteins
US7482124B2 (en) 2005-07-08 2009-01-27 Bristol-Myers Squibb Company Method of identifying a PPARgamma-agonist compound having a decreased likelihood of inducing dose-dependent peripheral edema
CN104072614B (en) 2005-07-08 2017-04-26 生物基因Ma公司 Anti-alpha[v]beta[6] antibodies and uses thereof
EP2238986A3 (en) 2005-07-08 2010-11-03 Biogen Idec MA Inc. Sp35 antibodies and uses thereof
NZ565631A (en) 2005-08-03 2011-01-28 Adelaide Res & Innovation Pty Polysaccharide synthases
SG2014010029A (en) 2005-08-19 2014-08-28 Abbott Lab Dual variable domain immunoglobin and uses thereof
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
EP2500355A3 (en) 2005-08-19 2012-10-24 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US8906864B2 (en) 2005-09-30 2014-12-09 AbbVie Deutschland GmbH & Co. KG Binding domains of proteins of the repulsive guidance molecule (RGM) protein family and functional fragments thereof, and their use
EP1945816B1 (en) 2005-10-21 2011-07-27 GeneNews Inc. Method and apparatus for correlating levels of biomarker products with disease
CA2628451A1 (en) 2005-11-04 2007-05-18 Biogen Idec Ma Inc. Methods for promoting neurite outgrowth and survival of dopaminergic neurons
US20100028358A1 (en) 2005-11-07 2010-02-04 Wolfram Ruf Compositions and Methods for Controlling Tissue Factor Signaling Specificity
WO2007111714A2 (en) 2005-11-28 2007-10-04 Zymogenetics, Inc. Il-21 antagonists
RS53270B2 (en) 2005-11-30 2018-05-31 Abbvie Deutschland Monoclonal antibodies against amyloid beta protein and uses thereof
AU2006319358B2 (en) 2005-11-30 2012-01-19 AbbVie Deutschland GmbH & Co. KG Anti-Abeta globulomer antibodies, antigen-binding moieties thereof, corresponding hybridomas, nucleic acids, vectors, host cells, methods of producing said antibodies, compositions comprising said antibodies, uses of said antibodies and methods of using said antibodies
CA2631181A1 (en) 2005-12-02 2007-06-07 Biogen Idec Ma Inc. Treatment of conditions involving demyelination
JP5183484B2 (en) 2005-12-09 2013-04-17 ユセベ ファルマ ソシエテ アノニム Antibody molecule having specificity for human IL-6
CN100366752C (en) * 2005-12-28 2008-02-06 河北省农林科学院遗传生理研究所 Method for deriving mulriple DNA sequences from atom destructurizing
JP5829373B2 (en) 2006-01-27 2015-12-09 バイオジェン・エムエイ・インコーポレイテッドBiogen MA Inc. Nogo receptor antagonist
EP2650306A1 (en) 2006-03-06 2013-10-16 Aeres Biomedical Limited Humanized Anti-CD22 antibodies and their use in treatment of oncology, transplantation and autoimmune disease
EP2034830B1 (en) * 2006-05-25 2014-09-03 Biogen Idec MA Inc. Anti-vla-1 antibody for treating stroke
GB0611116D0 (en) 2006-06-06 2006-07-19 Oxford Genome Sciences Uk Ltd Proteins
BRPI0713426A2 (en) 2006-06-14 2012-10-09 Macrogenics Inc methods of treating, slowing the progression, or ameliorating one or more symptoms of a disorder, and preventing or delaying the onset of a disorder
WO2008019199A2 (en) 2006-06-26 2008-02-14 Macrogenics, Inc. FCγRIIB-SPECIFIC ANTIBODIES AND METHODS OF USE THEREOF
US7572618B2 (en) 2006-06-30 2009-08-11 Bristol-Myers Squibb Company Polynucleotides encoding novel PCSK9 variants
EP2046374A4 (en) 2006-07-10 2010-05-05 Biogen Idec Inc Compositions and methods for inhibiting growth of smad4-deficient cancers
BRPI0714871A2 (en) 2006-07-18 2013-05-07 Sanofi Aventis antagonist antibody for cancer treatment
KR101314362B1 (en) 2006-08-28 2013-10-10 라 졸라 인스티튜트 포 앨러지 앤드 이뮤놀로지 Antagonistic human light-specific human monoclonal antibodies
MY188368A (en) 2006-09-08 2021-12-06 Abbott Lab Interleukin-13 binding proteins
US20100143254A1 (en) 2006-10-16 2010-06-10 Medimmune, Llc Molecules with reduced half-lives, compositions and uses thereof
GB0620729D0 (en) 2006-10-18 2006-11-29 Ucb Sa Biological products
EP1914242A1 (en) 2006-10-19 2008-04-23 Sanofi-Aventis Novel anti-CD38 antibodies for the treatment of cancer
CA2669205A1 (en) 2006-11-09 2008-05-15 Irm Llc Agonist trkb antibodies and uses thereof
US8785400B2 (en) 2006-11-22 2014-07-22 Curedm Group Holdings, Llc Methods and compositions relating to islet cell neogenesis
US8455626B2 (en) 2006-11-30 2013-06-04 Abbott Laboratories Aβ conformer selective anti-aβ globulomer monoclonal antibodies
EP2609932B1 (en) 2006-12-01 2022-02-02 Seagen Inc. Variant target binding agents and uses thereof
EP2687232A1 (en) 2006-12-06 2014-01-22 MedImmune, LLC Methods of treating systemic lupus erythematosus
TR201807425T4 (en) 2007-01-09 2018-06-21 Biogen Ma Inc Sp35 antibodies and their use.
US8128926B2 (en) 2007-01-09 2012-03-06 Biogen Idec Ma Inc. Sp35 antibodies and uses thereof
US8114606B2 (en) 2007-02-16 2012-02-14 The Board Of Trustees Of Southern Illinois University ARL-1 specific antibodies
US8685666B2 (en) 2007-02-16 2014-04-01 The Board Of Trustees Of Southern Illinois University ARL-1 specific antibodies and uses thereof
EP3118221B1 (en) 2007-02-26 2019-08-21 Oxford BioTherapeutics Ltd Proteins
WO2008104803A2 (en) 2007-02-26 2008-09-04 Oxford Genome Sciences (Uk) Limited Proteins
EP2486928A1 (en) 2007-02-27 2012-08-15 Abbott GmbH & Co. KG Method for the treatment of amyloidoses
CN103214577B (en) 2007-03-22 2015-09-02 生物基因Ma公司 Specific binding CD154 comprises associated proteins of antibody, antibody derivatives and antibody fragment and uses thereof
KR101540823B1 (en) 2007-03-30 2015-07-30 메디뮨 엘엘씨 Antibody formulation
EP2164868B1 (en) 2007-05-04 2015-03-25 Technophage, Investigação E Desenvolvimento Em Biotecnologia, SA Engineered rabbit antibody variable domains and uses thereof
RU2549701C2 (en) 2007-05-07 2015-04-27 Медиммун, Ллк Anti-icos antibodies and their application in treatment of oncological, transplantation-associated and autoimmune diseases
PL2068927T3 (en) 2007-05-14 2016-06-30 Medimmune Llc Methods of reducing eosinophil levels
EP2158221B1 (en) 2007-06-21 2018-08-29 MacroGenics, Inc. Covalent diabodies and uses thereof
PT2193142E (en) 2007-08-30 2015-04-22 Curedm Group Holdings Llc Compositions and methods of using proislet peptides and analogs thereof
GB0717337D0 (en) 2007-09-06 2007-10-17 Ucb Pharma Sa Method of treatment
ES2622460T3 (en) 2007-09-26 2017-07-06 Ucb Biopharma Sprl Fusions of antibodies with double specificity
CA2703705A1 (en) 2007-11-05 2009-05-14 Medimmune, Llc Methods of treating scleroderma
WO2009067546A2 (en) 2007-11-19 2009-05-28 Celera Corpration Lung cancer markers and uses thereof
EP2225275A4 (en) 2007-11-28 2013-04-03 Medimmune Llc Protein formulation
FR2924440B1 (en) * 2007-12-04 2015-01-09 Pf Medicament NEW METHOD FOR GENERATING AND SCREENING AN ANTIBODY BANK
GB0800277D0 (en) 2008-01-08 2008-02-13 Imagination Tech Ltd Video motion compensation
CA2711771C (en) 2008-01-11 2017-01-24 Gene Techno Science Co., Ltd. Humanized anti-.alpha.9 integrin antibodies and the uses thereof
ES2613963T3 (en) 2008-01-18 2017-05-29 Medimmune, Llc Cysteine manipulated antibodies for site specific conjugation
PT2250279E (en) 2008-02-08 2016-06-03 Medimmune Llc Anti-ifnar1 antibodies with reduced fc ligand affinity
US8962803B2 (en) 2008-02-29 2015-02-24 AbbVie Deutschland GmbH & Co. KG Antibodies against the RGM A protein and uses thereof
EP2098536A1 (en) 2008-03-05 2009-09-09 4-Antibody AG Isolation and identification of antigen- or ligand-specific binding proteins
US20110020368A1 (en) 2008-03-25 2011-01-27 Nancy Hynes Treating cancer by down-regulating frizzled-4 and/or frizzled-1
BRPI0906309A2 (en) 2008-04-02 2020-05-26 Macrogenics, Inc IMMUNOGLOBULIN, ANTIBODY, USE OF ANTIBODY AND PHARMACEUTICAL COMPOSITION
ES2654937T3 (en) 2008-04-02 2018-02-15 Macrogenics, Inc. Specific antibodies for the BCR complex and procedures for their use
SG189776A1 (en) 2008-04-11 2013-05-31 Seattle Genetics Inc Detection and treatment of pancreatic, ovarian and other cancers
GB0807413D0 (en) 2008-04-23 2008-05-28 Ucb Pharma Sa Biological products
WO2009131256A1 (en) 2008-04-24 2009-10-29 Gene Techno Science Co., Ltd. Humanized antibodies specific for amino acid sequence rgd of an extracellular matrix protein and the uses thereof
EP2282769A4 (en) 2008-04-29 2012-04-25 Abbott Lab Dual variable domain immunoglobulins and uses thereof
EP2282773B1 (en) 2008-05-02 2014-01-15 Seattle Genetics, Inc. Methods and compositions for making antibodies and antibody derivatives with reduced core fucosylation
ES2579554T3 (en) 2008-05-09 2016-08-12 Abbvie Deutschland Gmbh & Co Kg Antibodies for the recipient of advanced glycation terminal products (RAGE) and uses thereof
EP2304439A4 (en) 2008-05-29 2012-07-04 Nuclea Biotechnologies Llc Anti-phospho-akt antibodies
CA2725666A1 (en) 2008-06-03 2009-12-10 Abbott Laboratories Dual variable domain immunoglobulins and uses thereof
RU2010153580A (en) 2008-06-03 2012-07-20 Эбботт Лэборетриз (Us) IMMUNOGLOBULINS WITH TWO VARIABLE DOMAINS AND THEIR APPLICATION
WO2010033279A2 (en) 2008-06-04 2010-03-25 Macrogenics, Inc. Antibodies with altered binding to fcrn and methods of using same
JP5674654B2 (en) 2008-07-08 2015-02-25 アッヴィ・インコーポレイテッド Prostaglandin E2 double variable domain immunoglobulin and use thereof
CN104829718A (en) 2008-07-08 2015-08-12 艾伯维公司 Prostaglandin E2 binding proteins and uses thereof
US8058406B2 (en) 2008-07-09 2011-11-15 Biogen Idec Ma Inc. Composition comprising antibodies to LINGO or fragments thereof
WO2010048615A2 (en) 2008-10-24 2010-04-29 The Government Of The United States Of America As Represented By The Secretary, Department Of Health & Human Services, Center For Disease Control And Prevention Human ebola virus species and compositions and methods thereof
ES2719496T3 (en) 2008-11-12 2019-07-10 Medimmune Llc Antibody formulation
KR101940059B1 (en) 2008-12-19 2019-01-18 마크로제닉스, 인크. Covalent diabodies and uses thereof
CA2748757A1 (en) 2008-12-31 2010-07-08 Biogen Idec Ma Inc. Anti-lymphotoxin antibodies
GB0900425D0 (en) 2009-01-12 2009-02-11 Ucb Pharma Sa Biological products
WO2010082134A1 (en) 2009-01-14 2010-07-22 Iq Therapeutics Bv Combination antibodies for the treatment and prevention of disease caused by bacillus anthracis and related bacteria and their toxins
BRPI1007345A2 (en) 2009-01-20 2019-04-16 H. ZADEH Homayoun mediated antibody bone regeneration
AU2010207552A1 (en) 2009-01-21 2011-09-01 Oxford Biotherapeutics Ltd. PTA089 protein
UA102722C2 (en) 2009-01-29 2013-08-12 Эббви Инк. Il-1 binding proteins
US8852608B2 (en) 2009-02-02 2014-10-07 Medimmune, Llc Antibodies against and methods for producing vaccines for respiratory syncytial virus
WO2010093993A2 (en) 2009-02-12 2010-08-19 Human Genome Sciences, Inc. Use of b lymphocyte stimulator protein antagonists to promote transplantation tolerance
PE20120211A1 (en) 2009-02-17 2012-03-24 Ucb Pharma Sa ANTIBODIES THAT HAVE HUMAN OX40 SPECIFICITY
US8835610B2 (en) 2009-03-05 2014-09-16 Abbvie Inc. IL-17 binding proteins
EP2405920A1 (en) 2009-03-06 2012-01-18 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Novel therapy for anxiety
EP2406285B1 (en) 2009-03-10 2016-03-09 Gene Techno Science Co., Ltd. Generation, expression and characterization of the humanized k33n monoclonal antibody
GB0904214D0 (en) 2009-03-11 2009-04-22 Ucb Pharma Sa Biological products
EP2241323A1 (en) 2009-04-14 2010-10-20 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Tenascin-W and brain cancers
SG10201401604VA (en) 2009-04-20 2014-08-28 Oxford Biotherapeutics Ltd Antibodies Specific To Cadherin-17
JP5797642B2 (en) * 2009-05-20 2015-10-21 ノビミューン エスアー Synthetic polypeptide libraries and methods for generating naturally diversified polypeptide variants
DK2435568T3 (en) 2009-05-29 2014-09-08 Morphosys Ag Collection of synthetic antibodies to treat disease
BRPI1015234A2 (en) 2009-06-22 2018-02-20 Medimmune Llc fc regions designed for site specific conjugation.
ES2553440T3 (en) 2009-08-13 2015-12-09 Crucell Holland B.V. Antibodies against human respiratory syncytial virus (RSV) and method of use
EP2292266A1 (en) 2009-08-27 2011-03-09 Novartis Forschungsstiftung, Zweigniederlassung Treating cancer by modulating copine III
PE20121647A1 (en) 2009-08-29 2012-12-31 Abbvie Inc THERAPEUTIC BINDING PROTEINS TO DLL4
TW201119673A (en) 2009-09-01 2011-06-16 Abbott Lab Dual variable domain immunoglobulins and uses thereof
US20120283415A1 (en) 2009-09-10 2012-11-08 Ucb Pharma S.A. Multivalent Antibodies
US20120244170A1 (en) 2009-09-22 2012-09-27 Rafal Ciosk Treating cancer by modulating mex-3
GB201005063D0 (en) 2010-03-25 2010-05-12 Ucb Pharma Sa Biological products
AR078470A1 (en) 2009-10-02 2011-11-09 Sanofi Aventis ANTIBODIES THAT SPECIFICALLY JOIN THE EPHA2 RECEIVER
WO2011047083A1 (en) 2009-10-13 2011-04-21 Oxford Biotherapeutics Ltd. Antibodies against epha10
TW201119676A (en) 2009-10-15 2011-06-16 Abbott Lab Dual variable domain immunoglobulins and uses thereof
WO2011045352A2 (en) 2009-10-15 2011-04-21 Novartis Forschungsstiftung Spleen tyrosine kinase and brain cancers
GB0922434D0 (en) 2009-12-22 2010-02-03 Ucb Pharma Sa antibodies and fragments thereof
WO2011051350A1 (en) 2009-10-27 2011-05-05 Ucb Pharma S.A. Function modifying nav 1.7 antibodies
GB0922435D0 (en) 2009-12-22 2010-02-03 Ucb Pharma Sa Method
US9234037B2 (en) 2009-10-27 2016-01-12 Ucb Biopharma Sprl Method to generate antibodies to ion channels
UY32979A (en) 2009-10-28 2011-02-28 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
US20120213801A1 (en) 2009-10-30 2012-08-23 Ekaterina Gresko Phosphorylated Twist1 and cancer
WO2011053707A1 (en) 2009-10-31 2011-05-05 Abbott Laboratories Antibodies to receptor for advanced glycation end products (rage) and uses thereof
US20120282177A1 (en) 2009-11-02 2012-11-08 Christian Rohlff ROR1 as Therapeutic and Diagnostic Target
US9244063B2 (en) 2009-11-11 2016-01-26 Gentian As Immunoassay for assessing related analytes of different origin
GB0920127D0 (en) 2009-11-17 2009-12-30 Ucb Pharma Sa Antibodies
GB0920324D0 (en) 2009-11-19 2010-01-06 Ucb Pharma Sa Antibodies
CA2780069C (en) 2009-12-08 2018-07-17 Abbott Gmbh & Co. Kg Monoclonal antibodies against the rgm a protein for use in the treatment of retinal nerve fiber layer degeneration
GB201000467D0 (en) 2010-01-12 2010-02-24 Ucb Pharma Sa Antibodies
SG10201501562VA (en) 2010-03-02 2015-04-29 Abbvie Inc Therapeutic dll4 binding proteins
US20130004519A1 (en) 2010-03-05 2013-01-03 Ruth Chiquet-Ehrismann Smoci, tenascin-c and brain cancers
GB201005064D0 (en) 2010-03-25 2010-05-12 Ucb Pharma Sa Biological products
US10472426B2 (en) 2010-03-25 2019-11-12 Ucb Biopharma Sprl Disulfide stabilized DVD-Ig molecules
MX336196B (en) 2010-04-15 2016-01-11 Abbvie Inc Amyloid-beta binding proteins.
US20130034543A1 (en) 2010-04-19 2013-02-07 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Resear Modulating xrn1
EP2380909A1 (en) 2010-04-26 2011-10-26 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. PTK-7 protein involved in breast cancer
SI2563813T1 (en) 2010-04-30 2015-12-31 Alexion Pharmaceuticals, Inc. Anti-c5a antibodies and methods for using the antibodies
US20110293629A1 (en) 2010-05-14 2011-12-01 Bastid Jeremy Methods of Treating and/or Preventing Cell Proliferation Disorders with IL-17 Antagonists
HUE033063T2 (en) 2010-05-14 2017-11-28 Abbvie Inc Il-1 binding proteins
WO2011154485A1 (en) 2010-06-10 2011-12-15 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Treating cancer by modulating mammalian sterile 20-like kinase 3
US20120009196A1 (en) 2010-07-08 2012-01-12 Abbott Laboratories Monoclonal antibodies against hepatitis c virus core protein
UY33492A (en) 2010-07-09 2012-01-31 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
AR082149A1 (en) 2010-07-09 2012-11-14 Calmune Corp ANTIBODIES AGAINST HUMAN RESPIRATORY SYNTHETIC VIRUS (RSV) AND METHODS FOR USE
SG187682A1 (en) 2010-08-02 2013-03-28 Macrogenics Inc Covalent diabodies and uses thereof
CA2807014A1 (en) 2010-08-03 2012-02-09 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
WO2012024187A1 (en) 2010-08-14 2012-02-23 Abbott Laboratories Amyloid-beta binding proteins
US9505829B2 (en) 2010-08-19 2016-11-29 Zoetis Belgium S.A. Anti-NGF antibodies and their use
GB201014033D0 (en) 2010-08-20 2010-10-06 Ucb Pharma Sa Biological products
CA2809433A1 (en) 2010-08-26 2012-03-01 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
EP2614080A1 (en) 2010-09-10 2013-07-17 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Phosphorylated twist1 and metastasis
US20140093506A1 (en) 2010-11-15 2014-04-03 Marc Buehler Anti-fungal-agents
ES2696548T3 (en) 2010-11-19 2019-01-16 Morphosys Ag A collection of antibody sequences and their use
RU2627171C2 (en) 2010-12-21 2017-08-03 Эббви Инк. Il-1 alpha and beta bispecific immunoglobulins with double variable domains and their application
US20120275996A1 (en) 2010-12-21 2012-11-01 Abbott Laboratories IL-1 Binding Proteins
KR101941514B1 (en) 2010-12-22 2019-01-23 테바 파마슈티컬즈 오스트레일리아 피티와이 엘티디 Modified antibody with improved half-life
GB201100282D0 (en) 2011-01-07 2011-02-23 Ucb Pharma Sa Biological methods
US10208349B2 (en) 2011-01-07 2019-02-19 Ucb Biopharma Sprl Lipocalin 2 as a biomarker for IL-17 inhibitor therapy efficacy
SG10201510762YA (en) 2011-01-14 2016-01-28 Ucb Pharma Sa Antibody molecules which bind il-17a and il-17f
US20120189633A1 (en) 2011-01-26 2012-07-26 Kolltan Pharmaceuticals, Inc. Anti-kit antibodies and uses thereof
EP2699264B1 (en) 2011-04-20 2018-03-14 Medlmmune, LLC Antibodies and other molecules that bind b7-h1 and pd-1
US9376495B2 (en) 2011-05-21 2016-06-28 Macrogenics, Inc. Deimmunized serum-binding domains and their use in extending serum half-life
EP2717911A1 (en) 2011-06-06 2014-04-16 Novartis Forschungsstiftung, Zweigniederlassung Protein tyrosine phosphatase, non-receptor type 11 (ptpn11) and triple-negative breast cancer
WO2012170742A2 (en) 2011-06-07 2012-12-13 University Of Hawaii Treatment and prevention of cancer with hmgb1 antagonists
US9244074B2 (en) 2011-06-07 2016-01-26 University Of Hawaii Biomarker of asbestos exposure and mesothelioma
HUE038509T2 (en) 2011-06-10 2018-10-29 Medimmune Ltd Anti-pseudomonas psl binding molecules and uses thereof
JP6113721B2 (en) 2011-06-28 2017-04-12 オックスフォード ビオトヘラペウトイクス エルティーディー. Therapeutic and diagnostic targets
NZ618503A (en) 2011-06-28 2016-03-31 Oxford Biotherapeutics Ltd Antibodies to adp-ribosyl cyclase 2
MX2014000531A (en) 2011-07-13 2014-12-05 Abbvie Inc Methods and compositions for treating asthma using anti-il-13 antibodies.
GB201112056D0 (en) 2011-07-14 2011-08-31 Univ Leuven Kath Antibodies
AU2012296613B2 (en) 2011-08-15 2016-05-12 Amplimmune, Inc. Anti-B7-H4 antibodies and their uses
EP2753697A1 (en) 2011-09-05 2014-07-16 ETH Zürich Biosynthetic gene cluster for the production of peptide/protein analogues
US9447192B2 (en) 2011-09-09 2016-09-20 Medimmune Limited Anti-Siglec-15 antibodies and uses thereof
AR088513A1 (en) 2011-10-24 2014-06-18 Abbvie Inc IMMUNO LINKERS AGAINST SCLEROSTINE
WO2013067060A1 (en) 2011-11-01 2013-05-10 Bionomics, Inc. Anti-gpr49 antibodies
AU2012332588B2 (en) 2011-11-01 2017-09-07 Bionomics, Inc. Methods of blocking cancer stem cell growth
WO2013067057A1 (en) 2011-11-01 2013-05-10 Bionomics, Inc. Anti-gpr49 antibodies
US9221906B2 (en) 2011-11-01 2015-12-29 Bionomics Inc. Methods of inhibiting solid tumor growth by administering GPR49 antibodies
US20140314787A1 (en) 2011-11-08 2014-10-23 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute Treatment for neurodegenerative diseases
EP2776838A1 (en) 2011-11-08 2014-09-17 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Early diagnostic of neurodegenerative diseases
SG11201401649VA (en) 2011-11-11 2014-07-30 Ucb Pharma Sa Albumin binding antibodies and binding fragments thereof
CA2855570A1 (en) 2011-12-14 2013-06-20 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of iron-related disorders
US10118958B2 (en) 2011-12-14 2018-11-06 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of iron-related disorders
ES2721882T3 (en) 2011-12-23 2019-08-06 Pfizer Constant regions of genetically engineered antibody for site-specific conjugation and procedures and uses thereof
BR112014015851A2 (en) 2011-12-30 2019-09-24 Abbvie Inc double specific binding proteins directed against il-13 and / or il-17
WO2013102825A1 (en) 2012-01-02 2013-07-11 Novartis Ag Cdcp1 and breast cancer
GB201201332D0 (en) 2012-01-26 2012-03-14 Imp Innovations Ltd Method
ES2676725T3 (en) 2012-01-27 2018-07-24 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of diseases associated with the degeneration of neurites
ES2725569T3 (en) 2012-02-10 2019-09-24 Seattle Genetics Inc Diagnosis and treatment of cancers that express CD30
PL2814842T3 (en) 2012-02-15 2018-12-31 Novo Nordisk A/S Antibodies that bind peptidoglycan recognition protein 1
CN108103069B (en) 2012-02-15 2021-08-10 诺和诺德股份有限公司 Antibodies that bind to and block trigger receptor-1 (TREM-1) expressed by myeloid cells
US9550830B2 (en) 2012-02-15 2017-01-24 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
EP2814468B1 (en) 2012-02-16 2019-06-19 Santarus, Inc. Anti-vla1 (cd49a) antibody pharmaceutical compositions
GB201203071D0 (en) 2012-02-22 2012-04-04 Ucb Pharma Sa Biological products
GB201203051D0 (en) 2012-02-22 2012-04-04 Ucb Pharma Sa Biological products
CA2866185C (en) 2012-03-23 2021-04-06 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Pathogenic phlebovirus isolates and compositions and methods of use
US20150266961A1 (en) 2012-03-29 2015-09-24 Novartis Forschungsstiftung, Zweigniederlassung, Fridrich Miescher Institute Inhibition of interleukin-8 and/or its receptor cxcr1 in the treatment of her2/her3-overexpressing breast cancer
WO2013151649A1 (en) 2012-04-04 2013-10-10 Sialix Inc Glycan-interacting compounds
JP2015518829A (en) 2012-05-14 2015-07-06 バイオジェン・エムエイ・インコーポレイテッドBiogen MA Inc. LINGO-2 antagonist for treatment of conditions involving motor neurons
BR112014028306A2 (en) 2012-05-15 2018-04-17 Morphotek, Inc. methods for treating gastric cancer.
JP6629069B2 (en) 2012-06-06 2020-01-15 ゾエティス・エルエルシー Canine anti-NGF antibody and method thereof
US10048253B2 (en) 2012-06-28 2018-08-14 Ucb Biopharma Sprl Method for identifying compounds of therapeutic interest
WO2014001482A1 (en) 2012-06-29 2014-01-03 Novartis Forschungsstiftung, Zweigniererlassung, Friedrich Miescher Institute For Biomedical Research Treating diseases by modulating a specific isoform of mkl1
EP2870242A1 (en) 2012-07-05 2015-05-13 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research New treatment for neurodegenerative diseases
WO2014006115A1 (en) 2012-07-06 2014-01-09 Novartis Ag Combination of a phosphoinositide 3-kinase inhibitor and an inhibitor of the il-8/cxcr interaction
US9670276B2 (en) 2012-07-12 2017-06-06 Abbvie Inc. IL-1 binding proteins
GB201213652D0 (en) 2012-08-01 2012-09-12 Oxford Biotherapeutics Ltd Therapeutic and diagnostic target
WO2014055442A2 (en) 2012-10-01 2014-04-10 The Trustees Of The University Of Pennsylvania Compositions and methods for targeting stromal cells for the treatment of cancer
WO2014059251A1 (en) 2012-10-12 2014-04-17 The Brigham And Women's Hospital, Inc. Enhancement of the immune response
SG11201503412RA (en) 2012-11-01 2015-05-28 Abbvie Inc Anti-vegf/dll4 dual variable domain immunoglobulins and uses thereof
GB201223276D0 (en) 2012-12-21 2013-02-06 Ucb Pharma Sa Antibodies and methods of producing same
CA2896091C (en) 2012-12-21 2018-06-19 Amplimmune, Inc. Anti-h7cr antibodies
US9550986B2 (en) 2012-12-21 2017-01-24 Abbvie Inc. High-throughput antibody humanization
AU2013202668B2 (en) 2012-12-24 2014-12-18 Adelaide Research & Innovation Pty Ltd Inhibition of cancer growth and metastasis
EP2954323B1 (en) 2013-02-07 2020-04-15 The Regents of The University of California Use of translational profiling to identify target molecules for therapeutic treatment
GB201302447D0 (en) 2013-02-12 2013-03-27 Oxford Biotherapeutics Ltd Therapeutic and diagnostic target
JP6739329B2 (en) 2013-03-14 2020-08-12 アボット・ラボラトリーズAbbott Laboratories HCV core lipid binding domain monoclonal antibody
RU2015144186A (en) 2013-03-15 2017-04-24 Эббви Инк. CLEANING THE ANTIBODY MEDICINE (ADC) CONJUGATE
WO2014143739A2 (en) 2013-03-15 2014-09-18 Biogen Idec Ma Inc. Anti-alpha v beta 6 antibodies and uses thereof
WO2014144466A1 (en) 2013-03-15 2014-09-18 Biogen Idec Ma Inc. Anti-alpha v beta 6 antibodies and uses thereof
US9469686B2 (en) 2013-03-15 2016-10-18 Abbott Laboratories Anti-GP73 monoclonal antibodies and methods of obtaining the same
WO2014143765A1 (en) 2013-03-15 2014-09-18 Abbvie Deutschland Gmbh & Co.Kg Anti-egfr antibody drug conjugate formulations
CN105324396A (en) 2013-03-15 2016-02-10 艾伯维公司 Dual specific binding proteins directed against il-1 beta and il-17
JP2016521283A (en) 2013-05-06 2016-07-21 スカラー ロック インコーポレイテッドScholar Rock,Inc. Compositions and methods for growth factor modulation
CA2913312A1 (en) 2013-05-24 2014-11-27 Medimmune, Llc Anti-b7-h5 antibodies and their uses
SG11201509982UA (en) 2013-06-06 2016-04-28 Igenica Biotherapeutics Inc
JP6581572B2 (en) 2013-06-07 2019-09-25 デューク ユニバーシティ Complement factor H inhibitor
EP3030902B1 (en) 2013-08-07 2019-09-25 Friedrich Miescher Institute for Biomedical Research New screening method for the treatment friedreich's ataxia
KR20160054501A (en) 2013-08-26 2016-05-16 맵백스 테라퓨틱스, 인코포레이티드 Nucleic acids encoding human antibodies to sialyl-lewis a
AU2014342528A1 (en) 2013-10-28 2016-04-28 Dots Technology Corp. Allergen detection
US9914769B2 (en) 2014-07-15 2018-03-13 Kymab Limited Precision medicine for cholesterol treatment
US9045545B1 (en) 2014-07-15 2015-06-02 Kymab Limited Precision medicine by targeting PD-L1 variants for treatment of cancer
US8992927B1 (en) 2014-07-15 2015-03-31 Kymab Limited Targeting human NAV1.7 variants for treatment of pain
US9067998B1 (en) 2014-07-15 2015-06-30 Kymab Limited Targeting PD-1 variants for treatment of cancer
US8986694B1 (en) 2014-07-15 2015-03-24 Kymab Limited Targeting human nav1.7 variants for treatment of pain
GB201403775D0 (en) 2014-03-04 2014-04-16 Kymab Ltd Antibodies, uses & methods
US9738702B2 (en) 2014-03-14 2017-08-22 Janssen Biotech, Inc. Antibodies with improved half-life in ferrets
CN106536556B (en) 2014-04-04 2020-02-07 生态学有限公司 Humanized antibodies that bind LGR5
US20170267780A1 (en) 2014-05-16 2017-09-21 Medimmune, Llc Molecules with altered neonate fc receptor binding having enhanced therapeutic and diagnostic properties
NZ726513A (en) 2014-05-28 2023-07-28 Memorial Sloan Kettering Cancer Center Anti-gitr antibodies and methods of use thereof
GB201409558D0 (en) 2014-05-29 2014-07-16 Ucb Biopharma Sprl Method
NZ764877A (en) 2014-06-04 2023-12-22 Biontech Res And Development Inc Human monoclonal antibodies to ganglioside gd2
EP3154579A1 (en) 2014-06-13 2017-04-19 Friedrich Miescher Institute for Biomedical Research New treatment against influenza virus
WO2015198202A1 (en) 2014-06-23 2015-12-30 Friedrich Miescher Institute For Biomedical Research Methods for triggering de novo formation of heterochromatin and or epigenetic silencing with small rnas
GB201411320D0 (en) 2014-06-25 2014-08-06 Ucb Biopharma Sprl Antibody construct
WO2016001830A1 (en) 2014-07-01 2016-01-07 Friedrich Miescher Institute For Biomedical Research Combination of a brafv600e inhibitor and mertk inhibitor to treat melanoma
US9139648B1 (en) 2014-07-15 2015-09-22 Kymab Limited Precision medicine by targeting human NAV1.9 variants for treatment of pain
GB201412659D0 (en) 2014-07-16 2014-08-27 Ucb Biopharma Sprl Molecules
GB201412658D0 (en) 2014-07-16 2014-08-27 Ucb Biopharma Sprl Molecules
TN2017000008A1 (en) 2014-07-17 2018-07-04 Novo Nordisk As Site directed mutagenesis of trem-1 antibodies for decreasing viscosity.
EP3197557A1 (en) 2014-09-24 2017-08-02 Friedrich Miescher Institute for Biomedical Research Lats and breast cancer
RU2021125449A (en) 2014-10-01 2021-09-16 Медиммьюн Лимитед ANTIBODIES TO TICAGRELOR AND METHODS OF APPLICATION
MX2017006167A (en) 2014-11-12 2018-03-23 Siamab Therapeutics Inc Glycan-interacting compounds and methods of use.
US9879087B2 (en) 2014-11-12 2018-01-30 Siamab Therapeutics, Inc. Glycan-interacting compounds and methods of use
WO2016094881A2 (en) 2014-12-11 2016-06-16 Abbvie Inc. Lrp-8 binding proteins
PT3333191T (en) 2014-12-11 2020-12-15 Pf Medicament Anti-c10orf54 antibodies and uses thereof
US10221248B2 (en) 2014-12-22 2019-03-05 The Rockefeller University Anti-MERTK agonistic antibodies and uses thereof
EP3242893A1 (en) 2015-01-08 2017-11-15 Biogen MA Inc. Lingo-1 antagonists and uses for treatment of demyelinating disorders
PL3265123T3 (en) 2015-03-03 2023-03-13 Kymab Limited Antibodies, uses & methods
WO2016154177A2 (en) 2015-03-23 2016-09-29 Jounce Therapeutics, Inc. Antibodies to icos
CA2981142A1 (en) 2015-03-27 2016-10-06 University Of Southern California Car t-cell therapy directed to lhr for the treatment of solid tumors
GB201506870D0 (en) 2015-04-22 2015-06-03 Ucb Biopharma Sprl Method
GB201506869D0 (en) 2015-04-22 2015-06-03 Ucb Biopharma Sprl Method
EP3288542B1 (en) 2015-04-29 2021-12-29 University Of South Australia Compositions and methods for administering antibodies
EP3291836A4 (en) 2015-05-06 2018-11-14 Janssen Biotech, Inc. Prostate specific membrane antigen (psma) bispecific binding agents and uses thereof
AU2016256911B2 (en) 2015-05-07 2022-03-31 Agenus Inc. Anti-OX40 antibodies and methods of use thereof
MY195000A (en) 2015-05-27 2022-12-30 Ucb Biopharma Sprl Method for the treatment of neurological disease
HUE050750T2 (en) 2015-05-29 2021-01-28 Agenus Inc Anti-ctla-4 antibodies and methods of use thereof
PE20180193A1 (en) 2015-05-29 2018-01-26 Abbvie Inc ANTI-CD40 ANTIBODIES AND THEIR USES
EP3302559B1 (en) 2015-06-04 2022-01-12 University of Southern California Lym-1 and lym-2 targeted car cell immunotherapy
TW201710286A (en) 2015-06-15 2017-03-16 艾伯維有限公司 Binding proteins against VEGF, PDGF, and/or their receptors
GB201510758D0 (en) 2015-06-18 2015-08-05 Ucb Biopharma Sprl Novel TNFa structure for use in therapy
GB201601075D0 (en) 2016-01-20 2016-03-02 Ucb Biopharma Sprl Antibodies molecules
GB201601073D0 (en) 2016-01-20 2016-03-02 Ucb Biopharma Sprl Antibodies
GB201601077D0 (en) 2016-01-20 2016-03-02 Ucb Biopharma Sprl Antibody molecule
WO2017040790A1 (en) 2015-09-01 2017-03-09 Agenus Inc. Anti-pd-1 antibodies and methods of use thereof
AU2016326721C1 (en) 2015-09-23 2021-06-03 Cytoimmune Therapeutics, Inc. FLT3 directed car cells for immunotherapy
KR20180066236A (en) 2015-10-22 2018-06-18 조운스 테라퓨틱스, 인크. Gene traits for measuring ICOS expression
BR112018008075A2 (en) 2015-10-27 2018-12-04 Ucb Biopharma Sprl treatment methods using anti-il-17a / f antibodies
US20180348224A1 (en) 2015-10-28 2018-12-06 Friedrich Miescher Institute For Biomedical Resear Ch Tenascin-w and biliary tract cancers
KR20180088381A (en) 2015-11-12 2018-08-03 시아맙 쎄라퓨틱스, 인코포레이티드 Glycan-interacting compounds and methods of use
CN109415437B (en) 2015-12-02 2022-02-01 斯特库伯株式会社 Antibodies and molecules that immunospecifically bind to BTN1A1 and therapeutic uses thereof
CN108925136B (en) 2015-12-02 2022-02-01 斯特赛恩斯公司 Antibodies specific for glycosylated BTLA (B and T lymphocyte attenuating factor)
GB201521389D0 (en) 2015-12-03 2016-01-20 Ucb Biopharma Sprl Method
GB201521382D0 (en) 2015-12-03 2016-01-20 Ucb Biopharma Sprl Antibodies
GB201521393D0 (en) 2015-12-03 2016-01-20 Ucb Biopharma Sprl Antibodies
GB201521383D0 (en) 2015-12-03 2016-01-20 Ucb Biopharma Sprl And Ucb Celltech Method
GB201521391D0 (en) 2015-12-03 2016-01-20 Ucb Biopharma Sprl Antibodies
EP3386542B1 (en) 2015-12-10 2020-11-18 Katholieke Universiteit Leuven Anti adamts13 antibodies and their use for treatment or prevention of haemorrhagic disorders due to ventricular assist device
GB201602413D0 (en) 2016-02-10 2016-03-23 Nascient Ltd Method
MX2018010771A (en) 2016-03-10 2019-05-15 Viela Bio Inc Ilt7 binding molecules and methods of using the same.
US10745487B2 (en) 2016-03-22 2020-08-18 Bionomics Limited Method of treating cancer by administering an anti-LGR5 monoclonal antibody
CN116333130A (en) 2016-05-24 2023-06-27 英斯梅德股份有限公司 Antibodies and methods of making the same
KR102366813B1 (en) 2016-05-27 2022-02-24 아게누스 인코포레이티드 Anti-TIM-3 Antibodies and Methods of Using Same
AU2017292752B2 (en) 2016-07-06 2023-07-27 Celgene Corporation Antibodies with low immunogenicity and uses thereof
AU2017292184A1 (en) 2016-07-08 2019-02-07 Staten Biotechnology B.V. Anti-Apoc3 antibodies and methods of use thereof
GB201616596D0 (en) 2016-09-29 2016-11-16 Nascient Limited Epitope and antibodies
MX2019003473A (en) 2016-10-03 2019-10-15 Abbott Lab Improved methods of assessing uch-l1 status in patient samples.
IL265800B2 (en) 2016-10-11 2023-10-01 Agenus Inc Anti-lag-3 antibodies and methods of use thereof
WO2018085359A1 (en) 2016-11-02 2018-05-11 Immunogen, Inc. Combination treatment with antibody-drug conjugates and parp inhibitors
US11779604B2 (en) 2016-11-03 2023-10-10 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses and methods
AU2017353939A1 (en) 2016-11-07 2019-06-06 Neuracle Science Co., Ltd. Anti-family with sequence similarity 19, member A5 antibodies and method of use thereof
WO2018094143A1 (en) 2016-11-17 2018-05-24 Siamab Therapeutics, Inc. Glycan-interacting compounds and methods of use
WO2018100628A1 (en) * 2016-11-29 2018-06-07 国立大学法人東北大学 Establishing metal allergy animal model that is highly sensitive/susceptive to metal
KR102603681B1 (en) 2016-12-07 2023-11-17 아게누스 인코포레이티드 Antibodies and methods of using them
MA50949B1 (en) 2016-12-07 2023-12-29 Memorial Sloan Kettering Cancer Center ANTI-CTLA-4 ANTIBODIES AND METHODS OF USE THEREOF
GB201621635D0 (en) 2016-12-19 2017-02-01 Ucb Biopharma Sprl Crystal structure
EP3589319A4 (en) 2017-03-03 2021-07-14 Seagen Inc. Glycan-interacting compounds and methods of use
CA3052513A1 (en) 2017-03-23 2018-09-27 Abbott Laboratories Methods for aiding in the diagnosis and determination of the extent of traumatic brain injury in a human subject using the early biomarker ubiquitin carboxy-terminal hydrolase l1
TW201841942A (en) 2017-04-13 2018-12-01 美商艾吉納斯公司 Anti-CD137 antibodies and methods of use thereof
JP7344797B2 (en) 2017-04-15 2023-09-14 アボット・ラボラトリーズ Methods to aid in hyperacute diagnosis and determination of traumatic brain injury in human subjects using early biomarkers
EP3612560A1 (en) 2017-04-21 2020-02-26 Staten Biotechnology B.V. Anti-apoc3 antibodies and methods of use thereof
CN110603449A (en) 2017-04-28 2019-12-20 雅培实验室 Method for determining traumatic brain injury using early biomarkers from at least two samples of the same human subject for aiding hyperacute diagnosis
MA50957A (en) 2017-05-01 2020-10-14 Agenus Inc ANTI-TIGIT ANTIBODIES AND THEIR METHODS OF USE
US10865238B1 (en) 2017-05-05 2020-12-15 Duke University Complement factor H antibodies
JOP20190256A1 (en) 2017-05-12 2019-10-28 Icahn School Med Mount Sinai Newcastle disease viruses and uses thereof
CN110651190A (en) 2017-05-25 2020-01-03 雅培实验室 Method for using early biomarkers to help determine whether to perform imaging on a human subject who has suffered or may have suffered a head injury
AU2018275235A1 (en) 2017-05-30 2019-10-31 Abbott Laboratories Methods for aiding in diagnosing and evaluating a mild traumatic brain injury in a human subject using cardiac troponin I and early biomarkers
CN111051346A (en) 2017-05-31 2020-04-21 斯特库伯株式会社 Methods of treating cancer using antibodies and molecules that immunospecifically bind to BTN1a1
CA3065301A1 (en) 2017-05-31 2018-12-06 Stcube & Co., Inc. Antibodies and molecules that immunospecifically bind to btn1a1 and the therapeutic uses thereof
US11542331B2 (en) 2017-06-06 2023-01-03 Stcube & Co., Inc. Methods of treating cancer using antibodies and molecules that bind to BTN1A1 or BTN1A1-ligands
EP3649474A1 (en) 2017-07-03 2020-05-13 Abbott Laboratories Improved methods for measuring ubiquitin carboxy-terminal hydrolase l1 levels in blood
SG11201913137VA (en) 2017-07-11 2020-01-30 Compass Therapeutics Llc Agonist antibodies that bind human cd137 and uses thereof
EP3684811A2 (en) 2017-08-17 2020-07-29 Massachusetts Institute of Technology Multiple specificity binders of cxc chemokines and uses thereof
BR112020003533A2 (en) 2017-08-25 2020-11-17 Five Prime Therapeutics, Inc. b7-h4 antibodies and methods of using them
CN111630069A (en) 2017-10-13 2020-09-04 勃林格殷格翰国际有限公司 Human antibodies to Thomsen-novell (Tn) antigens
WO2019089753A2 (en) 2017-10-31 2019-05-09 Compass Therapeutics Llc Cd137 antibodies and pd-1 antagonists and uses thereof
US20190160089A1 (en) 2017-10-31 2019-05-30 Immunogen, Inc. Combination treatment with antibody-drug conjugates and cytarabine
BR112020008514A2 (en) 2017-10-31 2020-10-20 Staten Biotechnology B.V. anti-apoc3 antibodies and methods of using them
JP2021502125A (en) 2017-11-09 2021-01-28 ピンテオン セラピューティクス インコーポレイテッド Methods and Compositions for the Preparation and Use of Humanized Conformation-Specific Phosphorylated Tau Antibodies
EP3713961A2 (en) 2017-11-20 2020-09-30 Compass Therapeutics LLC Cd137 antibodies and tumor antigen-targeting antibodies and uses thereof
CN111727075B (en) 2017-11-27 2024-04-05 普渡制药公司 Humanized antibodies targeting human tissue factor
WO2019113525A2 (en) 2017-12-09 2019-06-13 Abbott Laboratories Methods for aiding in the diagnosis and evaluation of a subject who has sustained an orthopedic injury and that has or may have sustained an injury to the head, such as mild traumatic brain injury (tbi), using glial fibrillary acidic protein (gfap) and/or ubiquitin carboxy-terminal hydrolase l1 (uch-l1)
WO2019112860A1 (en) 2017-12-09 2019-06-13 Abbott Laboratories Methods for aiding in diagnosing and evaluating a traumatic brain injury in a human subject using a combination of gfap and uch-l1
GB201802486D0 (en) 2018-02-15 2018-04-04 Ucb Biopharma Sprl Methods
AU2019228600A1 (en) 2018-03-02 2020-09-24 Five Prime Therapeutics, Inc. B7-H4 antibodies and methods of use thereof
WO2019177690A1 (en) 2018-03-12 2019-09-19 Zoetis Services Llc Anti-ngf antibodies and methods thereof
NZ782442A (en) 2018-03-14 2022-01-28 Surface Oncology Inc Antibodies that bind cd39 and uses thereof
US11332524B2 (en) 2018-03-22 2022-05-17 Surface Oncology, Inc. Anti-IL-27 antibodies and uses thereof
JOP20200240A1 (en) 2018-04-02 2020-09-27 Bristol Myers Squibb Co Anti-trem-1 antibodies and uses thereof reference to sequence listing submitted electronically via efs-web
WO2019200357A1 (en) 2018-04-12 2019-10-17 Surface Oncology, Inc. Biomarker for cd47 targeting therapeutics and uses therefor
US20210230255A1 (en) 2018-04-27 2021-07-29 Fondazione Ebri Rita Levi-Montalcini Antibody directed against a tau-derived neurotoxic peptide and uses thereof
WO2019222130A1 (en) 2018-05-15 2019-11-21 Immunogen, Inc. Combination treatment with antibody-drug conjugates and flt3 inhibitors
WO2019226658A1 (en) 2018-05-21 2019-11-28 Compass Therapeutics Llc Multispecific antigen-binding compositions and methods of use
US20200109195A1 (en) 2018-05-21 2020-04-09 Compass Therapeutics Llc Compositions and methods for enhancing the killing of target cells by nk cells
AU2019289176A1 (en) 2018-06-18 2020-12-24 Oxford University Innovation Limited Gremlin-1 antagonist for the prevention and treatment of cancer
CN112533629A (en) 2018-06-19 2021-03-19 阿尔莫生物科技股份有限公司 Compositions and methods for combined use of IL-10 agents with chimeric antigen receptor cell therapy
TW202016144A (en) 2018-06-21 2020-05-01 日商第一三共股份有限公司 Compositions including cd3 antigen binding fragments and uses thereof
WO2020016459A1 (en) 2018-07-20 2020-01-23 Pierre Fabre Medicament Receptor for vista
WO2020033925A2 (en) 2018-08-09 2020-02-13 Compass Therapeutics Llc Antibodies that bind cd277 and uses thereof
US20210388089A1 (en) 2018-08-09 2021-12-16 Compass Therapeutics Llc Antigen binding agents that bind cd277 and uses thereof
US20210309746A1 (en) 2018-08-09 2021-10-07 Compass Therapeutics Llc Antibodies that bind cd277 and uses thereof
EP3856773A1 (en) 2018-09-28 2021-08-04 Kyowa Kirin Co., Ltd. Il-36 antibodies and uses thereof
JP2022504287A (en) 2018-10-03 2022-01-13 スターテン・バイオテクノロジー・ベー・フェー Antibodies specific for human and cynomolgus monkey APOC3, and methods of their use
GB201817311D0 (en) 2018-10-24 2018-12-05 Ucb Biopharma Sprl Antibodies
GB201817309D0 (en) 2018-10-24 2018-12-05 Ucb Biopharma Sprl Antibodies
BR112021008795A2 (en) 2018-11-13 2021-08-31 Compass Therapeutics Llc MULTISPECIFIC BINDING CONSTRUCTS AGAINST CHECKPOINT MOLECULES AND THEIR USES
JP2022514903A (en) 2018-12-20 2022-02-16 協和キリン株式会社 FN14 antibody and its use
MX2021008453A (en) 2019-01-16 2021-08-19 Compass Therapeutics Llc Formulations of antibodies that bind human cd137 and uses thereof.
GB201900732D0 (en) 2019-01-18 2019-03-06 Ucb Biopharma Sprl Antibodies
US11242407B2 (en) 2019-02-26 2022-02-08 Inspirna, Inc. High-affinity anti-MERTK antibodies and uses thereof
WO2020198731A2 (en) 2019-03-28 2020-10-01 Danisco Us Inc Engineered antibodies
WO2020209929A1 (en) * 2019-04-09 2020-10-15 Massachusetts Institute Of Technology Mutant subgenomic promoter library and uses thereof
CN114867751A (en) 2019-08-12 2022-08-05 阿帕特夫研究和发展有限公司 4-1BB and OX40 binding proteins and related compositions and methods, anti-4-1 BB antibodies, anti-OX 40 antibodies
WO2021042019A1 (en) 2019-08-30 2021-03-04 Agenus Inc. Anti-cd96 antibodies and methods of use thereof
CN114641501A (en) 2019-09-04 2022-06-17 Y生物股份有限公司 anti-VSIG 4 antibodies or antigen binding fragments and uses thereof
TW202124444A (en) 2019-09-16 2021-07-01 美商表面腫瘤學公司 Anti-cd39 antibody compositions and methods
BR112022004302A2 (en) 2019-09-25 2022-06-21 Surface Oncology Inc Anti-il-27 antibodies and uses thereof
WO2021062323A1 (en) 2019-09-26 2021-04-01 Stcube & Co. Antibodies specific to glycosylated ctla-4 and methods of use thereof
JP2022552282A (en) 2019-10-09 2022-12-15 エスティーキューブ アンド カンパニー Antibodies specific for glycosylated LAG3 and methods of use thereof
US11459389B2 (en) 2019-10-24 2022-10-04 Massachusetts Institute Of Technology Monoclonal antibodies that bind human CD161
GB201917480D0 (en) 2019-11-29 2020-01-15 Univ Oxford Innovation Ltd Antibodies
GB201919058D0 (en) 2019-12-20 2020-02-05 Ucb Biopharma Sprl Multi-specific antibodies
GB201919061D0 (en) 2019-12-20 2020-02-05 Ucb Biopharma Sprl Multi-specific antibody
GB201919062D0 (en) 2019-12-20 2020-02-05 Ucb Biopharma Sprl Antibody
TW202138388A (en) 2019-12-30 2021-10-16 美商西根公司 Methods of treating cancer with nonfucosylated anti-cd70 antibodies
IL294330A (en) 2020-01-06 2022-08-01 Vaccinex Inc Anti-ccr8 antibodies and uses thereof
CN115315273A (en) 2020-01-14 2022-11-08 辛德凯因股份有限公司 IL-2 orthologs and methods of use thereof
IL295387A (en) 2020-02-05 2022-10-01 Larimar Therapeutics Inc Tat peptide binding proteins and uses thereof
EP4103608A1 (en) 2020-02-13 2022-12-21 UCB Biopharma SRL Bispecific antibodies against cd9 and cd137
EP4103611B1 (en) 2020-02-13 2024-03-27 UCB Biopharma SRL Bispecific antibodies binding hvem and cd9
WO2021160269A1 (en) 2020-02-13 2021-08-19 UCB Biopharma SRL Anti cd44-ctla4 bispecific antibodies
US20230151109A1 (en) 2020-02-13 2023-05-18 UCB Biopharma SRL Bispecific antibodies against cd9
US20230096030A1 (en) 2020-02-13 2023-03-30 UCB Biopharma SRL Bispecific antibodies against cd9 and cd7
US20230220074A1 (en) 2020-02-18 2023-07-13 Alector Llc Pilra antibodies and methods of use thereof
JP2023516724A (en) 2020-03-06 2023-04-20 オーエヌエー セラピューティクス エセ.エレ. Anti-CD36 antibodies and their use to treat cancer
AU2021232853A1 (en) 2020-03-10 2022-09-22 Massachusetts Institute Of Technology Compositions and methods for immunotherapy of NPM1c-positive cancer
WO2021202473A2 (en) 2020-03-30 2021-10-07 Danisco Us Inc Engineered antibodies
CA3175523A1 (en) 2020-04-13 2021-10-21 Antti Virtanen Methods, complexes and kits for detecting or determining an amount of a .beta.-coronavirus antibody in a sample
AU2021275361A1 (en) 2020-05-17 2023-01-19 Astrazeneca Uk Limited SARS-CoV-2 antibodies and methods of selecting and using the same
AU2021313348A1 (en) 2020-07-20 2023-03-09 Astrazeneca Uk Limited SARS-CoV-2 proteins, anti-SARS-CoV-2 antibodies, and methods of using the same
EP4193149A1 (en) 2020-08-04 2023-06-14 Abbott Laboratories Improved methods and kits for detecting sars-cov-2 protein in a sample
EP4200018A1 (en) 2020-08-18 2023-06-28 Cephalon LLC Anti-par-2 antibodies and methods of use thereof
IL301859A (en) 2020-10-15 2023-06-01 UCB Biopharma SRL Binding molecules that multimerise cd45
US20230382978A1 (en) 2020-10-15 2023-11-30 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Antibody specific for sars-cov-2 receptor binding domain and therapeutic methods
WO2022087274A1 (en) 2020-10-21 2022-04-28 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Antibodies that neutralize type-i interferon (ifn) activity
US20220170948A1 (en) 2020-12-01 2022-06-02 Abbott Laboratories Use of one or more biomarkers to determine traumatic brain injury (tbi) in a human subject having received a head computerized tomography scan that is negative for a tbi
IL303328A (en) 2020-12-01 2023-07-01 Aptevo Res & Development Llc Heterodimeric psma and cd3-binding bispecific antibodies
WO2023102384A1 (en) 2021-11-30 2023-06-08 Abbott Laboratories Use of one or more biomarkers to determine traumatic brain injury (tbi) in a subject having received a head computerized tomography scan that is negative for a tbi
EP4271998A1 (en) 2020-12-30 2023-11-08 Abbott Laboratories Methods for determining sars-cov-2 antigen and anti-sars-cov-2 antibody in a sample
JP2024503657A (en) 2021-01-13 2024-01-26 メモリアル スローン-ケタリング キャンサー センター Antibody-pyrrolobenzodiazepine derivative conjugate
KR20230146522A (en) 2021-01-13 2023-10-19 메모리얼 슬로안 케터링 캔서 센터 Anti-DLL3 antibody-drug conjugate
TW202302645A (en) 2021-03-03 2023-01-16 法商皮爾法伯製藥公司 Anti-vsig4 antibody or antigen binding fragment and uses thereof
EP4067381A1 (en) 2021-04-01 2022-10-05 Julius-Maximilians-Universität Würzburg Novel tnfr2 binding molecules
AU2022270170A1 (en) 2021-05-07 2023-09-21 Surface Oncology, LLC Anti-il-27 antibodies and uses thereof
CA3216320A1 (en) 2021-05-18 2022-11-24 Abbott Laboratories Methods of evaluating brain injury in a pediatric subject
WO2022251446A1 (en) 2021-05-28 2022-12-01 Alexion Pharmaceuticals, Inc. Methods for detecting cm-tma biomarkers
IL309349A (en) 2021-06-14 2024-02-01 argenx BV Anti-il-9 antibodies and methods of use thereof
WO2022266034A1 (en) 2021-06-14 2022-12-22 Abbott Laboratories Methods of diagnosing or aiding in diagnosis of brain injury caused by acoustic energy, electromagnetic energy, an over pressurization wave, and/or blast wind
CA3221281A1 (en) 2021-06-29 2023-01-05 Seagen Inc. Methods of treating cancer with a combination of a nonfucosylated anti-cd70 antibody and a cd47 antagonist
WO2023285878A1 (en) 2021-07-13 2023-01-19 Aviation-Ophthalmology Methods for detecting, treating, and preventing gpr68-mediated ocular diseases, disorders, and conditions
AU2022320051A1 (en) 2021-07-30 2024-01-25 ONA Therapeutics S.L. Anti-cd36 antibodies and their use to treat cancer
CA3230038A1 (en) 2021-08-31 2023-03-09 Hongwei Zhang Methods and systems of diagnosing brain injury
AU2022354059A1 (en) 2021-09-30 2024-03-28 Abbott Laboratories Methods and systems of diagnosing brain injury
EP4177266A1 (en) 2021-11-05 2023-05-10 Katholieke Universiteit Leuven Neutralizing anti-sars-cov-2 human antibodies
WO2023114978A1 (en) 2021-12-17 2023-06-22 Abbott Laboratories Systems and methods for determining uch-l1, gfap, and other biomarkers in blood samples
US20230213536A1 (en) 2021-12-28 2023-07-06 Abbott Laboratories Use of biomarkers to determine sub-acute traumatic brain injury (tbi) in a subject having received a head computerized tomography (ct) scan that is negative for a tbi or no head ct scan
WO2023150652A1 (en) 2022-02-04 2023-08-10 Abbott Laboratories Lateral flow methods, assays, and devices for detecting the presence or measuring the amount of ubiquitin carboxy-terminal hydrolase l1 and/or glial fibrillary acidic protein in a sample
WO2023192436A1 (en) 2022-03-31 2023-10-05 Alexion Pharmaceuticals, Inc. Singleplex or multiplexed assay for complement markers in fresh biological samples
GB202205203D0 (en) 2022-04-08 2022-05-25 UCB Biopharma SRL Combination with inhibitor
GB202205200D0 (en) 2022-04-08 2022-05-25 Ucb Biopharma Sprl Combination with chemotherapy
WO2023209177A1 (en) 2022-04-29 2023-11-02 Astrazeneca Uk Limited Sars-cov-2 antibodies and methods of using the same
WO2023240124A1 (en) 2022-06-07 2023-12-14 Regeneron Pharmaceuticals, Inc. Pseudotyped viral particles for targeting tcr-expressing cells
WO2024006876A1 (en) 2022-06-29 2024-01-04 Abbott Laboratories Magnetic point-of-care systems and assays for determining gfap in biological samples
WO2024015953A1 (en) 2022-07-15 2024-01-18 Danisco Us Inc. Methods for producing monoclonal antibodies
WO2024013727A1 (en) 2022-07-15 2024-01-18 Janssen Biotech, Inc. Material and methods for improved bioengineered pairing of antigen-binding variable regions
WO2024050354A1 (en) 2022-08-31 2024-03-07 Washington University Alphavirus antigen binding antibodies and uses thereof
WO2024050524A1 (en) 2022-09-01 2024-03-07 University Of Georgia Research Foundation, Inc. Compositions and methods for directing apolipoprotein l1 to induce mammalian cell death
WO2024054436A1 (en) 2022-09-06 2024-03-14 Alexion Pharmaceuticals, Inc. Diagnostic and prognostic biomarker profiles in patients with hematopoietic stem cell transplant-associated thrombotic microangiopathy (hsct-tma)
WO2024059708A1 (en) 2022-09-15 2024-03-21 Abbott Laboratories Biomarkers and methods for differentiating between mild and supermild traumatic brain injury

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4683195A (en) * 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US5283173A (en) * 1990-01-24 1994-02-01 The Research Foundation Of State University Of New York System to detect protein-protein interactions
US5395750A (en) * 1992-02-28 1995-03-07 Hoffmann-La Roche Inc. Methods for producing proteins which bind to predetermined antigens
US5780225A (en) * 1990-01-12 1998-07-14 Stratagene Method for generating libaries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules
US6303313B1 (en) * 1990-01-11 2001-10-16 Stratagene Method for generating libraries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4683195A (en) * 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4683195B1 (en) * 1986-01-30 1990-11-27 Cetus Corp
US6303313B1 (en) * 1990-01-11 2001-10-16 Stratagene Method for generating libraries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules
US6635424B2 (en) * 1990-01-11 2003-10-21 Stratagene Method for generating libraries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules
US5780225A (en) * 1990-01-12 1998-07-14 Stratagene Method for generating libaries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules
US5283173A (en) * 1990-01-24 1994-02-01 The Research Foundation Of State University Of New York System to detect protein-protein interactions
US5395750A (en) * 1992-02-28 1995-03-07 Hoffmann-La Roche Inc. Methods for producing proteins which bind to predetermined antigens

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090318308A1 (en) * 2006-05-30 2009-12-24 Millegen Highly diversified antibody libraries

Also Published As

Publication number Publication date
US6635424B2 (en) 2003-10-21
US6303313B1 (en) 2001-10-16
US20030054001A1 (en) 2003-03-20
WO1991010737A1 (en) 1991-07-25
AU7247191A (en) 1991-08-05
US6479243B1 (en) 2002-11-12

Similar Documents

Publication Publication Date Title
US6635424B2 (en) Method for generating libraries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules
US5780225A (en) Method for generating libaries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules
Bakkus et al. Evidence that the clonogenic cell in multiple myeloma originates from a pre‐switched but somatically mutated B cell
US6987171B1 (en) Human CD28 specific monoclonal antibodies for antigen-non-specific activation of T-lymphocytes
Weiner Fully human therapeutic monoclonal antibodies
EP0403156B1 (en) Improved monoclonal antibodies against the human alpha/beta t-cell receptor, their production and use
AU658370B2 (en) CD53 cell surface antigen and use thereof
CN108463229A (en) Chimeric protein and immunotherapy method
CN110291200A (en) There is the chimeric transcription factor variant of the sensitivity increased to the medicament ligand induction of mammalian cell transgenic expression
JPH05500312A (en) CDR-grafted antibody
CN109476722A (en) The method of the effect of for improving immunocyte and expansion
EP0330191A2 (en) DNA encoding CD40
CN110462040A (en) The expression of polypeptide is adjusted by new gene switch expression system
CA2129445A1 (en) Design, cloning and expression of humanized monoclonal antibodies against human interleukin-5
JP2000506723A (en) Antibody variant
EP0491878B1 (en) Compositions for the inhibition of protein hormone formation and uses thereof
RU2204602C2 (en) Polypeptide able to formation of antigen-binding structure with specificity to rhesus-d-antigens, dna sequence, method for preparing polypeptide, method of polypeptide selection, complete anti- rhesus-d-antibody (variants), pharmaceutical composition, diagnostic agent for rhesus-d-typifying
RU99101121A (en) POLYEPEPTIDS ABLE TO FORM ANTIGEN-BINDING STRUCTURES SPECIFIC TO RES-D ANTIGENS, DNA ENCODING THEM, METHODS FOR PRODUCING AND APPLICATION
JP2002532066A (en) Antibody production using polynucleotide vaccines in birds
Dighiero et al. What is the CLL B-lymphocyte?
EP1358318A2 (en) Hybridoma cell line g250 and its use for producing monoclonal antibodies
JPH05502586A (en) Chimeric immunoglobulin against CD4 receptor
KR20030083698A (en) Substances
US20060099611A1 (en) In vivo affinity maturation scheme
JPWO2005110433A1 (en) Non-human animals in which Bach2 expression is artificially suppressed and their use

Legal Events

Date Code Title Description
AS Assignment

Owner name: STRATAGENE, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:WIGLER, MICHAEL H.;SORGE, JOSEPH A.;REEL/FRAME:014638/0057;SIGNING DATES FROM 19950406 TO 19950413

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: CATALYST ASSETS LLC, WYOMING

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SORGE, JOSEPH A.;AGILENT TECHNOLOGIES, INC.;AGILENT TECHNOLOGIES RESEARCH CORP.;AND OTHERS;REEL/FRAME:028009/0112

Effective date: 20120322