US20050163708A1 - Chimeric antibody with specificity to human B cell surface antigen - Google Patents

Chimeric antibody with specificity to human B cell surface antigen Download PDF

Info

Publication number
US20050163708A1
US20050163708A1 US10/941,768 US94176804A US2005163708A1 US 20050163708 A1 US20050163708 A1 US 20050163708A1 US 94176804 A US94176804 A US 94176804A US 2005163708 A1 US2005163708 A1 US 2005163708A1
Authority
US
United States
Prior art keywords
human
cells
antibody
molecule
chain
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
US10/941,768
Inventor
Randy Robinson
Alvin Liu
Jeffrey Ledbetter
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.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PCT/US1986/002269 external-priority patent/WO1987002671A1/en
Priority claimed from US07/665,939 external-priority patent/US5500362A/en
Application filed by Individual filed Critical Individual
Priority to US10/941,768 priority Critical patent/US20050163708A1/en
Publication of US20050163708A1 publication Critical patent/US20050163708A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2887Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6875Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody being a hybrid immunoglobulin
    • A61K47/6877Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody being a hybrid immunoglobulin the antibody being an immunoglobulin containing regions, domains or residues from different species
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • C07K16/461Igs containing Ig-regions, -domains or -residues form different species
    • C07K16/462Igs containing a variable region (Fv) from one specie and a constant region (Fc) from another
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/734Complement-dependent cytotoxicity [CDC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/40Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation

Definitions

  • This invention relates to recombinant DNA methods of preparing an antibody with specificity for an antigen on the surface of human B cells, genetic sequences coding therefor, as well as methods of obtaining such sequences.
  • mouse monoclonal antibodies may lack the ability to efficiently interact with human effector cells as assessed by functional assays such as antibody-dependent cellular cytotoxicity (ADCC) and complement-mediated cytolysis (CDC).
  • ADCC antibody-dependent cellular cytotoxicity
  • CDC complement-mediated cytolysis
  • the present invention bridges both the hybridoma and genetic engineering technologies to provide a quick and efficient method, as well as products derived therefrom, for the production of a chimeric human/non-human antibody.
  • the chimeric antibodies of the present invention embody a combination of the advantageous characteristics of monoclonal antibodies derived from mouse-mouse hybridomas and of human monoclonal antibodies.
  • the chimeric monoclonal antibodies like mouse monoclonal antibodies, can recognize and bind to a human target antigen; however, unlike mouse monoclonal antibodies, the species-specific properties of the chimeric antibodies will avoid the induction of harmful hypersensitivity reactions and may allow for resistance to clearance when used in humans in vivo.
  • the inclusion of appropriate human immunoglobulin sequences can result in an antibody which efficiently interacts with human effector cells in vivo to cause tumor cell lysis and the like.
  • any desired antibody isotype can be conferred upon a particular antigen combining site.
  • Boulianne G. L. et al., Nature, 312: 643 (December 13, 1984), also produced antibodies consisting of mouse variable regions joined to human constant regions. They constructed immunoglobulin genes in which the DNA segments encoding mouse variable regions specific for the hapten trinitrophenyl (TNP) were joined to segments encoding human mu and kappa constant regions. These chimeric genes were expressed as functional TNP binding chimeric IgM.
  • TNP hapten trinitrophenyl
  • NP 4-hydroxy-3-nitrophenacetyl
  • Taniguchi M., in European Patent Publication No. 171 496 (published Feb. 19, 1985) discloses the production of chimeric antibodies having variable regions with tumor specificty derived from experimental animals, and constant regions derived from human.
  • the corresponding heavy and light chain genes are produced in the genomic form, and expressed in mammalian cells.
  • Boss, M. A., European Patent Application 120694 shows expression in E. coli of non-chimeric immunoglobulin chains with 4-nitrophenyl specificity. There is a broad description of chimeric antibodies but no details (see p. 9).
  • Seno M. et al., Nucleic Acids Research, 11: 719-726 (1983), describe the preparation of a cDNA sequence and recombinant plasmids containing the same coding for part of the variable region and all of the constant region of the human IgE heavy chain (epsilon chain).
  • the invention provides a genetically engineered chimeric antibody of desired variable region specificity and constant region properties, through gene cloning and expression of light and heavy chains.
  • the cloned immunoglobulin gene products can be produced by expression in genetically engineered cells.
  • the invention provides cDNA sequences coding for immunoglobulin chains comprising a constant human region and a variable, non-human, region.
  • the immunoglobulin chains can be either heavy or light.
  • the invention provides gene sequences coding for immunoglobulin chains comprising a cDNA variable region of the desired specificity. These can be combined with genomic constant regions of human origin.
  • the invention provides sequences as above, present in recombinant DNA molecules in vehicles such as plasmid vectors, capable of expression in desired prokaryotic or eukaryotic hosts.
  • the invention provides hosts capable of producing, by culture, the chimeric antibodies and methods of using these hosts.
  • the invention also provides individual chimeric immunoglobulin individual chains, as well as complete assembled molecules having human constant regions and variable regions with a human B cell surface antigen specificity, wherein both variable regions have the same binding specificity.
  • immunoglobulin chains and/or molecules provided by the invention are:
  • the invention also provides for a genetic sequence, especially a cDNA sequence, coding for the variable region of desired specificity of an antibody molecule heavy and/or light chain, operably linked to a sequence coding for a polypeptide different than an immunoglobulin chain (e.g., an enzyme).
  • a genetic sequence especially a cDNA sequence, coding for the variable region of desired specificity of an antibody molecule heavy and/or light chain, operably linked to a sequence coding for a polypeptide different than an immunoglobulin chain (e.g., an enzyme).
  • cDNA sequences are particularly advantageous over genomic sequences (which contain introns), in that cDNA sequences can be expressed in bacteria or other hosts which lack appropriate RNA splicing systems.
  • FIG. 1 shows the DNA rearrangements and the expression of immunoglobulin mu and gamma heavy chain genes. This is a schematic representation of the human heavy chain gene complex (not shown to scale). Heavy chain variable V region formation occurs through the proper joining of V H , D and J H gene segments. This generates an active mu gene. A different kind of DNA rearrangement called “class switching” relocates the joined V H , D and J H region from the vicinity of mu constant C region to that of another heavy chain C region (switching to gamma is diagrammed here).
  • FIG. 2 shows the known nucleotide sequences of human and mouse J regions. Consensus sequences for the J regions are shown below the actual sequences.
  • the oligonucleotide sequence below the mouse kappa J region consensus sequence is a Universal Immunoglobulin Gene (UIG) oligonucleotide. Note that there are only a few J regions with relatively conserved sequences, especially near the constant regions, in each immunoglobulin gene locus.
  • UAG Universal Immunoglobulin Gene
  • FIG. 3 shows the nucleotide sequences of the mouse J regions. Shown below are the oligonucleotide primers UIG-H and UIG-K. Note that each contains a restriction enzyme site. They can be used as primers for the synthesis of cDNA complementary to the variable region of mRNA, and can also be used to mutagenize, in vitro, cloned cDNA.
  • FIG. 4 Human Constant Domain Module .
  • the ApaI site is 16 nucleotide residues into the C H 1 coding domain of Human gamma 1; and is used in a previous construction of a mouse-human chimeric heavy-chain immunoglobulin.
  • the BstEII site is in the J H region, and is used in the construction described in this application.
  • the human C K clone, pGML60 is a composite of two cDNA clones, one isolated from GM1500 (pK2-3), the other from GM2146 (pGML1).
  • the J K -C K junction sequence shown comes from pK2-3.
  • In vitro mutagenesis using the oligonucleotide, J K HindIII was carried out to engineer a HindIII site 14 nucleotide residues 5′ of the J-C junction. This changes a human GTG codon into a CTT codon.
  • FIG. 5 shows the nucleotide sequence of the V region of the 2H7 V H cDNA clone pH2-11. The sequence was determined by the dideoxytermination method using M13 subclones of gene fragments. Open circles denote amino acid residues confirmed by peptide sequence. A sequence homologous to D SP.2 in the CDR3 region is underlined. The NcoI site at 5′ end was converted to a SalI site by using SalI linkers.
  • FIG. 6 shows the nucleotide sequence of the V region of the 2H7 V K cDNA clone pL2-12.
  • the oligonucleotide primer used for site-directed mutagenesis is shown below the J K 5 segment. Open circles denote amino acid residues confirmed by peptide sequence.
  • FIG. 7 shows the construction of the light and heavy chain expression plasmids pING2106 (panel a) and pING2101 (panel B).
  • the SalI to BamHI fragment from pING2100 is identical to the SalI to BamHI fragment from pING2012E (see panel C).
  • a linear representation of the circular plasmid pING2012E is shown in panel C.
  • the 6.6 Kb SalI to BamHI fragment contains sequences from pSV2-neo, puc12, M8alphaRX12, and pL1.
  • the HindIII site in pSV2-neo was destroyed before assembly of pING2012E by HindIII cleavage, fill-in, and religation.
  • FIG. 8 shows the structure of several chimeric 2H7-V H expression plasmids.
  • pING2107 is a qpt version of the light chain plasmid, pING2106.
  • the larger ones are two-gene plasmids.
  • pHL2-11 and pHL2-26 contain both H and L genes, while pLL2-25 contains two L genes. They were constructed by joining an NdeI fragment containing either an H or L gene to partially digested (with NdeI) pING2106.
  • FIG. 9 shows a summary of the sequence alterations made in the construction of the 2H7 chimeric antibody expression plasmids. Residues underlined in the 5′ untranslated region are derived from the cloned mouse kappa and heavy-chain genes. Residues circled in the V/C boundary result from mutagenesis operations to engineer restriction enzyme sites in this region.
  • antibodies are composed of two light and two heavy chain molecules.
  • Light and heavy chains are divided into domains of structural and functional homology.
  • the variable domains of both the light (V L ) and the heavy (V H ) chains determine recognition and specificity.
  • the constant region domains of light (C L ) and heavy (C H ) chains confer important biological properties such as antibody chain association, secretion, transplacental mobility, complement binding, and the like.
  • V H , D and J H ; or V L and J L germ line gene segments
  • V H , D and J H ; or V L and J L germ line gene segments
  • V L -J L and V H -D-J H V segments are joined by DNA rearrangements to form the complete V regions expressed in heavy and light chains respectively ( FIG. 1 ).
  • the rearranged, joined (V L -J L and V H -D-J H )V segments then encode the complete variable regions or antigen binding domains of light and heavy chains, respectively.
  • Expression vector a plasmid DNA containing necessary regulatory signals for the synthesis of mRNA derived from any gene sequence, inserted into the vector.
  • Module vector a plasmid DNA containing a constant or variable region gene module.
  • Expression plasmid an expression vector that contains an inserted gene, such as a chimeric immunoglobulin gene.
  • the invention provides a novel approach for the cloning and production of a human/mouse chimeric antibody with specificity to a human B cell surface antigen.
  • the antigen is a polypeptide or comprises a polypeptide bound by the 2H7 monoclonal antibody described in Clark et al. Proc. Natl. Acad. Sci., U.S.A. 82: 1766-1770 (1985).
  • This antigen is a phosphoprotein designated (Bp35(CD20)) and is only expressed on cells of the B cell lineage.
  • Murine monoclonal antibodies to this antigen have been made before and are described in Clark et al., supra; see also Stashenko, P., et al., J. Immunol. 125: 1678-1685 (1980).
  • the method of production combines five elements:
  • One common feature of all immunoglobulin light and heavy chain genes and the encoded messenger RNAs is the so-called J region (i.e. joining region, see FIG. 1 ).
  • Heavy and light chain J regions have different sequences, but a high degree of sequence homology exists (greater than 80%) especially near the constant region, within the heavy J H regions or the kappa light chain J regions. This homology is exploited in this invention and consensus sequences of light and heavy chain J regions were used to design oligonucleotides (designated herein as UIGs) for use as primers or probes for cloning immunoglobulin light or heavy chain mRNAs or genes ( FIG. 3 ).
  • UIGs oligonucleotides
  • UIG may be capable of hybridizing to all immunoglobulin mRNAs or a specific one containing a particular J sequence.
  • Another utility of a particular UIG probe may be hybridization to light chain or heavy chain mRNAs of a specific constant region, such as UIG-MJK which detects all mouse J K -containing sequences ( FIG. 2 ).
  • UIG design can also include a sequence to introduce a restriction enzyme site into the cDNA copy of an immunoglobulin gene (see FIG. 3 ).
  • the invention may, for example, utilize chemical gene synthesis to generate the UIG probes for the cloning and modification of V regions from immunoglobulin mRNA.
  • oligonucleotides can be synthesized to recognize individually, the less conserved 5′-region of the J regions as a diagnostic aid in identifying the particular J region present in the immunoglobulin mRNA.
  • a multi-step procedure is utilized for generating complete V+C region cDNA clones from the hybridoma cell light and heavy chain mRNAs.
  • Clones are screened by hybridization with UIG oligonucleotide probes. Positive heavy and light chain clones identified by this screening procedure are mapped and sequenced to select those containing V region and leader coding sequences.
  • In vitro mutagenesis including, for example, the use of UIG oligonucleotides, is then used to engineer desired restriction enzyme cleavage sites. We used this approach for the chimeric 2H7 light chain.
  • An expedient method is to use synthetic UIG oligonucleotides as primers for the synthesis of cDNA.
  • This method has the advantage of simultaneously introducing a desired restriction enzyme site, such as BstEII ( FIG. 3 ) into a V region cDNA clone. We used this approach for the chimeric 2H7 heavy chain.
  • cDNA constant region module vectors are prepared from human cells. These cDNA clones are modified, when necessary, by site-directed mutagenesis to place a restriction site at the analogous position in the human sequence or at another desired location near a boundary of the constant region.
  • An alternative method utilizes genomic C region clones as the source for C region module vectors.
  • cloned V region segments generated as above are excised and ligated to light or heavy chain C region module vectors. For example, one can clone the complete human kappa light chain C region and the complete human gamma 1 C region. In addition, one can modify the human gamma 1 region to introduce a termination codon and thereby obtain a gene sequence which encodes the heavy chain portion of an Fab molecule.
  • Operationally linked means in-frame joining of coding sequences to derive a continuously translatable gene sequence without alterations or interruptions of the triplet reading frame.
  • cDNA genetic sequences in the present invention are the fact that they code continuously for immunoglobulin chains, either heavy or light.
  • continuous is meant that the sequences do not contain introns (i.e. are not genomic sequences, but rather, since derived from mRNA by reverse transcription, are sequences of contiguous exons).
  • This characteristic of the cDNA sequences provided by the invention allows them to be expressible in prokaryotic hosts, such as bacteria, or in lower eukaryotic hosts, such as yeast.
  • Another advantage of using cDNA cloning method is the ease and simplicity of obtaining variable region gene modules.
  • variable and variable are used functionally to denote those regions of the immunoglobulin chain, either heavy or light chain, which code for properties and features possessed by the variable and constant regions in natural non-chimeric antibodies. As noted, it is not necessary for the complete coding region for variable or constant regions to be present, as long as a functionally operating region is present and available.
  • Expression vehicles include plasmids or other vectors. Preferred among these are vehicles carrying a functionally complete human constant heavy or light chain sequence having appropriate restriction sites engineered so that any variable-heavy or light chain sequence with appropriate cohesive ends can be easily inserted thereinto. Human constant heavy or light chain sequence-containing vehicles are thus an important embodiment of the invention. These vehicles can be used as intermediates for the expression of any desired complete heavy or light chain in any appropriate host.
  • yeast provides substantial advantages for the production of immunoglobulin light and heavy chains. Yeasts carry out post-translational peptide modifications including glycosylation. A number of recombinant DNA strategies now exist which utilize strong promoter sequences and high copy number plasmids which can be used for overt production of the desired proteins in yeast. Yeast recognizes leader sequences on cloned mammalian gene products and secretes peptides bearing leader sequences (i.e. prepeptides) (Hitzman, et al., 11th International Conference on Yeast, Genetics and Molecular Biology, Montpelier, France, Sep. 13-17, 1982).
  • Yeast gene expression systems can be routinely evaluated for the level of heavy and light chain production, protein stability, and secretion. Any of a series of yeast gene expression systems incorporating promoter and termination elements from the actively expressed genes coding for glycolytic enzymes produced in large quantities when yeasts are grown in mediums rich in glucose can be utilized. Known glycolytic genes can also provide very efficient transcription control signals. For example, the promoter and terminator signals of the iso-1-cytochrome C (CYC-1) gene can be utilized.
  • CYC-1-cytochrome C CYC-1-cytochrome C
  • the following approach can be taken to develop and evaluate optimal expression plasmids for the expression of cloned immunoglobulin cDNAs in yeast.
  • E. coli K12 strain 294 (ATCC 31446) is particularly useful.
  • Other microbial strains which may be used include E. coli X1776 (ATCC 31537).
  • the aforementioned strains, as well as E. coli W3110 (ATCC 27325) and other enterobacteria such as Salmonella typhimurium or Serratia marcescens , and various Pseudomonas species may be used.
  • plasmid vectors containing replicon and control sequences which are derived from species compatible with a host cell are used in connection with these hosts.
  • the vector ordinarily carries a replication site, as well as specific genes which are capable of providing phenotypic selection in transformed cells.
  • E. coli is readily transformed using pBR322, a plasmid derived from an E. coli species (Bolivar, et al., Gene, 2: 95 (1977)).
  • pBR322 contains genes for ampicillin and tetracycline resistance, and thus provides easy means for identifying transformed cells.
  • the pBR322 plasmid or other microbial plasmids must also contain, or be modified to contain, promoters which can be used by the microbial organism for expression of its own proteins.
  • promoters most commonly used in recombinant DNA construction include the beta-lactamase (penicillinase) and lactose (beta-galactosidase) promoter systems (Chang et al., Nature, 275: 615 (1978); Itakura et al., Science, 198: 1056 (1977)); and tryptophan promoter systems (Goeddel et al., Nucleic Acids Research, 8: 4057 (1980); EPO Publication No. 0036776). While these are the most commonly used, other microbial promoters have been discovered and utilized.
  • a genetic construct for any heavy or light chimeric immunoglobulin chain can be placed under the control of the leftward promoter of bacteriophage lambda (P L ).
  • This promoter is one of the strongest known promoters which can be controlled. Control is exerted by the lambda repressor, and adjacent restriction sites are known.
  • lactose dependent E. coli chromosomal DNA comprises a lactose or lac operon which mediates lactose digestion by elaborating the enzyme beta-galactosidase.
  • the lac control elements may be obtained from bacteriophage lambda pLAC5, which is infective for E. coli .
  • the lac promoter-operator system can be induced by IPTG.
  • promoter/operator systems or portions thereof can be employed as well.
  • arabinose, colicine E1, galactose, alkaline phosphatase, tryptophan, xylose, tac, and the like can be used.
  • Mammalian cells are mammalian cells, grown in vitro in tissue culture, or in vivo in animals. Mammalian cells provide post-translational modifications to immunoglobulin protein molecules including leader peptide removal, correct folding and assembly of heavy and light chains, proper glycosylation at correct sites, and secretion of functional antibody protein.
  • Mammalian cells which may be useful as hosts for the production of antibody proteins include cells of lymphoid origin, such as the hybridoma Sp2/0-Ag14 (ATCC CRL 1581) or the myleoma P3X63Ag8 (ATCC TIB 9), and its derivatives. Others include cells of fibroblast origin, such as Vero (ATCC CRL 81) or CHO-K1 (ATCC CRL 61).
  • Several possible vector systems are available for the expression of cloned heavy chain and light chain genes in mammalian cells.
  • One class of vectors relies upon the integration of the desired gene sequences into the host cell genome.
  • Cells which have stably integrated DNA can be selected by simultaneously introducing drug resistance genes such as E. coli gpt (Mulligan, R. C. and Berg, P., Proc. Natl. Acad. Sci., USA, 78: 2072 (1981)) or Tn5 neo (Southern, P. J. and Berg, P., J. Mol. Appl. Genet., 1: 327 (1982)).
  • the selectable marker gene can be either linked to the DNA gene sequences to be expressed, or introduced into the same cell by co-transfection (Wigler, M. et al., Cell, 16: 77 (1979)).
  • a second class of vectors utilizes DNA elements which confer autonomously replicating capabilities to an extrachromosomal plasmid. These vectors can be derived from animal viruses, such as bovine papillomavirus (Sarver, N. et al., Proc. Natl. Acad. Sci., USA, 79: 7147 (1982)), polyoma virus (Deans, R. J. et al., Proc. Natl. Acad. Sci., USA, 81: 1292 (1984)), or SV40 virus (Lusky, M. and Botchan, M., Nature, 293: 79 (1981)).
  • an immunoglobulin cDNA is comprised only of sequences representing the mature mRNA encoding an antibody protein
  • additional gene expression elements regulating transcription of the gene and processing of the RNA are required for the synthesis of immunoglobulin mRNA.
  • These elements may include splice signals, transcription promoters, including inducible promoters enhancers, and termination signals.
  • cDNA expression vectors incorporating such elements include those described by Okayama, H. and Berg, P., Mol. Cell Biol., 3: 280 (1983); Cepko, C. L. et al., Cell, 37: 1053 (1984); and Kaufman, R. J., Proc. Natl. Acad. Sci., USA, 82: 689 (1985).
  • mammalian cells may express chimeric immunoglobulin genes which are comprised of a variable region cDNA module plus a constant region which is composed in whole or in part of genomic sequences.
  • a variable region cDNA module plus a constant region which is composed in whole or in part of genomic sequences.
  • human constant region genomic clones have been described (Ellison, J. W. et al., Nucl. Acids Res., 10: 4071 (1982), or Max, E. et al., Cell, 29: 691 (1982)).
  • the use of such genomic sequences may be convenient for the simultaneous introduction of immunoglobulin enhancers, splice signals, and transcription termination signals along with the constant region gene segment.
  • the invention provides “chimeric” immunoglobulin chains, either heavy or light.
  • a chimeric chain contains a constant region substantially similar to that present in a natural human immunoglobulin, and a variable region having the desired antigenic specificity of the invention, i.e., to the specified human B cell surface antigen.
  • the invention also provides immunoglobulin molecules having heavy and light chains associated so that the overall molecule exhibits any desired binding and recognition properties.
  • immunoglobulin molecules having heavy and light chains associated so that the overall molecule exhibits any desired binding and recognition properties.
  • Various types of immunoglobulin molecules are provided: monovalent, divalent, molecules with chimeric heavy chains and non-chimeric light chains, or molecules with the invention's variable binding domains attached to moieties carrying desired functions.
  • Antibodies having chimeric heavy chains of the same or different variable region binding specificity and non-chimeric (i.e., all human or all non-human) light chains can be prepared by appropriate association of the needed polypeptide chains. These chains are individually prepared by the modular assembly methods of the invention.
  • the antibodies of the invention having human constant region can be utilized for passive immunization, especially in humans, without negative immune reactions such as serum sickness or anaphylactic shock.
  • the antibodies can, of course, also be utilized in prior art immunodiagnostic assays and kits in detectably labelled form (e.g., enzymes, 125 I, 14 C, fluorescent labels, etc.), or in immunobilized form (on polymeric tubes, beads, etc.), in labelled form for in vivo imaging, wherein the label can be a radioactive emitter, or an NMR contrasting agent such as a carbon-13 nucleus, or an X-ray contrasting agent, such as a heavy metal nucleus.
  • the antibodies can also be used for in vitro localization of the antigen by appropriate labelling.
  • the antibodies can be used for therapeutic purposes, by themselves, in complement mediated lysis, or coupled to toxins or therapeutic moieties, such as ricin, etc.
  • Mixed antibody-enzyme molecules can be used for immunodiagnostic methods, such as ELISA.
  • Mixed antibody-peptide effector conjugates can be used for targeted delivery of the effector moiety with a high degree of efficacy and specificity.
  • the chimeric antibodies of this invention can be used for any and all uses in which the murine 2H7 monoclonal antibody can be used, with the obvious advantage that the chimeric ones are more compatible with the human body.
  • the human cell lines GM2146 and GM1500 were obtained from the Human Mutant Cell Repository (Camden, N.J.) and cultured in RPMI1640 plus 10% fetal bovine serum (M. A. Bioproducts).
  • the cell line Sp2/0 was obtained from the American Type Culture Collection and grown in Dulbecco's Modified Eagle Medium (DMEM) plus 4.5 g/l glucose (M. A. Bioproducts) plus 10% fetal bovine serum (Hyclone, Sterile Systems, Logan, Utah). Media were supplemented with penicillin/streptomycin (Irvine Scientific, Irvine, California).
  • the plasmids pBR322, pL1 and pUC12 were purchased from Pharmacia P-L Biochemicals (Milwaukee, Wisconsin).
  • the plasmids pSV2-neo and pSV2-qpt were obtained from BRL (Gaithersburg, Md.), and are available from the American Type Culture Collection (Rockville, Md.).
  • pHu-gamma-1 is a subclone of the 8.3 Kb HindIII to BamHI fragment of the human IgG1 chromosomal gene. An isolation method for of the human IgG1 chromosomal gene is described by Ellison, J. W. et al., Nucl. Acids Res., 10: 4071 (1982).
  • M8alphaRX12 contains the 0.7 Kb XbaI to EcoRI fragment containing the mouse heavy chain enhancer from the J-C intron region of the M603 chromosomal gene (Davis, M. et al., Nature, 283: 733, 1979) inserted into M13mp10.
  • DNA manipulations involving purification of plasmid DNA by buoyant density centrifugation, restriction endonuclease digestion, purification of DNA fragments by agarose gel electrophoresis, ligation and transformation of E. coli were as described by Maniatis, T. et al., Molecular Cloning: A Laboratory Manual , (1982) or other procedures. Restriction endonucleases and other DNA/RNA modifying enzymes were purchased from Boehringer-Mannheim (Indianapolis, Ind.), BRL, New England Biolabs (Beverly, Mass.) and Pharmacia P-L.
  • Oligonucleotides were either synthesized by the triester method of Ito et al. ( Nucl. Acids Res., 10: 1755 (1982)), or were purchased from ELESEN, Los Angeles, Calif. Tritylated, deblocked oligonucleotides were purified on Sephadex-G50, followed by reverse-phase HPLC with a 0-25% gradient of acetonitrile in 10 mM triethylamine-acetic acid, pH 7.2, on a C18 Bondapak column (Waters Associates). Detritylation was in 80% acetic acid for 30 min., followed by evaporation thrice. Oligonucleotides were labeled with [gamma- 32 P]ATP by T4 polynucleotide kinase.
  • Total cellular RNA was prepared from tissue culture cells by the method of Auffray, C. and Rougeon, F. ( Eur. J. Biochem., 107: 303 (1980)) or Chirgwin, J. M. et al. ( Biochemistry, 18: 5294 (1979)). Preparation of poly(A) + RNA, methyl-mercury agarose gel electrophoresis, and “Northern” transfer to nitrocellulose were as described by Maniatis, T. et al., supra. Total cellular RNA or poly(A) + RNA was directly bound to nitrocellulose by first treating the RNA with formaldehyde (White, B. A. and Bancroft, F. C., J. Biol.
  • Hybridization to filterbound RNA was with nick-translated DNA fragments using conditions described by Margulies, D. H. et al. ( Nature, 295: 168 (1982)) or with 32 P-labelled oligonucleotide using 4 ⁇ SSC, 10 ⁇ Denhardt's, 100 ug/ml salmon sperm DNA at 37° C. overnight, followed by washing in 4 ⁇ SSC at 37° C.
  • Oligo-dT primed cDNA libraries were prepared from poly(A) + RNA from GM1500 and GM2146 cells by the methods of Land, H. et al. ( Nucl. Acids Res., 9: 2251 (1981)) and Gubler, V. and Hoffman, B. J., Gene, 25: 263 (1983), respectively.
  • the cDNA libraries were screened by hybridization (Maniatis, T., supra) with 32 P-labelled oligonucleotides using the procedure of de Lange et al. ( Cell, 34: 891 (1983)), or with nick-translated DNA fragments.
  • Poly(A) + RNA (20 ug) was mixed with 1.2 ug primer in 40 ul of 64 mM KCl. After denaturation at 90° C. for 5 min. and then chilling in ice, 3 units Human Placental Ribonuclease Inhibitor (BRL) was added in 3 ul of 1M Tris-HCl, pH 8.3. The oligonucleotide was annealed to the RNA at 42° C. for 15 minutes, then 12 ul of 0.05M DTT, 0.05M MgCl 2 , and 1 mM each of dATP, dTTP, dCTP, and dGTP was added. 2 ul of alpha- 32 P-dATP (400 Ci/mmol, New England Nuclear) was added, followed by 3 ul of AMV reverse transcriptase (19 units/ul, Life Sciences).
  • BRL Human Placental Ribonuclease Inhibitor
  • RNA-DNA hybrid was extracted with phenol, then with chloroform, and precipitated with ethanol.
  • Second strand synthesis, homopolymer tailing with dGTP or dCTP, and insertion into homopolymer tailed vectors was as described by Gubler and Hoffman, supra.
  • Single stranded M13 subclone DNA (1 ug) was combined with 20 ng oligonucleotide primer in 12.5 ul of Hin buffer (7 mM Tris-HCl, pH 7.6, 7 mM MgCl 2 , 50 mM NaCl). After heating to 95° C. in a sealed tube, the primer was annealed to the template by slowly cooling from 70° C. to 37° C. for 90 minutes. 2 ul dNTPs (1 mM each), 1 ul 32 P-DATP (10 uCi), 1 ul DTT (0.1 M) and 0.4 ul Klenow DNA PolI (2u, Boehringer Mannheim) were added and chains extended at 37° C. for 30 minutes.
  • Hin buffer 7 mM Tris-HCl, pH 7.6, 7 mM MgCl 2 , 50 mM NaCl.
  • Cytoplasmic protein extract was prepared by vortexing 10 6 cells in 160 ul of 1% NP40, 0.15 M NaCl, 10 mM Tris, 1 mM EDTA, pH 7.6 and leaving the lysate at 0° C., 15 minutes, followed by centrifugation at 10,600 ⁇ g to remove insoluble debris.
  • a double antibody sandwich ELISA (Voller, A. et al., in Manual of Clinical Immunology, 2nd Ed., Eds. Rose, N. and Friedman, H., pp. 359-371, 1980) using affinity purified antisera was used to detect specific immunoglobulins.
  • the plate-bound antiserum is goat anti-human IgG (KPL, Gaithersburg, Md.) at 1/1000 dilution, while the peroxidase-bound antiserum is goat anti-human IgG (KPL or Tago, Burlingame) at 1/4000 dilution.
  • the plate-bound antiserum is goat anti-human kappa (Tago) at 1/500 dilution, while the peroxidase-bound antiserum is goat anti-human kappa (Cappel) at 1/1000 dilution.
  • the 2H7 mouse monoclonal antibody (gamma 2b, kappa) recognizes a human B-cell surface antigen, (Bp35(CD20)) Clark, E. A., et al., Proc. Natl. Acad. Sci., U.S.A. 82: 1766 (1985)).
  • the (Bp35(CD20)) molecules presumably play a role in B-cell activation.
  • the antibody 2H7 does not react with stem cells which are progenitors of B-cells epithelial, mesenchymal and fibroblastic cells of other organs.
  • Frozen cells were thawed on ice for 10 minutes and then at room temperature. The suspension was diluted with 15 ml PBS and the cells were centrifuged down. They were resuspended, after washes in PBS, in 16 ml 3M LiCl, 6M urea and disrupted in a polytron shear. The preparation of mRNA and the selection of the poly(A+) fraction were carried out according to Auffray, C. and Rougeon, F., Eur. J. Biochem. 107: 303, 1980.
  • the poly (A+) RNA from 2H7 was hybridized individually with labeled J H 1, J H 2, J H 3 and J H 4 oligonucleotides under conditions described by Nobrega et al. Anal. Biochem 131: 141, 1983). The products were then subjected to electrophoresis in a 1.7% agarose-TBE gel. The gel was fixed in 10% TCA, blotted dry and exposed for autoradiography. The result showed that the 2H7 V H contains J H 1, J H 2, or J H 4 but not J H 3 sequences.
  • V K mRNA For the analysis of the V K mRNA, the dot-blot method of White and Bancroft J. Biol. Chem. 257: 8569, (1982) was used. Poly (A+) RNA was immobilized on nitrocellulose filters and was hybridized to labeled probe-oligonucleotides at 400 in 4 ⁇ SSC. These experiments show that 2H7 contains J K 5 sequences.
  • a library primed by oligo (dT) on 2H7 poly (A+) RNA was screened for kappa clones with a mouse C K region probe. From the 2H7 library, several clones were isolated. A second screen with a 5′ J K 5 specific probe identified the 2H7 (J K 5) light-chain clones. Heavy chain clones of 2H7 were generated by priming the poly(A+) RNA with the UIGH(BstEII) oligonucleotide (see FIG. 3 ), and identified by screening with the UIGH(BstEII) oligonucleotide.
  • the heavy and light chain genes or gene fragments from the V H and V K cDNA clones pH2-11 and pL2-12 were inserted into M13 bacteriophage vectors for nucleotide sequence analysis.
  • the complete nucleotide sequences of the variable region of these clones were determined ( FIGS. 5 and 6 ) by the dideoxy chain termination method. These sequences predict V region amino acid compositions that agree well with the observed compositions, and predict peptide sequences which have been verified by direct amino acid sequencing of portions of the V regions.
  • the nucleotide sequences of the cDNA clones show that they are immunoglobulin V region clones as they contain amino acid residues diagnostic of V domains (Kabat et al., Sequences of Proteins of Immunological Interest; U.S. Dept of HHS, 1983).
  • the 2H7 V H has the J H 1 sequence.
  • the 2H7 V L is from the V K -KpnI family (Nishi et al. Proc. Nat. Acd. Sci. USA 82: 6399, 1985), and uses J K 5.
  • the cloned 2H7 V L predicts an amino acid sequence which was confirmed by amino acid sequencing of peptides from the 2H7 light chain corresponding to residues 81-100.
  • the cloned 2H7 V H predicts an amino acid sequence confirmed also by peptide sequencing, namely residues 1-12.
  • the J-region mutagenesis primer J K HindIII As shown in FIG. 6 , was utilized.
  • a human C K module derived from a cDNA clone was also mutagenized to contain the HindIII sequence (see FIG. 4 ).
  • the mutagenesis reaction was performed on M13 subclones of these genes. The frequency of mutant clones ranged from 0.5 to 1% of the plaques obtained.
  • oligo (dC) sequence upstream of the AUG codon in a V H chimeric gene interferes with proper splicing in one particular gene construct. It was estimated that perhaps as much as 70% of the RNA transcripts had undergone the mis-splicing, wherein a cryptic 3′ splice acceptor in the leader sequence was used. Therefore the oligo (dC) sequence upstream of the initiator AUG was removed in all of the clones.
  • an oligonucleotide was used which contains a SalI restriction site to mutagenize the 2H7 V K clone.
  • the primer used for this oligonucleotide-directed mutagenesis is a 22-mer which introduces a SalI site between the oligo (dC) and the initiator met codon ( FIG. 6 ).
  • the human C gamma 1 gene module is a cDNA derived from GM2146 cells (Human Genetic Mutant Cell Repository, Newark, N.J.). This C gamma 1 gene module was previously combined with a mouse V H gene module to form the chimeric expression plasmid pING2012E ( FIG. 7C ).
  • a 2H7 chimeric heavy chain expression plasmid was derived from the replacement of the V H module of pING2012E with the V H cDNA modules to give the expression plasmid pING2101 ( FIG. 7 b ). This plasmid directs the synthesis of chimeric 2H7 heavy chain when transfected into mammalian cells.
  • the SalI to HindIII fragment of the mouse V K module was joined to the human C K module by the procedure outlined in FIG. 7 a , forming pING2106.
  • Replacement of the neo sequence with the E. coli gpt gene derived from pSV2-gpt resulted in pING2107, which expresses 2H7 chimeric light chain and confers mycophenolic acid resistance when transfected into mammalian cells.
  • both heavy and light chain chimeric genes in the same plasmid allows for the introduction into transfected cells of a 1:1 gene ratio of heavy and light chain genes leading to a balanced gene dosage. This may improve expression and decrease manipulations of transfected cells for optimal chimeric antibody expression.
  • the DNA fragments derived from the chimeric heavy and light chain genes of pING2101 and pING2106 were combined into the expression plasmids pHL2-11 and pHL2-26 ( FIG. 8 ).
  • This expression plasmid contains a selectable neoR marker and separate transcription units for each chimeric gene, each including a mouse heavy chain enhancer.
  • Electroporation was used (Potter et al. supra; Toneguzzo et al. Mol. Cell Biol. 6: 703 1986) for the introduction of 2H7 chimeric expression plasmid DNA into mouse Sp2/0 cells.
  • the electroporation technique gave a transfection frequency of 10 4 ⁇ 10 5 for the Sp2/0 cells.
  • the expression plasmids, pING2101 and pING2106, were digested with NdeI; and the DNA was introduced into Sp2/0 cells by electroporation.
  • Transformant 1D6 was obtained which secretes chimeric 2H7 antibody.
  • Antibody isolated from this cell line was used for the functional assays done to characterize the chimeric antibody.
  • 1D6 Sp2/0.pING2101/pING2106.1D6 cells were grown in culture medium [DMEM (Gibco #320-1965), supplemented with 10% Fetal Bovine Serum (Hyclone #A-1111-D), 10 mM HEPES, 1 ⁇ Glutamine-Pen-Strep (Irvine Scientific #9316) to 1 ⁇ 10 6 cell/ml.
  • DMEM Gibco #320-1965
  • Fetal Bovine Serum Hyclone #A-1111-D
  • 10 mM HEPES 1 ⁇ Glutamine-Pen-Strep (Irvine Scientific #9316) to 1 ⁇ 10 6 cell/ml.
  • the cells were then centrifuged at 400 ⁇ g and resuspended in serum-free culture medium at 2 ⁇ 10 6 cell/ml for 18-24 hr.
  • the medium was centrifuged at 4000 RPM in a JS-4.2 rotor (3000 ⁇ g) for 15 min.
  • the supernatant was centrifuged at 2000 ⁇ g, 5 min., and then loaded onto a 40 ml DEAE column, which was preequilibrated with 10 mM sodium phosphate, pH8.0.
  • the bulk of the IgG was in the pH 3.5 elution and was pooled and concentrated over Centricon 30 (Amicon Corp.) to approximately 0.06 ml.
  • the buffer was changed to PBS (10 mM sodium phosphate pH 7.4, 0.15M NaCl) in Centricon 30 by repeated diluting with PBS and reconcentrating.
  • the IgG solution was then adjusted to 0.10 ml and bovine serum albumin (Fraction V, U.S. Biochemicals) was added to 1.0% as a stabilizing reagent.
  • the samples were tested with a binding assay, in which cells of both an 2H7 antigen-positive and an 2H7 antigen-negative cell line were incubated with standard mouse monoclonal antibody 2H7 with chimeric 2H7 antibody derived from the cell culture supernatants, followed by a second reagent, fluoresceinisothiocyanate (FITC)-conjugated goat antibodies to human (or mouse, for the standard) immunoglobulin.
  • FITC fluoresceinisothiocyanate
  • Binding Assays Cells from a human B cell line, T51, were used. Cells from human colon carcinoma line C3347 were used as a negative control, since they, according to previous testing, do not express detectable amounts of the 2H7 antigen.
  • the target cells were first incubated for 30 min at 4° C. with either the chimeric 2H7 or with mouse 2H7 standard, which had been purified from mouse ascites. This was followed by incubation with a second, FITC-labelled, reagent, which for the chimeric antibody was goat-anti-human immunoglobulin, obtained from TAGO (Burlingame, Calif.), and used at a dilution of 1:50.
  • TAGO Boundingame, Calif.
  • Antibody binding to the cell surface was determined using a Coulter Model EPIC-C cell sorter.
  • both the chimeric and the mouse standard 2H7 bound significantly, and to approximately the same extent, to the positive T51 line. They did not bind above background to the 2H7 negative C-3347 line.
  • ADCC antibody-dependent cellular cytotoxicity
  • Chimeric 2H7, but not mouse 2H7 antibody will be able to mediate both ADCC and CDC against human B lymphoma cells.
  • a hybridoma producing a non-functional mouse antibody can be converted to a hybridoma producing a chimeric antibody with ADCC and CDC activities.
  • Such a chimeric antibody is a prime candidate for the treatment or imaging of B-cell disorders, such as leukemias, lymphomas, and the like.
  • This invention therefore provides a method for making biologically functional antibodies when starting with a hybridoma which produces antibody which has the desired specificity for antigen but lacks biological effector functions such as ADCC and CDC.
  • the results presented above demonstrate that the chimeric 2H7 antibody binds to (Bp35(CD20)) antigen positive human B cells to approximately the same extent as the mouse 2H7 monoclonal antibody. This is significant because the 2H7 antibody defines a surface phosphoprotein antigen (Bp35(CD20)), of about 35,000 daltons, which is expressed on the cells of B cell lineage.
  • the 2H7 antibody does not bind detectably to various other cells such as fibroblasts, endothelial cells, or epithelial cells in the major organs or the stem cell precursors which give rise to B cells.
  • the chimeric 2H7 monoclonal antibodies will be advantageously used not only for therapy with unmodified chimeric antibodies, but also for development of various immunoconjugates with drugs, toxins, immunomodulators, isotopes, etc., as well as for diagnostic purposes such as in vivo imaging of B-cell tumors (for example, lymphomas and leukemias) using appropriately labelled chimeric 2H7 antibodies.
  • diagnostic purposes such as in vivo imaging of B-cell tumors (for example, lymphomas and leukemias) using appropriately labelled chimeric 2H7 antibodies.
  • Such immunoconjugation techniques are known to those skilled in the art and can be used to modify the chimeric 2H7 antibody molecules of the present invention.
  • the chimeric 2H7 antibody by virtue of its having the human constant portion, will possess biological activity in complement dependent and antibody dependent cytotoxicity which the mouse 2H7 does not.

Abstract

A chimeric antibody with human constant region and murine variable region, having specificity to a 35 kDA polypeptide (Bp35(CD20)) expressed on the surface of human B cells, methods of production, and uses.

Description

  • This application is a continuation in part of U.S. application Ser. No. ______, International Application No. PCT/US86/02269, filed Oct. 27, 1986, in the PCT receiving office of the U.S.
  • BACKGROUND OF THE INVENTION
  • 1. Field of the Invention
  • This invention relates to recombinant DNA methods of preparing an antibody with specificity for an antigen on the surface of human B cells, genetic sequences coding therefor, as well as methods of obtaining such sequences.
  • 2. Background Art
  • The application of cell-to-cell fusion for the production of monoclonal antibodies by Kohler and Milstein (Nature (London), 256: 495, 1975) spawned a revolution in biology equal in impact to that from the invention of recombinant DNA cloning. Monoclonal antibodies produced from hybridomas are already widely used in clinical and basic scientific studies. Applications of human monoclonal antibodies produced by human hybridomas hold great promise for the treatment of cancer, viral and microbial infections, certain immunodeficiencies with diminished antibody production, and other diseases and disorders of the immune system.
  • Unfortunately, a number of obstacles exist with respect to the development of human monoclonal antibodies. This is especially true when attempting to develop therapeutically useful monoclonal antibodies which define human cell surface antigens. Many of these human cell surface antigens are not recognized as foreign antigens by the human immune system; therefore, these antigens are not immunogenic in man. By contrast, human cellular antigens which are immunogenic in mice can be used for the production of mouse monoclonal antibodies that specifically recognize the human antigens. Although such antibodies may be used therapeutically in man, repeated injections of “foreign” antibodies, such as a mouse antibody, in humans, can lead to harmful hypersensitivity reactions as well as increased rate of clearance of the circulating antibody molecules so that the antibodies do not reach their target site. Furthermore, mouse monoclonal antibodies may lack the ability to efficiently interact with human effector cells as assessed by functional assays such as antibody-dependent cellular cytotoxicity (ADCC) and complement-mediated cytolysis (CDC).
  • Another problem faced by immunologists is that most human monoclonal antibodies obtained in cell culture are of the IgM type. When it is desirable to obtain human monoclonals of the IgG type, however, it has been necessary to use such techniques as cell sorting to identify and isolate the few cells which are producing antibodies of the IgG or other type from the majority producing antibodies of the IgM type. A need therefore exists for an efficient method of switching antibody classes, for any given antibody of a predetermined or desired antigenic specificity.
  • The present invention bridges both the hybridoma and genetic engineering technologies to provide a quick and efficient method, as well as products derived therefrom, for the production of a chimeric human/non-human antibody.
  • The chimeric antibodies of the present invention embody a combination of the advantageous characteristics of monoclonal antibodies derived from mouse-mouse hybridomas and of human monoclonal antibodies. The chimeric monoclonal antibodies, like mouse monoclonal antibodies, can recognize and bind to a human target antigen; however, unlike mouse monoclonal antibodies, the species-specific properties of the chimeric antibodies will avoid the induction of harmful hypersensitivity reactions and may allow for resistance to clearance when used in humans in vivo. Also, the inclusion of appropriate human immunoglobulin sequences can result in an antibody which efficiently interacts with human effector cells in vivo to cause tumor cell lysis and the like. Moreover, using the methods disclosed in the present invention, any desired antibody isotype can be conferred upon a particular antigen combining site.
  • Information Disclosure Statement*
  • Approaches to the problem of producing chimeric antibodies have been published by various authors.
  • Morrison, S. L. et al., Proc. Natl. Acad. Sci., USA, 81: 6851-6855 (November 1984), describe the production of a mouse-human antibody molecule of defined antigen binding specificity, produced by joining the variable region genes of a mouse antibody-producing myeloma cell line with known antigen binding specificity to human immunoglobulin constant region genes using recombinant DNA techniques. Chimeric genes were constructed, wherein the heavy chain variable region exon from the myeloma cell line S107 well joined to human IgG1 or IgG2 heavy chain constant region exons, and the light chain variable region exon from the same myeloma to the human kappa light chain exon. These genes were transfected into mouse myeloma cell lines and. Transformed cells producing chimeric mouse-human antiphosphocholine antibodies were thus developed.
  • Morrison, S. L. et al., European Patent Publication No. 173494 (published Mar. 5, 1986), disclose chimeric “receptors” (e.g. antibodies) having variable regions derived from one species and constant regions derived from another. Mention is made of utilizing cDNA cloning to construct the genes, although no details of cDNA cloning or priming are shown. (see pp 5, 7 and 8).
    * Note: The present Information Disclosure Statement is subject to the provisions of 37 C.F.R. 1.97(b). In addition, Applicants reserve the right to demonstrate that their invention was made prior to any one or more of the mentioned publications.
  • Boulianne, G. L. et al., Nature, 312: 643 (December 13, 1984), also produced antibodies consisting of mouse variable regions joined to human constant regions. They constructed immunoglobulin genes in which the DNA segments encoding mouse variable regions specific for the hapten trinitrophenyl (TNP) were joined to segments encoding human mu and kappa constant regions. These chimeric genes were expressed as functional TNP binding chimeric IgM. For a commentary on the work of Boulianne et al. and Morrison et al., see Munro, Nature, 312: 597 (Dec. 13, 1984), Dickson, Genetic Engineerinq News, 5, No. 3 (March 1985), or Marx, Science, 229: 455 (August 1985).
  • Neuberger, M. S. et al., Nature, 314: 268 (Mar. 25, 1985), also constructed a chimeric heavy chain immunoglobulin gene in which a DNA segment encoding a mouse variable region specific for the hapten 4-hydroxy-3-nitrophenacetyl (NP) was joined to a segment encoding the human epsilon region. When this chimeric gene was transfected into the J558L cell line, an antibody was produced which bound to the NP hapten and had human IgE properties.
  • Neuberger, M. S. et al., have also published work showing the preparation of cell lines that secrete hapten-specific antibodies in which the Fc portion has been replaced either with an active enzyme moiety (Williams, G. and Neuberger, M. S. Gene 43: 319, 1986) or with a polypeptide displaying c-myc antigenic determinants (Nature, 312: 604, 1984).
  • Neuberger, M. et al., PCT Publication WO 86/01533, (published Mar. 13, 1986) also disclose production of chimeric antibodies (see p. 5) and suggests, among the technique's many uses the concept of “class switching” (see p. 6).
  • Taniguchi, M., in European Patent Publication No. 171 496 (published Feb. 19, 1985) discloses the production of chimeric antibodies having variable regions with tumor specificty derived from experimental animals, and constant regions derived from human. The corresponding heavy and light chain genes are produced in the genomic form, and expressed in mammalian cells.
  • Takeda, S. et al., Nature, 314: 452 (Apr. 4, 1985) have described a potential method for the construction of chimeric immunoglobulin genes which have intron sequences removed by the use of a retrovirus vector. However, an unexpected splice donor site caused the deletion of the V region leader sequence. Thus, this approach did not yield complete chimeric antibody molecules.
  • Cabilly, S. et al., Proc. Natl. Acad. Sci., USA, 81: 3273-3277 (June 1984), describe plasmids that direct the synthesis in E. coli of heavy chains and/or light chains of anti-carcinoembryonic antigen (CEA) antibody. Another plasmid was constructed for expression of a truncated form of heavy chain (Fd′) fragment in E. coli. Functional CEA-binding activity was obtained by in vitro reconstitution, in E. coli extracts, of a portion of the heavy chain with light chain.
  • Cabilly, S., et al., European Patent Publication 125023 (published Nov. 14, 1984) describes chimeric immunoglobulin genes and their presumptive products as well as other modified forms. On pages 21, 28 and 33 it discusses cDNA cloning and priming.
  • Boss, M. A., European Patent Application 120694 (published Oct. 3, 1984) shows expression in E. coli of non-chimeric immunoglobulin chains with 4-nitrophenyl specificity. There is a broad description of chimeric antibodies but no details (see p. 9).
  • Wood, C. R. et al., Nature, 314: 446 (April, 1985) describe plasmids that direct the synthesis of mouse anti-NP antibody proteins in yeast. Heavy chain mu antibody proteins appeared to be glycosylated in the yeast cells. When both heavy and light chains were synthesized in the same cell, some of the protein was assembled into functional antibody molecules, as detected by anti-NP binding activity in soluble protein prepared from yeast cells.
  • Alexander, A. et al., Proc. Nat. Acad. Sci. USA, 79: 3260-3264 (1982), describe the preparation of a cDNA sequence coding for an abnormally short human Ig gamma heavy chain (OMM gamma3 HCD serum protein) containing a 19-amino acid leader followed by the first 15 residues of the V region. An extensive internal deletion removes the remainder of the V and the entire C H1 domain. This is cDNA coding for an internally deleted molecule.
  • Dolby, T. W. et al., Proc. Natl. Acad. Sci., USA, 77: 6027-6031 (1980), describe the preparation of a cDNA sequence and recombinant plasmids containing the same coding for mu and kappa human immunoglobulin polypeptides. One of the recombinant DNA molecules contained codons for part of the CH3 constant region domain and the entire 3′ noncoding sequence.
  • Seno, M. et al., Nucleic Acids Research, 11: 719-726 (1983), describe the preparation of a cDNA sequence and recombinant plasmids containing the same coding for part of the variable region and all of the constant region of the human IgE heavy chain (epsilon chain).
  • Kurokawa, T. et al., ibid, 11: 3077-3085 (1983), show the construction, using cDNA, of three expression plasmids coding for the constant portion of the human IgE heavy chain.
  • Liu, F. T. et al., Proc. Nat. Acad. Sci., USA, 81: 5369-5373 (September 1984), describe the preparation of a cDNA sequence and recombinant plasmids containing the same encoding about two-thirds of the CH2, and all of the CH3 and C H4 domains of human IgE heavy chain.
  • Tsujimoto, Y. et al., Nucleic Acids Res., 12: 8407-8414 (November 1984), describe the preparation of a human V lambda cDNA sequence from an Ig lambda-producing human Burkitt lymphoma cell line, by taking advantage of a cloned constant region gene as a primer for cDNA synthesis.
  • Murphy, J., PCT Publication WO 83/03971 (published Nov. 24, 1983) discloses hybrid proteins made of fragments comprising a toxin and a cell-specific ligand (which is suggested as possibly being an antibody).
  • Tan, et al., J. Immunol. 135: 8564 (November, 1985), obtained expression of a chimeric human-mouse immunoglobulin genomic gene after transfection into mouse myeloma cells.
  • Jones, P. T., et al., Nature 321: 552 (May 1986) constructed and expressed a genomic construct where CDR domains of variable regions from a mouse monolonal antibody were used to substitute for the corresponding domains in a human antibody.
  • Sun, L. K., et al., Hybridoma 5 suppl. 1 S17 (1986), describes a chimeric human/mouse antibody with potential tumor specificty. The chimeric heavy and light chain genes are genomic constructs and expressed in mammalian cells.
  • Sahagan et al., J. Immun. 137-1066-1074 (August 1986) describe a chimeric antibody with specificity to a human tumor associated antigen, the genes for which are assembled from genomic sequences.
  • For a recent review of the field see also Morrison, S. L., Science 229: 1202-1207 (Sep. 20, 1985) and Oi, V. T., et al., BioTechniques 4: 214 (1986).
  • The Oi, et al., paper is relevant as it argues that the production of chimeric antibodies from cDNA constructs in yeast and/or bacteria is not necessarily advantageous.
  • See also Commentary on page 835 in Biotechnology 4 (1986).
  • SUMMARY OF THE INVENTION
  • The invention provides a genetically engineered chimeric antibody of desired variable region specificity and constant region properties, through gene cloning and expression of light and heavy chains. The cloned immunoglobulin gene products can be produced by expression in genetically engineered cells.
  • The application of oligodeoxyribonucleotide synthesis, recombinant DNA cloning, and production of specific immunoglobulin chains in various prokaryotic and eukaryotic cells provides a means for the large scale production of a chimeric human/mouse monoclonal antibody with specificity to a human B cell surface antigen.
  • The invention provides cDNA sequences coding for immunoglobulin chains comprising a constant human region and a variable, non-human, region. The immunoglobulin chains can be either heavy or light.
  • The invention provides gene sequences coding for immunoglobulin chains comprising a cDNA variable region of the desired specificity. These can be combined with genomic constant regions of human origin.
  • The invention provides sequences as above, present in recombinant DNA molecules in vehicles such as plasmid vectors, capable of expression in desired prokaryotic or eukaryotic hosts.
  • The invention provides hosts capable of producing, by culture, the chimeric antibodies and methods of using these hosts.
  • The invention also provides individual chimeric immunoglobulin individual chains, as well as complete assembled molecules having human constant regions and variable regions with a human B cell surface antigen specificity, wherein both variable regions have the same binding specificity.
  • Among other immunoglobulin chains and/or molecules provided by the invention are:
      • (a) a complete functional, immunoglobulin molecule comprising:
        • (i) two identical chimeric heavy chains comprising a variable region with a human B cell surface antigen specificity and human constant region and
        • (ii) two identical all (i.e. non-chimeric) human light chains.
      • (b) a complete, functional, immunoglobulin molecule comprising:
        • (i) two identical chimeric heavy chains comprising a variable region as indicated, and a human constant region, and
        • (ii) two identical all (i.e. non-chimeric) non-human light chains.
      • (c) a monovalent antibody, i.e., a complete, functional immunoglobulin molecule comprising:
        • (i) two identical chimeric heavy chains comprising a variable region as indicated, and a human constant region, and
        • (ii) two different light chains, only one of which has the same specificity as the variable region of the heavy chains. The resulting antibody molecule binds only to one end thereof and is therefore incapable of divalent binding.
  • Genetic sequences, especially cDNA sequences, coding for the aforementioned combinations of chimeric chains or of non-chimeric chains are also provided herein.
  • The invention also provides for a genetic sequence, especially a cDNA sequence, coding for the variable region of desired specificity of an antibody molecule heavy and/or light chain, operably linked to a sequence coding for a polypeptide different than an immunoglobulin chain (e.g., an enzyme). These sequences can be assembled by the methods of the invention, and expressed to yield mixed-function molecules.
  • The use of cDNA sequences is particularly advantageous over genomic sequences (which contain introns), in that cDNA sequences can be expressed in bacteria or other hosts which lack appropriate RNA splicing systems.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 shows the DNA rearrangements and the expression of immunoglobulin mu and gamma heavy chain genes. This is a schematic representation of the human heavy chain gene complex (not shown to scale). Heavy chain variable V region formation occurs through the proper joining of VH, D and JH gene segments. This generates an active mu gene. A different kind of DNA rearrangement called “class switching” relocates the joined VH, D and JH region from the vicinity of mu constant C region to that of another heavy chain C region (switching to gamma is diagrammed here).
  • FIG. 2 shows the known nucleotide sequences of human and mouse J regions. Consensus sequences for the J regions are shown below the actual sequences. The oligonucleotide sequence below the mouse kappa J region consensus sequence is a Universal Immunoglobulin Gene (UIG) oligonucleotide. Note that there are only a few J regions with relatively conserved sequences, especially near the constant regions, in each immunoglobulin gene locus.
  • FIG. 3 shows the nucleotide sequences of the mouse J regions. Shown below are the oligonucleotide primers UIG-H and UIG-K. Note that each contains a restriction enzyme site. They can be used as primers for the synthesis of cDNA complementary to the variable region of mRNA, and can also be used to mutagenize, in vitro, cloned cDNA.
  • FIG. 4 Human Constant Domain Module. The human C gamma 1 clone, pGMH6, was isolated from the cell line GM2146. The sequence at its JH-C H1 junction is shown. Two restriction enzyme sites are useful as joints in recombining the C H1 gene with different VH genes. The ApaI site is 16 nucleotide residues into the C H1 coding domain of Human gamma 1; and is used in a previous construction of a mouse-human chimeric heavy-chain immunoglobulin. The BstEII site is in the JH region, and is used in the construction described in this application.
  • The human CK clone, pGML60, is a composite of two cDNA clones, one isolated from GM1500 (pK2-3), the other from GM2146 (pGML1). The JK-CK junction sequence shown comes from pK2-3. In vitro mutagenesis using the oligonucleotide, JKHindIII, was carried out to engineer a HindIII site 14 nucleotide residues 5′ of the J-C junction. This changes a human GTG codon into a CTT codon.
  • FIG. 5 shows the nucleotide sequence of the V region of the 2H7 VH cDNA clone pH2-11. The sequence was determined by the dideoxytermination method using M13 subclones of gene fragments. Open circles denote amino acid residues confirmed by peptide sequence. A sequence homologous to DSP.2 in the CDR3 region is underlined. The NcoI site at 5′ end was converted to a SalI site by using SalI linkers.
  • FIG. 6 shows the nucleotide sequence of the V region of the 2H7 VK cDNA clone pL2-12. The oligonucleotide primer used for site-directed mutagenesis is shown below the J K5 segment. Open circles denote amino acid residues confirmed by peptide sequence.
  • FIG. 7 shows the construction of the light and heavy chain expression plasmids pING2106 (panel a) and pING2101 (panel B). The SalI to BamHI fragment from pING2100 is identical to the SalI to BamHI fragment from pING2012E (see panel C). A linear representation of the circular plasmid pING2012E is shown in panel C. The 6.6 Kb SalI to BamHI fragment contains sequences from pSV2-neo, puc12, M8alphaRX12, and pL1. The HindIII site in pSV2-neo was destroyed before assembly of pING2012E by HindIII cleavage, fill-in, and religation.
  • FIG. 8 shows the structure of several chimeric 2H7-VH expression plasmids. pING2107 is a qpt version of the light chain plasmid, pING2106. The larger ones are two-gene plasmids. pHL2-11 and pHL2-26 contain both H and L genes, while pLL2-25 contains two L genes. They were constructed by joining an NdeI fragment containing either an H or L gene to partially digested (with NdeI) pING2106.
  • FIG. 9 shows a summary of the sequence alterations made in the construction of the 2H7 chimeric antibody expression plasmids. Residues underlined in the 5′ untranslated region are derived from the cloned mouse kappa and heavy-chain genes. Residues circled in the V/C boundary result from mutagenesis operations to engineer restriction enzyme sites in this region.
  • DESCRIPTION OF THE PREFERRED EMBODIMENTS Introduction
  • Generally, antibodies are composed of two light and two heavy chain molecules. Light and heavy chains are divided into domains of structural and functional homology. The variable domains of both the light (VL) and the heavy (VH) chains determine recognition and specificity. The constant region domains of light (CL) and heavy (CH) chains confer important biological properties such as antibody chain association, secretion, transplacental mobility, complement binding, and the like.
  • A complex series of events leads to immunoglobulin gene expression in the antibody producing cells. The V region gene sequences conferring antigen specificity and binding are located in separate germ line gene segments called VH, D and JH; or VL and JL. These gene segments are joined by DNA rearrangements to form the complete V regions expressed in heavy and light chains respectively (FIG. 1). The rearranged, joined (VL-JL and VH-D-JH)V segments then encode the complete variable regions or antigen binding domains of light and heavy chains, respectively.
  • Definitions
  • Certain terms and phrases are used throughout the specification and claims. The following definitions are provided for purposes of clarity and consistency.
  • 1. Expression vector—a plasmid DNA containing necessary regulatory signals for the synthesis of mRNA derived from any gene sequence, inserted into the vector.
  • 2. Module vector—a plasmid DNA containing a constant or variable region gene module.
  • 3. Expression plasmid—an expression vector that contains an inserted gene, such as a chimeric immunoglobulin gene.
  • 4. Gene cloning—synthesis of a gene, insertion into DNA vectors, identification by hybridization, sequence analysis and the like.
  • 5. Transfection—the transfer of DNA into mammalian cells.
  • Genetic Processes and Products
  • The invention provides a novel approach for the cloning and production of a human/mouse chimeric antibody with specificity to a human B cell surface antigen. The antigen is a polypeptide or comprises a polypeptide bound by the 2H7 monoclonal antibody described in Clark et al. Proc. Natl. Acad. Sci., U.S.A. 82: 1766-1770 (1985). This antigen is a phosphoprotein designated (Bp35(CD20)) and is only expressed on cells of the B cell lineage. Murine monoclonal antibodies to this antigen have been made before and are described in Clark et al., supra; see also Stashenko, P., et al., J. Immunol. 125: 1678-1685 (1980).
  • The method of production combines five elements:
      • (1) Isolation of messenger RNA (mRNA) from the mouse hybridoma line producing the monoclonal antibody, cloning and cDNA production therefrom;
      • (2) Preparation of Universal Immunoglobulin Gene (UIG) oligonucleotides, useful as primers and/or probes for cloning of the variable region gene segments in the light and heavy chain mRNA from the hybridoma cell line, and cDNA production therefrom;
      • (3) Preparation of constant region gene segment modules by cDNA preparation and cloning, or genomic gene preparation and cloning;
      • (4) Construction of complete heavy or light chain coding sequences by linkage of the cloned specific immunoglobulin variable region gene segments of part (2) above to cloned human constant region gene segment modules;
      • (5) Expression and production of light and heavy chains in selected hosts, including prokaryotic and eukaryotic cells, either in separate fermentations followed by assembly of antibody molecules in vitro, or through production of both chains in the same cell.
  • One common feature of all immunoglobulin light and heavy chain genes and the encoded messenger RNAs is the so-called J region (i.e. joining region, see FIG. 1). Heavy and light chain J regions have different sequences, but a high degree of sequence homology exists (greater than 80%) especially near the constant region, within the heavy JH regions or the kappa light chain J regions. This homology is exploited in this invention and consensus sequences of light and heavy chain J regions were used to design oligonucleotides (designated herein as UIGs) for use as primers or probes for cloning immunoglobulin light or heavy chain mRNAs or genes (FIG. 3). Depending on the sequence of a particular UIG, it may be capable of hybridizing to all immunoglobulin mRNAs or a specific one containing a particular J sequence. Another utility of a particular UIG probe may be hybridization to light chain or heavy chain mRNAs of a specific constant region, such as UIG-MJK which detects all mouse JK-containing sequences (FIG. 2).
  • UIG design can also include a sequence to introduce a restriction enzyme site into the cDNA copy of an immunoglobulin gene (see FIG. 3). The invention may, for example, utilize chemical gene synthesis to generate the UIG probes for the cloning and modification of V regions from immunoglobulin mRNA. On the other hand, oligonucleotides can be synthesized to recognize individually, the less conserved 5′-region of the J regions as a diagnostic aid in identifying the particular J region present in the immunoglobulin mRNA.
  • A multi-step procedure is utilized for generating complete V+C region cDNA clones from the hybridoma cell light and heavy chain mRNAs. First, the complementary strand of oligodT-primed cDNA is synthesized, and this double-stranded cDNA is cloned in appropriate cDNA cloning vectors such as pBR322 (Gubler and Hoffman, Gene, 25: 263 (1983)). Clones are screened by hybridization with UIG oligonucleotide probes. Positive heavy and light chain clones identified by this screening procedure are mapped and sequenced to select those containing V region and leader coding sequences. In vitro mutagenesis including, for example, the use of UIG oligonucleotides, is then used to engineer desired restriction enzyme cleavage sites. We used this approach for the chimeric 2H7 light chain.
  • An expedient method is to use synthetic UIG oligonucleotides as primers for the synthesis of cDNA. This method has the advantage of simultaneously introducing a desired restriction enzyme site, such as BstEII (FIG. 3) into a V region cDNA clone. We used this approach for the chimeric 2H7 heavy chain.
  • Second, cDNA constant region module vectors are prepared from human cells. These cDNA clones are modified, when necessary, by site-directed mutagenesis to place a restriction site at the analogous position in the human sequence or at another desired location near a boundary of the constant region. An alternative method utilizes genomic C region clones as the source for C region module vectors.
  • Third, cloned V region segments generated as above are excised and ligated to light or heavy chain C region module vectors. For example, one can clone the complete human kappa light chain C region and the complete human gamma1 C region. In addition, one can modify the human gamma1 region to introduce a termination codon and thereby obtain a gene sequence which encodes the heavy chain portion of an Fab molecule.
  • The coding sequences having operationally linked V and C regions are then transferred into appropriate expression vehicles for expression in appropriate hosts, prokaryotic or eukaryotic. Operationally linked means in-frame joining of coding sequences to derive a continuously translatable gene sequence without alterations or interruptions of the triplet reading frame.
  • One particular advantage of using cDNA genetic sequences in the present invention is the fact that they code continuously for immunoglobulin chains, either heavy or light. By “continuously” is meant that the sequences do not contain introns (i.e. are not genomic sequences, but rather, since derived from mRNA by reverse transcription, are sequences of contiguous exons). This characteristic of the cDNA sequences provided by the invention allows them to be expressible in prokaryotic hosts, such as bacteria, or in lower eukaryotic hosts, such as yeast.
  • Another advantage of using cDNA cloning method is the ease and simplicity of obtaining variable region gene modules.
  • The terms “constant” and “variable” are used functionally to denote those regions of the immunoglobulin chain, either heavy or light chain, which code for properties and features possessed by the variable and constant regions in natural non-chimeric antibodies. As noted, it is not necessary for the complete coding region for variable or constant regions to be present, as long as a functionally operating region is present and available.
  • Expression vehicles include plasmids or other vectors. Preferred among these are vehicles carrying a functionally complete human constant heavy or light chain sequence having appropriate restriction sites engineered so that any variable-heavy or light chain sequence with appropriate cohesive ends can be easily inserted thereinto. Human constant heavy or light chain sequence-containing vehicles are thus an important embodiment of the invention. These vehicles can be used as intermediates for the expression of any desired complete heavy or light chain in any appropriate host.
  • One preferred host is yeast. Yeast provides substantial advantages for the production of immunoglobulin light and heavy chains. Yeasts carry out post-translational peptide modifications including glycosylation. A number of recombinant DNA strategies now exist which utilize strong promoter sequences and high copy number plasmids which can be used for overt production of the desired proteins in yeast. Yeast recognizes leader sequences on cloned mammalian gene products and secretes peptides bearing leader sequences (i.e. prepeptides) (Hitzman, et al., 11th International Conference on Yeast, Genetics and Molecular Biology, Montpelier, France, Sep. 13-17, 1982).
  • Yeast gene expression systems can be routinely evaluated for the level of heavy and light chain production, protein stability, and secretion. Any of a series of yeast gene expression systems incorporating promoter and termination elements from the actively expressed genes coding for glycolytic enzymes produced in large quantities when yeasts are grown in mediums rich in glucose can be utilized. Known glycolytic genes can also provide very efficient transcription control signals. For example, the promoter and terminator signals of the iso-1-cytochrome C (CYC-1) gene can be utilized.
  • The following approach can be taken to develop and evaluate optimal expression plasmids for the expression of cloned immunoglobulin cDNAs in yeast.
      • (1) The cloned immunoglobulin DNA linking V and C regions is attached to different transcription promoters and terminator DNA fragments;
      • (2) The chimeric genes are placed on yeast plasmids (see, for example, Broach, J. R. in Methods in Enzymology—Vol. 101: 307 ed. Wu, R. et al., 1983));
      • (3) Additional genetic units such as a yeast leader peptide may be included on immunoglobulin DNA constructs to obtain antibody secretion.
      • (4) A portion of the sequence, frequently the first 6 to 20 codons of the gene sequence may be modified to represent preferred yeast codon usage.
      • (5) The chimeric genes are placed on plasmids used for integration into yeast chromosomes.
  • The following approaches can be taken to simultaneously express both light and heavy chain genes in yeast.
      • (1) The light and heavy chain genes are each attached to a yeast promoter and a terminator sequence and placed on the same plasmid. This plasmid can be designed for either autonomous replication in yeast or integration at specific sites in the yeast chromosome.
      • (2) The light and heavy chain genes are each attached to a yeast promoter and terminator sequence on separate plasmids containing different selectable markers. For example, the light chain gene can be placed on a plasmid containing the trp1 gene as a selectable marker, while the heavy chain gene can be placed on a plasmid containing ura3 as a selectable marker. The plasmids can be designed for either autonomous replication in yeast or integration at specific sites in yeast chromosomes. A yeast strain defective for both selectable markers is either simultaneously or sequentially transformed with the plasmid containing the light chain gene and with the plasmid containing the heavy chain gene.
      • (3) The light and heavy chain genes are each attached to a yeast promoter and terminator sequence on separate plasmids each containing different selectable markers as described in (2) above. A yeast mating type “a” strain defective in the selectable markers found on the light and heavy chain expression plasmids (trp1 and ura3 in the above example) is transformed with the plasmid containing the light chain gene by selection for one of the two selectable markers (trp1 in the above example). A yeast mating type “alpha” strain defective in the same selectable markers as the “a” strain (i.e. trp1 and ura3 as examples) is transformed with a plasmid containing the heavy chain gene by selection for the alternate selectable marker (i.e. ura3 in the above example). The “a” strain containing the light chain plasmid (phenotype: Trp+Ura in the above example) and the strain containing the heavy chain plasmid (phenotype: TrpUra+ in the above example) are mated and diploids are selected which are prototrophic for both of the above selectable markers (Trp+Ura+ in the above example).
  • Among bacterial hosts which may be utilized as transformation hosts, E. coli K12 strain 294 (ATCC 31446) is particularly useful. Other microbial strains which may be used include E. coli X1776 (ATCC 31537). The aforementioned strains, as well as E. coli W3110 (ATCC 27325) and other enterobacteria such as Salmonella typhimurium or Serratia marcescens, and various Pseudomonas species may be used.
  • In general, plasmid vectors containing replicon and control sequences which are derived from species compatible with a host cell are used in connection with these hosts. The vector ordinarily carries a replication site, as well as specific genes which are capable of providing phenotypic selection in transformed cells. For example, E. coli is readily transformed using pBR322, a plasmid derived from an E. coli species (Bolivar, et al., Gene, 2: 95 (1977)). pBR322 contains genes for ampicillin and tetracycline resistance, and thus provides easy means for identifying transformed cells. The pBR322 plasmid or other microbial plasmids must also contain, or be modified to contain, promoters which can be used by the microbial organism for expression of its own proteins. Those promoters most commonly used in recombinant DNA construction include the beta-lactamase (penicillinase) and lactose (beta-galactosidase) promoter systems (Chang et al., Nature, 275: 615 (1978); Itakura et al., Science, 198: 1056 (1977)); and tryptophan promoter systems (Goeddel et al., Nucleic Acids Research, 8: 4057 (1980); EPO Publication No. 0036776). While these are the most commonly used, other microbial promoters have been discovered and utilized.
  • For example, a genetic construct for any heavy or light chimeric immunoglobulin chain can be placed under the control of the leftward promoter of bacteriophage lambda (PL). This promoter is one of the strongest known promoters which can be controlled. Control is exerted by the lambda repressor, and adjacent restriction sites are known.
  • The expression of the immunoglobulin chain sequence can also be placed under control of other regulatory sequences which may be “homologous” to the organism in its untransformed state. For example, lactose dependent E. coli chromosomal DNA comprises a lactose or lac operon which mediates lactose digestion by elaborating the enzyme beta-galactosidase. The lac control elements may be obtained from bacteriophage lambda pLAC5, which is infective for E. coli. The lac promoter-operator system can be induced by IPTG.
  • Other promoter/operator systems or portions thereof can be employed as well. For example, arabinose, colicine E1, galactose, alkaline phosphatase, tryptophan, xylose, tac, and the like can be used.
  • Other preferred hosts are mammalian cells, grown in vitro in tissue culture, or in vivo in animals. Mammalian cells provide post-translational modifications to immunoglobulin protein molecules including leader peptide removal, correct folding and assembly of heavy and light chains, proper glycosylation at correct sites, and secretion of functional antibody protein.
  • Mammalian cells which may be useful as hosts for the production of antibody proteins include cells of lymphoid origin, such as the hybridoma Sp2/0-Ag14 (ATCC CRL 1581) or the myleoma P3X63Ag8 (ATCC TIB 9), and its derivatives. Others include cells of fibroblast origin, such as Vero (ATCC CRL 81) or CHO-K1 (ATCC CRL 61).
  • Several possible vector systems are available for the expression of cloned heavy chain and light chain genes in mammalian cells. One class of vectors relies upon the integration of the desired gene sequences into the host cell genome. Cells which have stably integrated DNA can be selected by simultaneously introducing drug resistance genes such as E. coli gpt (Mulligan, R. C. and Berg, P., Proc. Natl. Acad. Sci., USA, 78: 2072 (1981)) or Tn5 neo (Southern, P. J. and Berg, P., J. Mol. Appl. Genet., 1: 327 (1982)). The selectable marker gene can be either linked to the DNA gene sequences to be expressed, or introduced into the same cell by co-transfection (Wigler, M. et al., Cell, 16: 77 (1979)). A second class of vectors utilizes DNA elements which confer autonomously replicating capabilities to an extrachromosomal plasmid. These vectors can be derived from animal viruses, such as bovine papillomavirus (Sarver, N. et al., Proc. Natl. Acad. Sci., USA, 79: 7147 (1982)), polyoma virus (Deans, R. J. et al., Proc. Natl. Acad. Sci., USA, 81: 1292 (1984)), or SV40 virus (Lusky, M. and Botchan, M., Nature, 293: 79 (1981)).
  • Since an immunoglobulin cDNA is comprised only of sequences representing the mature mRNA encoding an antibody protein additional gene expression elements regulating transcription of the gene and processing of the RNA are required for the synthesis of immunoglobulin mRNA. These elements may include splice signals, transcription promoters, including inducible promoters enhancers, and termination signals. cDNA expression vectors incorporating such elements include those described by Okayama, H. and Berg, P., Mol. Cell Biol., 3: 280 (1983); Cepko, C. L. et al., Cell, 37: 1053 (1984); and Kaufman, R. J., Proc. Natl. Acad. Sci., USA, 82: 689 (1985).
  • An additional advantage of mammalian cells as hosts is their ability to express chimeric immunoglobulin genes which are derived from genomic sequences. Thus, mammalian cells may express chimeric immunoglobulin genes which are comprised of a variable region cDNA module plus a constant region which is composed in whole or in part of genomic sequences. Several human constant region genomic clones have been described (Ellison, J. W. et al., Nucl. Acids Res., 10: 4071 (1982), or Max, E. et al., Cell, 29: 691 (1982)). The use of such genomic sequences may be convenient for the simultaneous introduction of immunoglobulin enhancers, splice signals, and transcription termination signals along with the constant region gene segment.
  • Different approaches can be followed to obtain complete H2L2 antibodies.
  • First, one can separately express the light and heavy chains followed by in vitro assembly of purified light and heavy chains into complete H2L2 IgG antibodies. The assembly pathways used for generation of complete H2L2 IgG molecules in cells have been extensively studied (see, for example, Scharff, M., Harvey Lectures, 69: 125 (1974)). In vitro reaction parameters for the formation of IgG antibodies from reduced isolated light and heavy chains have been defined by Beychok, S., Cells of Immunoglobulin Synthesis, Academic Press, New York, page 69, 1979.
  • Second, it is possible to co-express light and heavy chains in the same cells to achieve intracellular association and linkage of heavy and light chains into complete H2L2 IgG antibodies. The co-expression can occur by using either the same or different plasmids in the same host.
  • Polypeptide Products
  • The invention provides “chimeric” immunoglobulin chains, either heavy or light. A chimeric chain contains a constant region substantially similar to that present in a natural human immunoglobulin, and a variable region having the desired antigenic specificity of the invention, i.e., to the specified human B cell surface antigen.
  • The invention also provides immunoglobulin molecules having heavy and light chains associated so that the overall molecule exhibits any desired binding and recognition properties. Various types of immunoglobulin molecules are provided: monovalent, divalent, molecules with chimeric heavy chains and non-chimeric light chains, or molecules with the invention's variable binding domains attached to moieties carrying desired functions.
  • Antibodies having chimeric heavy chains of the same or different variable region binding specificity and non-chimeric (i.e., all human or all non-human) light chains, can be prepared by appropriate association of the needed polypeptide chains. These chains are individually prepared by the modular assembly methods of the invention.
  • Uses
  • The antibodies of the invention having human constant region can be utilized for passive immunization, especially in humans, without negative immune reactions such as serum sickness or anaphylactic shock. The antibodies can, of course, also be utilized in prior art immunodiagnostic assays and kits in detectably labelled form (e.g., enzymes, 125I, 14C, fluorescent labels, etc.), or in immunobilized form (on polymeric tubes, beads, etc.), in labelled form for in vivo imaging, wherein the label can be a radioactive emitter, or an NMR contrasting agent such as a carbon-13 nucleus, or an X-ray contrasting agent, such as a heavy metal nucleus. The antibodies can also be used for in vitro localization of the antigen by appropriate labelling.
  • The antibodies can be used for therapeutic purposes, by themselves, in complement mediated lysis, or coupled to toxins or therapeutic moieties, such as ricin, etc.
  • Mixed antibody-enzyme molecules can be used for immunodiagnostic methods, such as ELISA. Mixed antibody-peptide effector conjugates can be used for targeted delivery of the effector moiety with a high degree of efficacy and specificity.
  • Specifically, the chimeric antibodies of this invention can be used for any and all uses in which the murine 2H7 monoclonal antibody can be used, with the obvious advantage that the chimeric ones are more compatible with the human body.
  • Having now generally described the invention, the same will be further understood by reference to certain specific examples which are included herein for purposes of illustration only and are not intended to be limiting unless otherwise specified.
  • Experimental
  • Materials and Methods
  • Tissue Culture Cell Lines
  • The human cell lines GM2146 and GM1500 were obtained from the Human Mutant Cell Repository (Camden, N.J.) and cultured in RPMI1640 plus 10% fetal bovine serum (M. A. Bioproducts). The cell line Sp2/0 was obtained from the American Type Culture Collection and grown in Dulbecco's Modified Eagle Medium (DMEM) plus 4.5 g/l glucose (M. A. Bioproducts) plus 10% fetal bovine serum (Hyclone, Sterile Systems, Logan, Utah). Media were supplemented with penicillin/streptomycin (Irvine Scientific, Irvine, California).
  • Recombinant Plasmid and Bacteriophage DNAs
  • The plasmids pBR322, pL1 and pUC12 were purchased from Pharmacia P-L Biochemicals (Milwaukee, Wisconsin). The plasmids pSV2-neo and pSV2-qpt were obtained from BRL (Gaithersburg, Md.), and are available from the American Type Culture Collection (Rockville, Md.). pHu-gamma-1 is a subclone of the 8.3 Kb HindIII to BamHI fragment of the human IgG1 chromosomal gene. An isolation method for of the human IgG1 chromosomal gene is described by Ellison, J. W. et al., Nucl. Acids Res., 10: 4071 (1982). M8alphaRX12 contains the 0.7 Kb XbaI to EcoRI fragment containing the mouse heavy chain enhancer from the J-C intron region of the M603 chromosomal gene (Davis, M. et al., Nature, 283: 733, 1979) inserted into M13mp10. DNA manipulations involving purification of plasmid DNA by buoyant density centrifugation, restriction endonuclease digestion, purification of DNA fragments by agarose gel electrophoresis, ligation and transformation of E. coli were as described by Maniatis, T. et al., Molecular Cloning: A Laboratory Manual, (1982) or other procedures. Restriction endonucleases and other DNA/RNA modifying enzymes were purchased from Boehringer-Mannheim (Indianapolis, Ind.), BRL, New England Biolabs (Beverly, Mass.) and Pharmacia P-L.
  • Oligonucleotide Preparation
  • Oligonucleotides were either synthesized by the triester method of Ito et al. (Nucl. Acids Res., 10: 1755 (1982)), or were purchased from ELESEN, Los Angeles, Calif. Tritylated, deblocked oligonucleotides were purified on Sephadex-G50, followed by reverse-phase HPLC with a 0-25% gradient of acetonitrile in 10 mM triethylamine-acetic acid, pH 7.2, on a C18 Bondapak column (Waters Associates). Detritylation was in 80% acetic acid for 30 min., followed by evaporation thrice. Oligonucleotides were labeled with [gamma-32P]ATP by T4 polynucleotide kinase.
  • RNA Preparation and Analysis
  • Total cellular RNA was prepared from tissue culture cells by the method of Auffray, C. and Rougeon, F. (Eur. J. Biochem., 107: 303 (1980)) or Chirgwin, J. M. et al. (Biochemistry, 18: 5294 (1979)). Preparation of poly(A)+ RNA, methyl-mercury agarose gel electrophoresis, and “Northern” transfer to nitrocellulose were as described by Maniatis, T. et al., supra. Total cellular RNA or poly(A)+ RNA was directly bound to nitrocellulose by first treating the RNA with formaldehyde (White, B. A. and Bancroft, F. C., J. Biol. Chem., 257: 8569 (1982)). Hybridization to filterbound RNA was with nick-translated DNA fragments using conditions described by Margulies, D. H. et al. (Nature, 295: 168 (1982)) or with 32P-labelled oligonucleotide using 4×SSC, 10× Denhardt's, 100 ug/ml salmon sperm DNA at 37° C. overnight, followed by washing in 4×SSC at 37° C.
  • cDNA Preparation and Cloning
  • Oligo-dT primed cDNA libraries were prepared from poly(A)+ RNA from GM1500 and GM2146 cells by the methods of Land, H. et al. (Nucl. Acids Res., 9: 2251 (1981)) and Gubler, V. and Hoffman, B. J., Gene, 25: 263 (1983), respectively. The cDNA libraries were screened by hybridization (Maniatis, T., supra) with 32P-labelled oligonucleotides using the procedure of de Lange et al. (Cell, 34: 891 (1983)), or with nick-translated DNA fragments.
  • Oligonucleotide Primer Extension and Cloning
  • Poly(A)+ RNA (20 ug) was mixed with 1.2 ug primer in 40 ul of 64 mM KCl. After denaturation at 90° C. for 5 min. and then chilling in ice, 3 units Human Placental Ribonuclease Inhibitor (BRL) was added in 3 ul of 1M Tris-HCl, pH 8.3. The oligonucleotide was annealed to the RNA at 42° C. for 15 minutes, then 12 ul of 0.05M DTT, 0.05M MgCl2, and 1 mM each of dATP, dTTP, dCTP, and dGTP was added. 2 ul of alpha-32P-dATP (400 Ci/mmol, New England Nuclear) was added, followed by 3 ul of AMV reverse transcriptase (19 units/ul, Life Sciences).
  • After incubation at 42° C. for 105 min., 2 ul 0.5 M EDTA and 50 ul 10 mM Tris, 1 mM EDTA, pH 7.6 were added. Unincorporated nucleotides were removed by Sephadex G-50 spin column chromatography, and the RNA-DNA hybrid was extracted with phenol, then with chloroform, and precipitated with ethanol. Second strand synthesis, homopolymer tailing with dGTP or dCTP, and insertion into homopolymer tailed vectors was as described by Gubler and Hoffman, supra.
  • Site-Directed Mutagenesis
  • Single stranded M13 subclone DNA (1 ug) was combined with 20 ng oligonucleotide primer in 12.5 ul of Hin buffer (7 mM Tris-HCl, pH 7.6, 7 mM MgCl2, 50 mM NaCl). After heating to 95° C. in a sealed tube, the primer was annealed to the template by slowly cooling from 70° C. to 37° C. for 90 minutes. 2 ul dNTPs (1 mM each), 1 ul 32P-DATP (10 uCi), 1 ul DTT (0.1 M) and 0.4 ul Klenow DNA PolI (2u, Boehringer Mannheim) were added and chains extended at 37° C. for 30 minutes. To this was added 1 ul (10 ng) M13 reverse primer (New England Biolabs), and the heating/annealing and chain extension steps were repeated. The reaction was stopped with 2 ul of 0.5M EDTA, pH 8, plus 80 ul of 10 mM Tris-HCl, pH 7.6, 1 mM EDTA. The products were phenol extracted and purified by Sephadex G-50 spun column chromatography and ethanol precipitated prior to restriction enzyme digestion and ligation to the appropriate vector.
  • Transfection of Myeloma Tissue Culture Cells
  • The electroporation method of Potter, H. et al. (Proc. Natl. Acad. Sci., USA, 81: 7161 (1984)) was used. After transfection, cells were allowed to recover in complete DMEM for 48-72 hours, then were seeded at 10,000 to 50,000 cells per well in 96-well culture plates in the presence of selective medium. G418 (GIBCO) selection was at 0.8 mg/ml, and mycophenolic acid (Calbiochem) was at 6 ug/ml plus 0.25 mg/ml xanthine.
  • Assays for Immunoglobulin Synthesis and Secretion
  • Secreted immunoglobulin was measured directly from tissue culture cell supernatants. Cytoplasmic protein extract was prepared by vortexing 106 cells in 160 ul of 1% NP40, 0.15 M NaCl, 10 mM Tris, 1 mM EDTA, pH 7.6 and leaving the lysate at 0° C., 15 minutes, followed by centrifugation at 10,600×g to remove insoluble debris.
  • A double antibody sandwich ELISA (Voller, A. et al., in Manual of Clinical Immunology, 2nd Ed., Eds. Rose, N. and Friedman, H., pp. 359-371, 1980) using affinity purified antisera was used to detect specific immunoglobulins. For detection of human IgG, the plate-bound antiserum is goat anti-human IgG (KPL, Gaithersburg, Md.) at 1/1000 dilution, while the peroxidase-bound antiserum is goat anti-human IgG (KPL or Tago, Burlingame) at 1/4000 dilution. For detection of human immunoglobulin kappa, the plate-bound antiserum is goat anti-human kappa (Tago) at 1/500 dilution, while the peroxidase-bound antiserum is goat anti-human kappa (Cappel) at 1/1000 dilution.
  • EXAMPLE 1 A Chimeric Mouse-Human Immunoglobulin with Specificity for a Human B-Cell Surface Antigen
  • (1) Antibody 2H7.
  • The 2H7 mouse monoclonal antibody (gamma 2b, kappa) recognizes a human B-cell surface antigen, (Bp35(CD20)) Clark, E. A., et al., Proc. Natl. Acad. Sci., U.S.A. 82: 1766 (1985)). The (Bp35(CD20)) molecules presumably play a role in B-cell activation. The antibody 2H7 does not react with stem cells which are progenitors of B-cells epithelial, mesenchymal and fibroblastic cells of other organs.
  • (2) Identification of J Sequences in the Immunoglobulin mRNA of 2H7.
  • Frozen cells were thawed on ice for 10 minutes and then at room temperature. The suspension was diluted with 15 ml PBS and the cells were centrifuged down. They were resuspended, after washes in PBS, in 16 ml 3M LiCl, 6M urea and disrupted in a polytron shear. The preparation of mRNA and the selection of the poly(A+) fraction were carried out according to Auffray, C. and Rougeon, F., Eur. J. Biochem. 107: 303, 1980.
  • The poly (A+) RNA from 2H7 was hybridized individually with labeled J H1, JH2, JH3 and J H4 oligonucleotides under conditions described by Nobrega et al. Anal. Biochem 131: 141, 1983). The products were then subjected to electrophoresis in a 1.7% agarose-TBE gel. The gel was fixed in 10% TCA, blotted dry and exposed for autoradiography. The result showed that the 2H7 VH contains J H1, JH2, or J H4 but not JH3 sequences.
  • For the analysis of the VK mRNA, the dot-blot method of White and Bancroft J. Biol. Chem. 257: 8569, (1982) was used. Poly (A+) RNA was immobilized on nitrocellulose filters and was hybridized to labeled probe-oligonucleotides at 400 in 4×SSC. These experiments show that 2H7 contains J K5 sequences.
  • (3) V Region cDNA Clones.
  • A library primed by oligo (dT) on 2H7 poly (A+) RNA was screened for kappa clones with a mouse CK region probe. From the 2H7 library, several clones were isolated. A second screen with a 5′ J K5 specific probe identified the 2H7 (JK5) light-chain clones. Heavy chain clones of 2H7 were generated by priming the poly(A+) RNA with the UIGH(BstEII) oligonucleotide (see FIG. 3), and identified by screening with the UIGH(BstEII) oligonucleotide.
  • The heavy and light chain genes or gene fragments from the VH and VK cDNA clones pH2-11 and pL2-12 were inserted into M13 bacteriophage vectors for nucleotide sequence analysis. The complete nucleotide sequences of the variable region of these clones were determined (FIGS. 5 and 6) by the dideoxy chain termination method. These sequences predict V region amino acid compositions that agree well with the observed compositions, and predict peptide sequences which have been verified by direct amino acid sequencing of portions of the V regions.
  • The nucleotide sequences of the cDNA clones show that they are immunoglobulin V region clones as they contain amino acid residues diagnostic of V domains (Kabat et al., Sequences of Proteins of Immunological Interest; U.S. Dept of HHS, 1983).
  • The 2H7 VH has the J H1 sequence. The 2H7 VL is from the VK-KpnI family (Nishi et al. Proc. Nat. Acd. Sci. USA 82: 6399, 1985), and uses J K5. The cloned 2H7 VL predicts an amino acid sequence which was confirmed by amino acid sequencing of peptides from the 2H7 light chain corresponding to residues 81-100. The cloned 2H7 VH predicts an amino acid sequence confirmed also by peptide sequencing, namely residues 1-12.
  • (4) In Vitro Mutagenesis to Engineer Restriction Enzyme Sites in the J Region for Joining to a Human C-Module, and to Remove Oligo (dC) Sequences 5′ to the V Modules.
  • For the 2H7 VK, the J-region mutagenesis primer JKHindIII, as shown in FIG. 6, was utilized. A human CK module derived from a cDNA clone was also mutagenized to contain the HindIII sequence (see FIG. 4). The mutagenesis reaction was performed on M13 subclones of these genes. The frequency of mutant clones ranged from 0.5 to 1% of the plaques obtained.
  • It had been previously observed that the oligo (dC) sequence upstream of the AUG codon in a VH chimeric gene interferes with proper splicing in one particular gene construct. It was estimated that perhaps as much as 70% of the RNA transcripts had undergone the mis-splicing, wherein a cryptic 3′ splice acceptor in the leader sequence was used. Therefore the oligo (dC) sequence upstream of the initiator AUG was removed in all of the clones.
  • In one approach, an oligonucleotide was used which contains a SalI restriction site to mutagenize the 2H7 VK clone. The primer used for this oligonucleotide-directed mutagenesis is a 22-mer which introduces a SalI site between the oligo (dC) and the initiator met codon (FIG. 6).
  • In a different approach, a convenient NcoI site was utilized to delete the 5′ untranslated region and oligo (dC) of the 2H7 VH clone (see FIG. 5).
  • The human C gamma 1 gene module is a cDNA derived from GM2146 cells (Human Genetic Mutant Cell Repository, Newark, N.J.). This C gamma 1 gene module was previously combined with a mouse VH gene module to form the chimeric expression plasmid pING2012E (FIG. 7C).
  • (5) Chimeric 2H7 Expression Plasmids.
  • A 2H7 chimeric heavy chain expression plasmid was derived from the replacement of the VH module of pING2012E with the VH cDNA modules to give the expression plasmid pING2101 (FIG. 7 b). This plasmid directs the synthesis of chimeric 2H7 heavy chain when transfected into mammalian cells.
  • For the 2H7 light chain chimeric gene, the SalI to HindIII fragment of the mouse VK module was joined to the human C K module by the procedure outlined in FIG. 7 a, forming pING2106. Replacement of the neo sequence with the E. coli gpt gene derived from pSV2-gpt resulted in pING2107, which expresses 2H7 chimeric light chain and confers mycophenolic acid resistance when transfected into mammalian cells.
  • The inclusion of both heavy and light chain chimeric genes in the same plasmid allows for the introduction into transfected cells of a 1:1 gene ratio of heavy and light chain genes leading to a balanced gene dosage. This may improve expression and decrease manipulations of transfected cells for optimal chimeric antibody expression. For this purpose, the DNA fragments derived from the chimeric heavy and light chain genes of pING2101 and pING2106 were combined into the expression plasmids pHL2-11 and pHL2-26 (FIG. 8). This expression plasmid contains a selectable neoR marker and separate transcription units for each chimeric gene, each including a mouse heavy chain enhancer.
  • The modifications and V-C joint regions of the 2H7 chimeric genes are summarized in FIG. 9.
  • (6) Stable Transfection of Mouse Lymphoid Cells for the Production of Chimeric Antibody.
  • Electroporation was used (Potter et al. supra; Toneguzzo et al. Mol. Cell Biol. 6: 703 1986) for the introduction of 2H7 chimeric expression plasmid DNA into mouse Sp2/0 cells. The electroporation technique gave a transfection frequency of 104×105 for the Sp2/0 cells.
  • The expression plasmids, pING2101 and pING2106, were digested with NdeI; and the DNA was introduced into Sp2/0 cells by electroporation. Transformant 1D6 was obtained which secretes chimeric 2H7 antibody. Antibody isolated from this cell line was used for the functional assays done to characterize the chimeric antibody. We have also obtained transformants from experiments using the two-gene plasmids.
  • (7) Purification of Chimeric 2H7 Antibody Secreted in Tissue Culture.
  • a. 1D6 (Sp2/0.pING2101/pING2106.1D6) cells were grown in culture medium [DMEM (Gibco #320-1965), supplemented with 10% Fetal Bovine Serum (Hyclone #A-1111-D), 10 mM HEPES, 1× Glutamine-Pen-Strep (Irvine Scientific #9316) to 1×106 cell/ml.
  • b. The cells were then centrifuged at 400×g and resuspended in serum-free culture medium at 2×106 cell/ml for 18-24 hr.
  • c. The medium was centrifuged at 4000 RPM in a JS-4.2 rotor (3000×g) for 15 min.
  • d. 1.6 liter of supernatant was then filtered through a 0.45 micron filter and then concentrated over a YM30 (Amicon Corp.) filter to 25 ml.
  • e. The conductance of the concentrated supernatant was adjusted to 5.7-5.6 mS/cm CDM 80 radiometer and the pH was adjusted to 8.0.
  • f. The supernatant was centrifuged at 2000×g, 5 min., and then loaded onto a 40 ml DEAE column, which was preequilibrated with 10 mM sodium phosphate, pH8.0.
  • g. The flow through fraction was collected and loaded onto a 1 ml protein A-Sepharose (Sigma) column preequilibrated with 10 mM sodium phosphate, pH8.0.
  • h. The column was washed first with 6 ml 10 mM sodium phosphate buffer pH 8.0, followed by 8 ml 0.1M sodium citrate pH 3.5, then by 6 ml 0.1M citric acid (pH 2.2). Fractions of 0.5 ml were collected in tubes containing 50 ul 2M Tris base (Sigma).
  • i. The bulk of the IgG was in the pH 3.5 elution and was pooled and concentrated over Centricon 30 (Amicon Corp.) to approximately 0.06 ml.
  • j. The buffer was changed to PBS (10 mM sodium phosphate pH 7.4, 0.15M NaCl) in Centricon 30 by repeated diluting with PBS and reconcentrating.
  • k. The IgG solution was then adjusted to 0.10 ml and bovine serum albumin (Fraction V, U.S. Biochemicals) was added to 1.0% as a stabilizing reagent.
  • (9) Chimeric 2H7 Antibody, Like the Mouse 2H7 Antibody, Specifically Binds to Human B Cells.
  • First, the samples were tested with a binding assay, in which cells of both an 2H7 antigen-positive and an 2H7 antigen-negative cell line were incubated with standard mouse monoclonal antibody 2H7 with chimeric 2H7 antibody derived from the cell culture supernatants, followed by a second reagent, fluoresceinisothiocyanate (FITC)-conjugated goat antibodies to human (or mouse, for the standard) immunoglobulin.
  • Binding Assays. Cells from a human B cell line, T51, were used. Cells from human colon carcinoma line C3347 were used as a negative control, since they, according to previous testing, do not express detectable amounts of the 2H7 antigen. The target cells were first incubated for 30 min at 4° C. with either the chimeric 2H7 or with mouse 2H7 standard, which had been purified from mouse ascites. This was followed by incubation with a second, FITC-labelled, reagent, which for the chimeric antibody was goat-anti-human immunoglobulin, obtained from TAGO (Burlingame, Calif.), and used at a dilution of 1:50. For the mouse standard, it was goat-anti-mouse immunoglobulin, also obtained from TAGO and used at a dilution of 1:50. Antibody binding to the cell surface was determined using a Coulter Model EPIC-C cell sorter.
  • As shown in Table I, both the chimeric and the mouse standard 2H7 bound significantly, and to approximately the same extent, to the positive T51 line. They did not bind above background to the 2H7 negative C-3347 line.
  • Functional Assays.
  • In previous studies, antibody 2H7 was tested for antibody-dependent cellular cytotoxicity (ADCC) measured by its ability to lyse Cr-labelled human B lymphona cells in the presence of human peripheral blood leukocytes as the source of effector cells. It was also tested for its ability to lyse 51Cr labelled hum B cells in the presence of human serum as the source of complement. These tests were carried out as previously described for mouse monoclonal anti-carcinoma antibody L6, which can mediate ADCC, as well as complement-mediated cytoxicity, CDC. The techniques used and the data described for the L6 antibody have been previously described. Hellstrom, et al., Proc. Natl. Acad. Sci. U.S.A. 83: 7059-7063 (1986).
  • Chimeric 2H7, but not mouse 2H7 antibody, will be able to mediate both ADCC and CDC against human B lymphoma cells. Thus a hybridoma producing a non-functional mouse antibody can be converted to a hybridoma producing a chimeric antibody with ADCC and CDC activities. Such a chimeric antibody is a prime candidate for the treatment or imaging of B-cell disorders, such as leukemias, lymphomas, and the like.
  • This invention therefore provides a method for making biologically functional antibodies when starting with a hybridoma which produces antibody which has the desired specificity for antigen but lacks biological effector functions such as ADCC and CDC.
  • Conclusions
  • The results presented above demonstrate that the chimeric 2H7 antibody binds to (Bp35(CD20)) antigen positive human B cells to approximately the same extent as the mouse 2H7 monoclonal antibody. This is significant because the 2H7 antibody defines a surface phosphoprotein antigen (Bp35(CD20)), of about 35,000 daltons, which is expressed on the cells of B cell lineage. The 2H7 antibody does not bind detectably to various other cells such as fibroblasts, endothelial cells, or epithelial cells in the major organs or the stem cell precursors which give rise to B cells.
  • Although the prospect of attempting tumor therapy using monoclonal antibodies is attractive, with some partial tumor regressions being reported, to date such monoclonal antibody therapy has been met with limited success (Houghton et al., February 1985, Proc. Natl. Acad. Sci. 82: 1242-1246). Murine monoclonal anti-(Bp35(CD20)) antibody has been used for therapy of B cell malignancies (Press, et al.,) Blood: February 1987, in press). The therapeutic efficacy of mouse monoclonal antibodies (which are the ones that have been tried so far) appears to be too low for most practical purposes. Because of the “human” properties which may make the chimeric 2H7 monoclonal antibodies more resistant to clearance and less immunogenic in vivo, the chimeric 2H7 monoclonal antibodies will be advantageously used not only for therapy with unmodified chimeric antibodies, but also for development of various immunoconjugates with drugs, toxins, immunomodulators, isotopes, etc., as well as for diagnostic purposes such as in vivo imaging of B-cell tumors (for example, lymphomas and leukemias) using appropriately labelled chimeric 2H7 antibodies. Such immunoconjugation techniques are known to those skilled in the art and can be used to modify the chimeric 2H7 antibody molecules of the present invention. The chimeric 2H7 antibody, by virtue of its having the human constant portion, will possess biological activity in complement dependent and antibody dependent cytotoxicity which the mouse 2H7 does not.
  • An illustrative cell line secreting chimeric 2H7 antibody was deposited prior to the U.S. filing date at the ATCC, Rockville Md. This is a transfected hybridoma (corresponds to 1D6 cells supra) ATCC HB 9303.
  • The present invention is not to be limited in scope by the cell lines deposited since the deposited embodiment is intended as a single illustration of one aspect of the invention and all cell lines which are functionally equivalent are within the scope of the invention. Indeed, various modifications of the invention in addition to those shown in the art from the foregoing description and accompanying drawings. Such modifications are intended to fall within the scope of the appended claims.
    TABLE 1
    Binding Assays Of Chimeric 2H7 Antibody and Mouse 2H7
    Monoclonal Antibody to a B cell Line Expressing (Bp35(CD20))
    and a Cell Line Not Expressing This Antigen.
    Antibody GAM GAH
    Binding
    Ratio* for
    T51 B-Cells
    2H7 Mouse 37 ND
    2H7 Chimeric ND 29
    L6 Mouse 1 ND
    Binding
    Ratio* for
    C3347 Cells
    2H7 Mouse 1.4 ND
    2H7 Chimeric ND 1.3
    L6 Mouse 110 ND

    *All assays were conducted using an antibody concentration of 10 ug/ml. The binding ratio is the number of times brighter a test sample is than a control sample treated with GAM (FITC-Conjugated goat anti-mouse) or GAH (FITC conjugated goat anti-human) alone. A ratio of 1 means that the test sample is just as bright as the control; a ratio of 2 means the test sample is twice as bright as the control and so on.

    ND—not done

Claims (29)

1. A polynucleotide molecule comprising a cDNA sequence coding for the variable region of an immunoglobulin chain having specificity to a 35-kDa polypeptide (Bp35(CD20)) expressed on the surface of B cells.
2. The molecule of claim 1 wherein said chain is a heavy chain.
3. The molecule of claim 1 wherein said chain is a light chain.
4. The molecule of claim 1 which further comprises an additional sequence coding for the constant C region of a human immunoglobulin chain, both said sequences in operable linkage with each other.
5. The molecule of claim 4 wherein said additional sequence is a cDNA sequence.
6. The molecule of claim 4 wherein said additional sequence is a genomic sequence.
7. The molecule of claim 1 which is a recombinant DNA molecule.
8. The molecule of claim 7 which is in double-stranded DNA form.
9. The molecule of claim 7 which is an expressible vehicle.
10. The molecule of claim 9 wherein said vehicle is a plasmid.
11. A prokaryotic host transformed with the molecule of claim 4.
12. The host of claim 11 which is a bacterium.
13. A eukaryotic host transfected with the molecule of claim 4.
14. The host of claim 13 which is yeast or a mammalian cell.
15. A heavy immunoglobulin chain comprising a constant human region and a variable region having specificity to a 35 kDa polypeptide (Bp35(CD20)) expressed on the surface of human B cells.
16. A light immunoglobulin chain comprising a constant human region and a variable region having specificity to a 35 kDa polypeptide (Bp35(CD20)) expressed on the surface of human B cells.
17. A chimeric antibody molecule comprising two light chains and two heavy chains, each of said chains comprising a constant human region and a variable region having specificity to a 35 kDa polypeptide (Bp35(CD20)) expressed on the surface of human B cells.
18. The antibody of claim 17 in detectably labelled form.
19. The antibody of claim 17 immobilized on an aqueous-insoluble solid phase.
20. A process of preparing an immunoglobulin heavy chain having a constant human region and a variable region having specificity to a 35 kDa polypeptide (Bp35(CD20)) expressed on the surface of human B cells which comprises:
culturing a host capable of expressing said chain under culturing conditions and
recovering from said culture said heavy chain.
21. A process of preparing an immunoglobulin light chain having a constant human region and a variable region with specificity to a 35 kDa polypeptide (Bp35(CD20)) expressed on the surface of human B cells which comprises:
culturing a host capable of expressing said chain under culturing conditions; and
recovering from said culture said light chain.
22. A process of preparing a chimeric immunoglobulin containing a heavy chain and a light chain, each of said heavy and light chains having a constant human region and a variable region with specificity to a 35 kDa polypeptide (Bp35(CD20)) expressed on the surface of human B cells which comprises:
culturing a host capable of expressing said heavy chain, or said light chain, or both, under culturing conditions; and
recovering from said culture said chimeric immunoglobulin molecule.
23. The process of any of claims 20, 21 or 22 wherein said host is prokaryotic.
24. The process of any of claims 20, 21 or 22 wherein said host is eukaryotic.
25. An immunoassay method for the detection of a 35 kDa polypeptide normally expressed on the surface of B cells in a sample, which comprises:
contacting said sample with the antibody of claim 17 and
detecting whether said antibody binds to said antigen.
26. An in vivo or in vitro imaging method to detect an antigen comprising a 35 kDa polypeptide normally expressed on the surface of ‘B ’ cells which comprises contacting said antigen with the labelled antibody of claim 18 and detecting said antibody.
27. A method of killing cells carrying an antigen thereon, which antigen comprising a 35 kDa polypeptide normally expressed on the surface of B cells which comprises:
contacting said cells with the antibody of claim 17.
28. The method of claim 27 wherein said killing occurs by complement mediated lysis of said cells.
29. The method of claim 27 wherein said killing occurs by ADCC.
US10/941,768 1986-10-27 2004-09-15 Chimeric antibody with specificity to human B cell surface antigen Abandoned US20050163708A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/941,768 US20050163708A1 (en) 1986-10-27 2004-09-15 Chimeric antibody with specificity to human B cell surface antigen

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
PCT/US1986/002269 WO1987002671A1 (en) 1985-11-01 1986-10-27 Modular assembly of antibody genes, antibodies prepared thereby and use
US1620287A 1987-01-08 1987-01-08
US19596188A 1988-05-13 1988-05-13
US07/665,939 US5500362A (en) 1987-01-08 1991-03-05 Chimeric antibody with specificity to human B cell surface antigen
US08/471,984 US5721108A (en) 1987-01-08 1995-06-06 Chimeric antibody with specificity to human B cell surface antigen
US09/021,934 US6120767A (en) 1986-10-27 1998-02-12 Chimeric antibody with specificity to human B cell surface antigen
US09/630,198 US6893625B1 (en) 1986-10-27 2000-08-01 Chimeric antibody with specificity to human B cell surface antigen
US10/941,768 US20050163708A1 (en) 1986-10-27 2004-09-15 Chimeric antibody with specificity to human B cell surface antigen

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/630,198 Continuation US6893625B1 (en) 1986-10-27 2000-08-01 Chimeric antibody with specificity to human B cell surface antigen

Publications (1)

Publication Number Publication Date
US20050163708A1 true US20050163708A1 (en) 2005-07-28

Family

ID=34577905

Family Applications (3)

Application Number Title Priority Date Filing Date
US09/630,198 Expired - Fee Related US6893625B1 (en) 1986-10-27 2000-08-01 Chimeric antibody with specificity to human B cell surface antigen
US09/724,138 Expired - Fee Related US6652852B1 (en) 1986-10-27 2000-11-28 Chimeric antibody with specificity to human B cell surface antigen
US10/941,768 Abandoned US20050163708A1 (en) 1986-10-27 2004-09-15 Chimeric antibody with specificity to human B cell surface antigen

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US09/630,198 Expired - Fee Related US6893625B1 (en) 1986-10-27 2000-08-01 Chimeric antibody with specificity to human B cell surface antigen
US09/724,138 Expired - Fee Related US6652852B1 (en) 1986-10-27 2000-11-28 Chimeric antibody with specificity to human B cell surface antigen

Country Status (1)

Country Link
US (3) US6893625B1 (en)

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020058029A1 (en) * 2000-09-18 2002-05-16 Nabil Hanna Combination therapy for treatment of autoimmune diseases using B cell depleting/immunoregulatory antibody combination
US20020197255A1 (en) * 1992-11-13 2002-12-26 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabelled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US20030147885A1 (en) * 1992-11-13 2003-08-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US20030206903A1 (en) * 1998-08-11 2003-11-06 Idec Pharmaceuticals Corporation Combination therapies for B-cell lynphomas comprising administration of anti-CD20 antibody
US20050123546A1 (en) * 2003-11-05 2005-06-09 Glycart Biotechnology Ag Antigen binding molecules with increased Fc receptor binding affinity and effector function
US20080206818A1 (en) * 2006-09-22 2008-08-28 Wacker Chemie Ag Process for the fermentative production of antibodies
US20090074760A1 (en) * 1998-11-09 2009-03-19 Grillo-Lopez Antonio J Use of chimeric anti-cd20 antibody as in vitro or in vivo purging agent in patients receiving bmt or pbsc transplant
US7682612B1 (en) 1998-11-09 2010-03-23 Biogen Idec Inc. Treatment of hematologic malignancies associated with circulating tumor cells using chimeric anti-CD20 antibody
US7820161B1 (en) 1999-05-07 2010-10-26 Biogen Idec, Inc. Treatment of autoimmune diseases
US20100303806A1 (en) * 2009-05-27 2010-12-02 Synageva Biopharma Corp. Avian derivedantibodies
US20110076273A1 (en) * 2009-09-11 2011-03-31 Genentech, Inc. Highly Concentrated Pharmaceutical Formulations
US20110201022A1 (en) * 2008-07-30 2011-08-18 Biomarin Pharmaceutical Inc. Assays for detection of phenylalanine ammonia-lyase and antibodies to phenylalanine ammonia-lyase
WO2011097335A3 (en) * 2010-02-04 2011-10-20 Biomarin Pharmaceutical Inc. Compositions of prokaryotic phenylalanine ammonia-lyase variants and methods of using compositions thereof
US8277651B2 (en) 2009-03-13 2012-10-02 Terrasep, Llc Methods and apparatus for centrifugal liquid chromatography
US8557244B1 (en) 1999-08-11 2013-10-15 Biogen Idec Inc. Treatment of aggressive non-Hodgkins lymphoma with anti-CD20 antibody
WO2014031648A3 (en) * 2012-08-21 2014-05-01 Ortho-Clinical Diagnostics, Inc Antibodies to risperidone and use thereof
WO2014031630A3 (en) * 2012-08-21 2014-05-08 Ortho-Clinical Diagnostics, Inc Antibodies to paliperidone and use thereof
US9410972B2 (en) 2012-08-21 2016-08-09 Janssen Pharmaceutica Nv Antibodies to quetiapine and use thereof
US9494608B2 (en) 2012-08-21 2016-11-15 Janssen Pharmaceutica Nv Antibodies to olanzapine and use thereof
US9494607B2 (en) 2012-08-21 2016-11-15 Janssen Pharmaceutica Nv Antibodies to aripiprazole and use thereof
US9611332B2 (en) 2012-08-21 2017-04-04 Janssen Pharmaceutica Nv Antibodies to aripiprazole haptens and use thereof
US9751953B2 (en) 2012-08-21 2017-09-05 Janssen Pharmaceutica Nv Antibodies to risperidone haptens and use thereof
US9850318B2 (en) 2012-08-21 2017-12-26 Janssen Pharmaceutica Nv Antibodies to quetiapine haptens and use thereof
US10370457B2 (en) 2012-08-21 2019-08-06 Janssen Pharmaceutica Nv Antibodies to paliperidone haptens and use thereof
US10435478B2 (en) 2015-12-17 2019-10-08 Janssen Pharmaceutica Nv Antibodies to quetiapine and use thereof
US10444250B2 (en) 2015-12-17 2019-10-15 Janssen Pharmaceutica Nv Antibodies to risperidone and use thereof
US10712353B2 (en) 2012-08-21 2020-07-14 Janssen Pharmaceutica Nv Antibodies to olanzapine haptens and use thereof

Families Citing this family (89)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5776093A (en) * 1985-07-05 1998-07-07 Immunomedics, Inc. Method for imaging and treating organs and tissues
US6893625B1 (en) 1986-10-27 2005-05-17 Royalty Pharma Finance Trust Chimeric antibody with specificity to human B cell surface antigen
US5736137A (en) * 1992-11-13 1998-04-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
EP1255558B1 (en) * 2000-02-16 2006-06-14 Genentech, Inc. Anti-april antibodies and hybridoma cells
US7754208B2 (en) * 2001-01-17 2010-07-13 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
US20030133939A1 (en) * 2001-01-17 2003-07-17 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
US7829084B2 (en) * 2001-01-17 2010-11-09 Trubion Pharmaceuticals, Inc. Binding constructs and methods for use thereof
EA007984B1 (en) * 2001-08-03 2007-02-27 Дженентек, Инк. TACIs AND BR3 POLYPEPTIDES AND USE THEREOF
SI2289936T1 (en) * 2002-12-16 2017-10-30 Genentech, Inc. Immunoglobulin variants and uses thereof
WO2004098634A2 (en) * 2003-04-30 2004-11-18 Government Of The United States Of America As Represented By The Sercretary Of The Department Of Health And Human Services National Institutes Of Health Protein arginine n-methyltransferase 2 (prmt-2)
CN1802388B (en) * 2003-05-09 2011-01-05 杜克大学 CD20-specific antibodies and methods of employing same
US20050163775A1 (en) * 2003-06-05 2005-07-28 Genentech, Inc. Combination therapy for B cell disorders
PL2272868T4 (en) 2003-06-05 2016-07-29 Genentech Inc Combination therapy for B cell disorders
US7902338B2 (en) 2003-07-31 2011-03-08 Immunomedics, Inc. Anti-CD19 antibodies
US20080166718A1 (en) * 2003-12-15 2008-07-10 Institut Pasteur Repertoire determination of a lymphocyte B population
BRPI0509412A (en) * 2004-04-16 2007-09-04 Genentech Inc polychondritis or multiplex mononeuritis treatment method in mammals and industrialized article
MXPA06012674A (en) * 2004-05-05 2007-03-26 Genentech Inc Preventing autoimmune disease.
AU2005249566B2 (en) * 2004-06-04 2010-11-11 Genentech, Inc. Method for treating multiple sclerosis
WO2005120437A2 (en) * 2004-06-04 2005-12-22 Genentech, Inc. Method for treating lupus
KR20070036187A (en) * 2004-07-22 2007-04-02 제넨테크, 인크. Method of treating sjogren's syndrome
KR100864549B1 (en) 2004-08-04 2008-10-20 어플라이드 몰리큘라 에볼류션, 인코포레이티드 Variant fc regions
WO2006023382A2 (en) * 2004-08-23 2006-03-02 Albert Einstein College Of Medicine Of Yeshiva University Collagen vi and cancer
WO2006041680A2 (en) * 2004-10-05 2006-04-20 Genentech, Inc. Method for treating vasculitis
JP2008526998A (en) * 2005-01-13 2008-07-24 ジェネンテック・インコーポレーテッド Method of treatment
DOP2006000029A (en) * 2005-02-07 2006-08-15 Genentech Inc ANTIBODY VARIANTS AND USES THEREOF. (VARIATIONS OF AN ANTIBODY AND USES OF THE SAME)
AR053579A1 (en) * 2005-04-15 2007-05-09 Genentech Inc TREATMENT OF INTESTINAL INFLAMMATORY DISEASE (IBD)
JP2008538767A (en) * 2005-04-22 2008-11-06 ジェネンテック・インコーポレーテッド Method for treating dementia or Alzheimer's disease with CD20 antibody
CN101223448B (en) * 2005-05-20 2012-01-18 健泰科生物技术公司 Pretreatment of a biological sample from an autoimmune disease subject
AU2006272555B2 (en) 2005-07-25 2012-03-08 Aptevo Research And Development Llc B-cell reduction using CD37-specific and CD20-specific binding molecules
MY149159A (en) 2005-11-15 2013-07-31 Hoffmann La Roche Method for treating joint damage
ES2618543T3 (en) 2005-11-23 2017-06-21 Genentech, Inc. Methods and compositions related to B lymphocyte assays
KR20080110833A (en) * 2006-03-21 2008-12-19 와이어쓰 Methods for preventing and treating amyloidogenic diseases
BRPI0712223A2 (en) 2006-06-02 2012-07-10 Aveo Pharmaceuticals Inc hepatocyte growth factor binding proteins (hgf)
DK2027156T3 (en) 2006-06-02 2011-04-26 Aveo Pharmaceuticals Inc Hepatocyte growth factor (HGF) binding protein
WO2007146172A2 (en) 2006-06-07 2007-12-21 Bioalliance C.V. Antibodies recognizing a carbohydrate containing epitope on cd-43 and cea expressed on cancer cells and methods using same
JP2009539413A (en) 2006-06-12 2009-11-19 トゥルビオン・ファーマシューティカルズ・インコーポレーテッド Single-chain multivalent binding protein with effector function
SG173371A1 (en) 2006-07-13 2011-08-29 Univ Iowa Res Found Methods and reagents for treatment and diagnosis of vascular disorders and age-related macular degeneration
US7879984B2 (en) * 2007-07-31 2011-02-01 Regeneron Pharmaceuticals, Inc. Human antibodies to human CD20 and method of using thereof
CN101868246A (en) 2007-09-21 2010-10-20 加利福尼亚大学董事会 Targeted interferon demonstrates potent apoptotic and anti-tumor activities
NZ585959A (en) 2007-12-18 2012-09-28 Bioalliance Cv Antibodies recognizing a carbohydrate containing epitope on cd-43 and cea expressed on cancer cells and methods using same
US7914785B2 (en) 2008-01-02 2011-03-29 Bergen Teknologieverforing As B-cell depleting agents, like anti-CD20 antibodies or fragments thereof for the treatment of chronic fatigue syndrome
EP2077281A1 (en) 2008-01-02 2009-07-08 Bergen Teknologioverforing AS Anti-CD20 antibodies or fragments thereof for the treatment of chronic fatigue syndrome
CN102099377A (en) * 2008-04-11 2011-06-15 新兴产品开发西雅图有限公司 CD37 immunotherapeutic and combination with bifunctional chemotherapeutic thereof
AR073295A1 (en) 2008-09-16 2010-10-28 Genentech Inc METHODS TO TREAT PROGRESSIVE MULTIPLE SCLEROSIS. MANUFACTURING ARTICLE.
WO2010075249A2 (en) 2008-12-22 2010-07-01 Genentech, Inc. A method for treating rheumatoid arthritis with b-cell antagonists
US20110142836A1 (en) * 2009-01-02 2011-06-16 Olav Mella B-cell depleting agents for the treatment of chronic fatigue syndrome
HUE060624T2 (en) 2009-02-13 2023-04-28 Immunomedics Inc Immunoconjugates with an intracellularly-cleavable linkage
US8287865B2 (en) 2009-09-16 2012-10-16 Immunomedics, Inc. Class I anti-CEA antibodies and uses thereof
CA2782194C (en) 2009-12-02 2018-01-16 Immunomedics, Inc. Combination of radiolabelled antibodies (rait) and antibody-drug conjugates (adc) for treatment of pancreatic cancer
US20110165161A1 (en) * 2009-12-23 2011-07-07 Shih-Yao Lin Anti-epcam antibodies that induce apoptosis of cancer cells and methods using same
AR080154A1 (en) 2010-02-10 2012-03-14 Immunogen Inc CD20 ANTIBODIES AND ITS USE
KR101764453B1 (en) 2010-04-09 2017-08-02 아베오 파마슈티컬즈, 인크. Anti-erbb3 antibodies
WO2011156617A2 (en) 2010-06-09 2011-12-15 Aveo Pharmaceuticals, Inc. Anti-egfr antibodies
WO2012003472A1 (en) 2010-07-02 2012-01-05 Aveo Pharmaceuticals, Inc. Anti-notch1 antibodies
AR082194A1 (en) 2010-07-06 2012-11-21 Aveo Pharmaceuticals Inc ANTI-RON ANTIBODIES
JP6024025B2 (en) 2011-05-02 2016-11-09 イミューノメディクス、インコーポレイテッドImmunomedics, Inc. Ultrafiltration concentration of allotype-selected antibodies for small volume administration
CA2853637C (en) 2011-10-26 2023-04-04 Novartis Ag Anti-canine cd20 monoclonal antibodies and methods of use
CA2874864C (en) 2012-08-14 2023-02-21 Ibc Pharmaceuticals, Inc. T-cell redirecting bispecific antibodies for treatment of disease
JOP20200236A1 (en) 2012-09-21 2017-06-16 Regeneron Pharma Anti-cd3 antibodies, bispecific antigen-binding molecules that bind cd3 and cd20, and uses thereof
US9803021B2 (en) 2012-12-07 2017-10-31 The Regents Of The University Of California CD138-targeted interferon demonstrates potent apoptotic and anti-tumor activities
WO2017004144A1 (en) 2015-07-01 2017-01-05 Immunomedics, Inc. Antibody-sn-38 immunoconjugates with a cl2a linker
ES2909746T3 (en) 2012-12-13 2022-05-10 Immunomedics Inc SN-38 and Antibody Immunoconjugate Dosages for Improved Efficacy and Decreased Toxicity
PL2935330T3 (en) 2012-12-19 2019-11-29 Aveo Pharmaceuticals Inc Anti-notch3 antibodies
EP2934584B1 (en) 2012-12-21 2020-02-19 Aveo Pharmaceuticals, Inc. Anti-gdf15 antibodies
WO2014194100A1 (en) 2013-05-29 2014-12-04 The Regents Of The University Of California Anti-cspg4 fusions with interferon for the treatment of malignancy
US9321834B2 (en) 2013-12-05 2016-04-26 Leidos, Inc. Anti-malarial compositions
AU2015219495B2 (en) 2014-02-21 2019-11-21 Ibc Pharmaceuticals, Inc. Disease therapy by inducing immune response to Trop-2 expressing cells
EP3110445A4 (en) 2014-02-25 2017-09-27 Immunomedics, Inc. Humanized rfb4 anti-cd22 antibody
TWI754319B (en) 2014-03-19 2022-02-01 美商再生元醫藥公司 Methods and antibody compositions for tumor treatment
EP3157952B1 (en) 2014-06-20 2021-12-22 Aveo Pharmaceuticals, Inc. Treatment of congestive heart failure and other cardiac dysfunction using a gdf15 modulator
EP3157953B1 (en) 2014-06-20 2021-12-22 Aveo Pharmaceuticals, Inc. Treatment of chronic kidney disease and other renal dysfunction using a gdf15 modulator
CA2953567C (en) 2014-06-24 2023-09-05 Immunomedics, Inc. Anti-histone therapy for vascular necrosis in severe glomerulonephritis
PT3197493T (en) 2014-09-25 2021-06-04 Aveo Pharmaceuticals Inc Methods of reversing cachexia and prolonging survival comprising administering a gdf15 modulator and an anti-cancer agent
CA2961774C (en) 2014-10-07 2023-05-23 Immunomedics, Inc. Neoadjuvant use of antibody-drug conjugates
ES2809455T3 (en) 2014-11-17 2021-03-04 Regeneron Pharma Methods for tumor treatment using CD3xCD20 bispecific antibody
CN107428837A (en) 2015-04-22 2017-12-01 免疫医疗公司 Circulate separation, detection, diagnosis and/or the identification of the positive cancer cells of TROP 2
EP3303373B1 (en) 2015-05-30 2020-04-08 Molecular Templates, Inc. De-immunized, shiga toxin a subunit scaffolds and cell-targeting molecules comprising the same
RS62986B1 (en) 2015-06-24 2022-03-31 Hoffmann La Roche Anti-transferrin receptor antibodies with tailored affinity
EP4257134A3 (en) 2015-06-25 2024-01-24 Immunomedics, Inc. Combining anti-hla-dr or anti-trop-2 antibodies with microtubule inhibitors, parp inhibitors, bruton kinase inhibitors or phosphoinositide 3-kinase inhibitors significantly improves therapeutic outcome in cancer
UA126278C2 (en) 2015-09-21 2022-09-14 Аптево Рісьорч Енд Девелопмент Ллс Cd3 binding polypeptides
CN114057884A (en) 2015-10-02 2022-02-18 豪夫迈·罗氏有限公司 Bispecific anti-human CD 20/human transferrin receptor antibodies and methods of use
AR106189A1 (en) 2015-10-02 2017-12-20 Hoffmann La Roche BIESPECTIFIC ANTIBODIES AGAINST HUMAN A-b AND THE HUMAN TRANSFERRINE RECEIVER AND METHODS OF USE
AU2017206074B2 (en) 2016-01-08 2023-09-07 Altrubio Inc. Tetravalent anti-PSGL-1 antibodies and uses thereof
US10314910B2 (en) 2016-01-10 2019-06-11 Neotx Therapeutics Ltd. Methods and compositions for enhancing the potency of superantigen mediated cancer immunotherapy
AU2019331024A1 (en) 2018-08-31 2021-03-18 Regeneron Pharmaceuticals, Inc. Dosing strategy that mitigates cytokine release syndrome for CD3/C20 bispecific antibodies
KR20220009428A (en) 2019-05-15 2022-01-24 네오티엑스 테라퓨틱스 엘티디. cancer treatment
WO2021155295A1 (en) 2020-01-31 2021-08-05 The Cleveland Clinic Foundation Anti-müllerian hormone receptor 2 antibodies and methods of use
JPWO2022239720A1 (en) 2021-05-10 2022-11-17
CA3229448A1 (en) 2021-08-23 2023-03-02 Immunitas Therapeutics, Inc. Anti-cd161 antibodies and uses thereof

Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4468464A (en) * 1974-11-04 1984-08-28 The Board Of Trustees Of The Leland Stanford Junior University Biologically functional molecular chimeras
US4486538A (en) * 1978-07-07 1984-12-04 Samuel Bogoch Detection of malignant tumor cells
US4650756A (en) * 1981-08-31 1987-03-17 Sloan Kettering Institute For Cancer Research Monoclonal antibodies to cell surface antigens of human renal cancer
US4708862A (en) * 1983-02-22 1987-11-24 Xoma Corporation Radioimmuno detection of human cancers using anti-tumor monoclonal antibody
US4724213A (en) * 1985-05-24 1988-02-09 Northwestern University Murine hybridoma Lym-1 and diagnostic antibody produced thereby
US4724212A (en) * 1985-05-24 1988-02-09 Northwestern University Murine hybridoma Lym-2 and diagnostic antibody produced thereby
US4863726A (en) * 1987-05-29 1989-09-05 Cetus Corporation Combination therapy using immunotoxins with interleukin-2
US5091178A (en) * 1986-02-21 1992-02-25 Oncogen Tumor therapy with biologically active anti-tumor antibodies
US5354847A (en) * 1986-10-27 1994-10-11 Bristol-Myers Squibb Company Chimeric antibody with specificity to human tumor antigen
US5500362A (en) * 1987-01-08 1996-03-19 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
US5500302A (en) * 1989-04-26 1996-03-19 Flex Products, Inc. Barrier film and method
US5545405A (en) * 1990-10-17 1996-08-13 Burroughs Wellcome Co. Method for treating a mammal suffering from cancer with a cho-glycosylated antibody
US5595721A (en) * 1993-09-16 1997-01-21 Coulter Pharmaceutical, Inc. Radioimmunotherapy of lymphoma using anti-CD20
US5807715A (en) * 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
US6652852B1 (en) * 1986-10-27 2003-11-25 Royalty Pharma Finance Trust Chimeric antibody with specificity to human B cell surface antigen

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES522315A0 (en) 1982-05-12 1984-08-16 Harvard College A PROCEDURE FOR OBTAINING A HYBRID PROTEIN.
GB8308235D0 (en) 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
JPS6147500A (en) 1984-08-15 1986-03-07 Res Dev Corp Of Japan Chimera monoclonal antibody and its preparation
EP0173494A3 (en) 1984-08-27 1987-11-25 The Board Of Trustees Of The Leland Stanford Junior University Chimeric receptors by dna splicing and expression
GB8422238D0 (en) 1984-09-03 1984-10-10 Neuberger M S Chimeric proteins
JPS61134325A (en) 1984-12-04 1986-06-21 Teijin Ltd Expression of hybrid antibody gene
US4770993A (en) 1985-10-31 1988-09-13 Beatrice Companies, Inc. Hybridoma tumor cell lines and their monoclonal antibodies to thaumatin
EP0247091B1 (en) 1985-11-01 1993-09-29 Xoma Corporation Modular assembly of antibody genes, antibodies prepared thereby and use

Patent Citations (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4468464A (en) * 1974-11-04 1984-08-28 The Board Of Trustees Of The Leland Stanford Junior University Biologically functional molecular chimeras
US4486538A (en) * 1978-07-07 1984-12-04 Samuel Bogoch Detection of malignant tumor cells
US4650756A (en) * 1981-08-31 1987-03-17 Sloan Kettering Institute For Cancer Research Monoclonal antibodies to cell surface antigens of human renal cancer
US4708862A (en) * 1983-02-22 1987-11-24 Xoma Corporation Radioimmuno detection of human cancers using anti-tumor monoclonal antibody
US5807715A (en) * 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
US4724213A (en) * 1985-05-24 1988-02-09 Northwestern University Murine hybridoma Lym-1 and diagnostic antibody produced thereby
US4724212A (en) * 1985-05-24 1988-02-09 Northwestern University Murine hybridoma Lym-2 and diagnostic antibody produced thereby
US5091178A (en) * 1986-02-21 1992-02-25 Oncogen Tumor therapy with biologically active anti-tumor antibodies
US5354847A (en) * 1986-10-27 1994-10-11 Bristol-Myers Squibb Company Chimeric antibody with specificity to human tumor antigen
US6893625B1 (en) * 1986-10-27 2005-05-17 Royalty Pharma Finance Trust Chimeric antibody with specificity to human B cell surface antigen
US6652852B1 (en) * 1986-10-27 2003-11-25 Royalty Pharma Finance Trust Chimeric antibody with specificity to human B cell surface antigen
US6120767A (en) * 1986-10-27 2000-09-19 Pharmaceutical Royalties, L.L.C. Chimeric antibody with specificity to human B cell surface antigen
US5500362A (en) * 1987-01-08 1996-03-19 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
US5677180A (en) * 1987-01-08 1997-10-14 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
US5721108A (en) * 1987-01-08 1998-02-24 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
US4863726A (en) * 1987-05-29 1989-09-05 Cetus Corporation Combination therapy using immunotoxins with interleukin-2
US5500302A (en) * 1989-04-26 1996-03-19 Flex Products, Inc. Barrier film and method
US5545403A (en) * 1990-10-17 1996-08-13 Burroughs Wellcome Co. Method for treating a mammal by administering a CHO-glycosylated antibody
US5545404A (en) * 1990-10-17 1996-08-13 Burroughs Wellcome Co. Method for treating a mammal suffering from a T-cell medicated disorder with a CHO-Glycosylated antibody
US5545405A (en) * 1990-10-17 1996-08-13 Burroughs Wellcome Co. Method for treating a mammal suffering from cancer with a cho-glycosylated antibody
US5843398A (en) * 1993-09-16 1998-12-01 Coulter Pharmaceutical, Inc. Radioimmunotherapy of lymphoma using anti-CD20 antibodies
US6015542A (en) * 1993-09-16 2000-01-18 Coulter Pharmaceutical, Inc. Radioimmunotherapy of lymphoma using anti-CD20 antibodies
US6090365A (en) * 1993-09-16 2000-07-18 Coulter Pharmaceutical, Inc. Radioimmunotherapy of lymphoma using anti-CD20 antibodies
US5595721A (en) * 1993-09-16 1997-01-21 Coulter Pharmaceutical, Inc. Radioimmunotherapy of lymphoma using anti-CD20

Cited By (81)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7744877B2 (en) 1992-11-13 2010-06-29 Biogen Idec Inc. Expression and use of anti-CD20 Antibodies
US20020197255A1 (en) * 1992-11-13 2002-12-26 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabelled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US20030147885A1 (en) * 1992-11-13 2003-08-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US20080089893A9 (en) * 1992-11-13 2008-04-17 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabelled antibodies to human b lymphocyte restricted differentiation antigen for treatment of b cell lymphoma
US7381560B2 (en) 1992-11-13 2008-06-03 Biogen Idec Inc. Expression and use of anti-CD20 antibodies
US7422739B2 (en) 1992-11-13 2008-09-09 Biogen Idec Inc. Anti-CD20 antibodies
US20030206903A1 (en) * 1998-08-11 2003-11-06 Idec Pharmaceuticals Corporation Combination therapies for B-cell lynphomas comprising administration of anti-CD20 antibody
US9296821B2 (en) 1998-08-11 2016-03-29 Biogen Inc. Combination therapies for B-cell lymphomas comprising administration of anti-CD20 antibodies
US20080038261A1 (en) * 1998-08-11 2008-02-14 Biogen Idec Inc. Combination therapies for b-cell lymphomas comprising administration of anti-cd20 antibody
US10113000B2 (en) 1998-08-11 2018-10-30 Biogen Inc. Combination therapies for B-cell lymphomas comprising administration of anti-CD20 antibody
US8329172B2 (en) 1998-08-11 2012-12-11 Biogen Idec Combination therapies for B-cell lymphomas comprising administration of anti-CD20 antibody
US20110165159A1 (en) * 1998-11-09 2011-07-07 Biogen Idec Inc. Use of chimeric anti-cd20 antibody as in vitro or in vivo purging agent in patients receiving bmt or pbsc transplant
US20100080769A1 (en) * 1998-11-09 2010-04-01 Biogen Idec Inc. Treatment of Chronic Lymphocytic Leukemia using Anti-CD20 Antibodies
US7682612B1 (en) 1998-11-09 2010-03-23 Biogen Idec Inc. Treatment of hematologic malignancies associated with circulating tumor cells using chimeric anti-CD20 antibody
US8206711B2 (en) 1998-11-09 2012-06-26 Biogen Idec Inc. Treatment of chronic lymphocytic leukemia using anti-CD20 antibodies
US20090074760A1 (en) * 1998-11-09 2009-03-19 Grillo-Lopez Antonio J Use of chimeric anti-cd20 antibody as in vitro or in vivo purging agent in patients receiving bmt or pbsc transplant
US20110008250A1 (en) * 1999-05-07 2011-01-13 Genentech, Inc. Treatment of Autoimmune Diseases
US20110008338A1 (en) * 1999-05-07 2011-01-13 Genentech, Inc. Treatment of Autoimmune Diseases
US20110008337A1 (en) * 1999-05-07 2011-01-13 Genetech, Inc. Treatment of Autoimmune Diseases
US20110008336A1 (en) * 1999-05-07 2011-01-13 Genentech, Inc. Treatment of Autoimmune Diseases
US9993550B2 (en) 1999-05-07 2018-06-12 Genentech, Inc. Treatment of pemphigus
US7820161B1 (en) 1999-05-07 2010-10-26 Biogen Idec, Inc. Treatment of autoimmune diseases
US8545843B2 (en) 1999-05-07 2013-10-01 Genentech, Inc. Treatment of vasculitis
US9504744B2 (en) 1999-08-11 2016-11-29 Biogen Inc. Treatment of diffuse large-cell lymphoma with anti-CD20 antibody
US10400043B2 (en) 1999-08-11 2019-09-03 Biogen, Inc. Treatment of diffuse large-cell lymphoma with anti-CD20 antibody
US8821873B2 (en) 1999-08-11 2014-09-02 Biogen Idec Inc. Treatment of diffuse large-cell lymphoma with anti-CD20 antibody
US8557244B1 (en) 1999-08-11 2013-10-15 Biogen Idec Inc. Treatment of aggressive non-Hodgkins lymphoma with anti-CD20 antibody
US20020058029A1 (en) * 2000-09-18 2002-05-16 Nabil Hanna Combination therapy for treatment of autoimmune diseases using B cell depleting/immunoregulatory antibody combination
US9296820B2 (en) 2003-11-05 2016-03-29 Roche Glycart Ag Polynucleotides encoding anti-CD20 antigen binding molecules with increased Fc receptor binding affinity and effector function
US20050123546A1 (en) * 2003-11-05 2005-06-09 Glycart Biotechnology Ag Antigen binding molecules with increased Fc receptor binding affinity and effector function
US8883980B2 (en) 2003-11-05 2014-11-11 Roche Glycart Ag Antigen binding molecules with increased Fc receptor binding affinity and effector function
US8216573B2 (en) 2006-09-22 2012-07-10 Wacker Chemie Ag Process for the fermentative production of antibodies
US20080206818A1 (en) * 2006-09-22 2008-08-28 Wacker Chemie Ag Process for the fermentative production of antibodies
US20110201022A1 (en) * 2008-07-30 2011-08-18 Biomarin Pharmaceutical Inc. Assays for detection of phenylalanine ammonia-lyase and antibodies to phenylalanine ammonia-lyase
US9557340B2 (en) 2008-07-30 2017-01-31 Biomarin Pharmaceutical Inc. Assays for detection of phenylalanine ammonia-lyase and antibodies to phenylalanine ammonia-lyase
US9052304B2 (en) 2009-03-13 2015-06-09 Terrasep, Llc Methods and apparatus for centrifugal liquid chromatography
US8293101B2 (en) 2009-03-13 2012-10-23 Terrasep, Llc Methods and apparatus for centrifugal liquid chromatography
US8277651B2 (en) 2009-03-13 2012-10-02 Terrasep, Llc Methods and apparatus for centrifugal liquid chromatography
US8277650B2 (en) 2009-03-13 2012-10-02 Terrasep, Llc Methods and apparatus for centrifugal liquid chromatography
US8293100B2 (en) 2009-03-13 2012-10-23 Terrasep, Llc Methods and apparatus for centrifugal liquid chromatography
US8815242B2 (en) 2009-05-27 2014-08-26 Synageva Biopharma Corp. Avian derived antibodies
US20100303806A1 (en) * 2009-05-27 2010-12-02 Synageva Biopharma Corp. Avian derivedantibodies
US10752696B2 (en) 2009-09-11 2020-08-25 Genentech, Inc. Highly concentrated pharmaceutical formulations
US10377831B2 (en) 2009-09-11 2019-08-13 Genentech, Inc. Highly concentrated pharmaceutical formulations
US10280227B2 (en) 2009-09-11 2019-05-07 Genentech, Inc. Highly concentrated pharmaceutical formulations
US20110076273A1 (en) * 2009-09-11 2011-03-31 Genentech, Inc. Highly Concentrated Pharmaceutical Formulations
US10221408B2 (en) 2010-02-04 2019-03-05 Biomarin Pharmaceutical Inc. Compositions of prokaryotic phenylalanine ammonia-lyase variants and methods of using compositions thereof
WO2011097335A3 (en) * 2010-02-04 2011-10-20 Biomarin Pharmaceutical Inc. Compositions of prokaryotic phenylalanine ammonia-lyase variants and methods of using compositions thereof
US11505790B2 (en) 2010-02-04 2022-11-22 Biomarin Pharmaceutical Inc. Compositions of prokaryotic phenylalanine ammonia-lyase variants and methods of using compositions thereof
US9410972B2 (en) 2012-08-21 2016-08-09 Janssen Pharmaceutica Nv Antibodies to quetiapine and use thereof
US10793644B2 (en) 2012-08-21 2020-10-06 Janssen Pharmaceutica Nv Antibodies to risperidone haptens and use thereof
US9850318B2 (en) 2012-08-21 2017-12-26 Janssen Pharmaceutica Nv Antibodies to quetiapine haptens and use thereof
US9664700B2 (en) 2012-08-21 2017-05-30 Janssen Pharmaceutica Nv Antibodies to risperidone and use thereof
US9611332B2 (en) 2012-08-21 2017-04-04 Janssen Pharmaceutica Nv Antibodies to aripiprazole haptens and use thereof
US10175257B2 (en) 2012-08-21 2019-01-08 Janssen Pharmaceutica Nv Antibodies to aripiprazole and use thereof
US9494607B2 (en) 2012-08-21 2016-11-15 Janssen Pharmaceutica Nv Antibodies to aripiprazole and use thereof
US9494608B2 (en) 2012-08-21 2016-11-15 Janssen Pharmaceutica Nv Antibodies to olanzapine and use thereof
US10288631B2 (en) 2012-08-21 2019-05-14 Janssen Pharmaceutica Nv Antibodies to quetiapine and use thereof
US10344098B2 (en) 2012-08-21 2019-07-09 Janssen Pharmaceutica Nv Antibodies to olanzapine and use thereof
US10370457B2 (en) 2012-08-21 2019-08-06 Janssen Pharmaceutica Nv Antibodies to paliperidone haptens and use thereof
US10379105B2 (en) 2012-08-21 2019-08-13 Janssen Pharmaceutica Nv Antibodies to aripiprazole haptens and use thereof
US9465041B2 (en) 2012-08-21 2016-10-11 Janssen Pharmaceutica Nv Antibodies to paliperidone and use thereof
US10379129B2 (en) 2012-08-21 2019-08-13 Janssen Pharmaceutica Nv Antibodies to paliperidone and use thereof
CN104737017A (en) * 2012-08-21 2015-06-24 奥索临床诊断有限公司 Antibodies to risperidone and use thereof
WO2014031648A3 (en) * 2012-08-21 2014-05-01 Ortho-Clinical Diagnostics, Inc Antibodies to risperidone and use thereof
US11385246B2 (en) 2012-08-21 2022-07-12 Saladax Biomedical Inc. Antibodies to paliperidone and use thereof
US10465013B2 (en) 2012-08-21 2019-11-05 Janssen Pharmaceutica Nv Antibodies to quetiapine haptens and use thereof
US10488401B2 (en) 2012-08-21 2019-11-26 Janssen Pharmaceutica Nv Antibodies to aripiprazole haptens and use thereof
US10690686B2 (en) 2012-08-21 2020-06-23 Janssen Pharmaceutica Nv Antibodies to risperidone and use thereof
US10712353B2 (en) 2012-08-21 2020-07-14 Janssen Pharmaceutica Nv Antibodies to olanzapine haptens and use thereof
WO2014031630A3 (en) * 2012-08-21 2014-05-08 Ortho-Clinical Diagnostics, Inc Antibodies to paliperidone and use thereof
US9751953B2 (en) 2012-08-21 2017-09-05 Janssen Pharmaceutica Nv Antibodies to risperidone haptens and use thereof
US10816561B2 (en) 2012-08-21 2020-10-27 Janssen Pharmaceutica Nv Antibodies to aripiprazole and use thereof
US11226345B2 (en) 2012-08-21 2022-01-18 Janssen Pharmaceutica Nv Antibodies to olanzapine haptens and use thereof
US11046786B2 (en) 2012-08-21 2021-06-29 Janssen Pharmaceutica Nv Antibodies to olanzapine and use thereof
US11225527B2 (en) 2012-08-21 2022-01-18 Janssen Pharmaceutica Nv Antibodies to paliperidone haptens and use thereof
US11105793B2 (en) 2012-08-21 2021-08-31 Janssen Pharmaceutica Nv Antibodies to aripiprazole haptens and use thereof
US11104742B2 (en) 2015-12-17 2021-08-31 Janssen Pharmaceutica Nv Antibodies to quetiapine and use thereof
US10852313B2 (en) 2015-12-17 2020-12-01 Janssen Pharmaceutica Nv Antibodies to risperidone and use thereof
US10444250B2 (en) 2015-12-17 2019-10-15 Janssen Pharmaceutica Nv Antibodies to risperidone and use thereof
US10435478B2 (en) 2015-12-17 2019-10-08 Janssen Pharmaceutica Nv Antibodies to quetiapine and use thereof

Also Published As

Publication number Publication date
US6652852B1 (en) 2003-11-25
US6893625B1 (en) 2005-05-17

Similar Documents

Publication Publication Date Title
US5677180A (en) Chimeric antibody with specificity to human B cell surface antigen
US6893625B1 (en) Chimeric antibody with specificity to human B cell surface antigen
EP0266663B1 (en) Chimeric antibody with specificity to human tumor antigen
EP0247091B1 (en) Modular assembly of antibody genes, antibodies prepared thereby and use
EP0550400B1 (en) Modular assembly of antibody genes, antibodies prepared thereby and use
US5595898A (en) Modular assembly of antibody genes, antibodies prepared thereby and use
US5846818A (en) Pectate lyase signal sequence
EP0364096B1 (en) Gene expression elements and the production of chimeric mouse-human antibodies
CA2019323A1 (en) Chimeric mouse-human km10 antibody with specificity to a human tumor cell antigen
AU606653C (en) Chimeric antibody with specificity to human B cell surface antigen
EP0536566A1 (en) Modular assembly of antibody genes, antibodies prepared thereby and use

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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