US20030035799A1 - Glycosylated antibody - Google Patents

Glycosylated antibody Download PDF

Info

Publication number
US20030035799A1
US20030035799A1 US10/145,992 US14599202A US2003035799A1 US 20030035799 A1 US20030035799 A1 US 20030035799A1 US 14599202 A US14599202 A US 14599202A US 2003035799 A1 US2003035799 A1 US 2003035799A1
Authority
US
United States
Prior art keywords
antibody
human
cell
disease
cho
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
US10/145,992
Inventor
Martin Page
James Crowe
Nicholas Rapson
Michael Keen
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.)
SmithKline Beecham Corp
Original Assignee
Glaxo Wellcome Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=10683862&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20030035799(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Glaxo Wellcome Inc filed Critical Glaxo Wellcome Inc
Priority to US10/145,992 priority Critical patent/US20030035799A1/en
Assigned to SMITHKLINE BEECHAM CORPORATION reassignment SMITHKLINE BEECHAM CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CROWE, JAMES S., RAPSON, NICHOLAS T., KEEN, MICHAEL J., PAGE, MARTIN J.
Publication of US20030035799A1 publication Critical patent/US20030035799A1/en
Priority to US12/122,123 priority patent/US20080279852A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2812Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD4
    • 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
    • C07K16/2893Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD52
    • 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
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation

Definitions

  • the present invention relates to Chinese hamster ovary (CHO) cell lines, to the production of proteins, in particular antibodies from such cell lines, also to antibodies having CHO glycosylation.
  • CHO Chinese hamster ovary
  • Antibodies or immunoglobulins are proteinaceous bifunctional molecules.
  • One region which is highly variable between different antibodies is responsible for binding to an antigen (Fab region), for example the many different infectious agents that the body may encounter, whilst the second, constant region (or Fc region) is responsible for binding to the Fc receptors of cells and also activates complement.
  • Fab region antigen
  • Fc region constant region
  • An antibody molecule is composed of two light chains and two heavy chains that are held together by interchain disulphide bonds. Each light chain is linked to a heavy chain by disulphide bonds and the two heavy chains are linked to each other by disulphide bonds.
  • Each heavy chain has at one end a variable domain followed by a number of constant domains, and each light chain has a variable domain at one end and a constant domain at the other end.
  • the light chain variable domain is aligned with the variable domain of the heavy chain.
  • the light chain constant domain is aligned with the first constant domain of the heavy chain.
  • the remaining constant domains of the heavy chains are aligned with each other. The constant domains in the light and heavy chains are not involved directly in binding the antibody to the antigen.
  • variable domains of each pair of light and heavy chains form the antigen binding site. They have the same general structure with each domain comprising a framework of four regions, whose sequences are relatively conserved, connected by three complementarity determining regions (CDRs).
  • CDRs complementarity determining regions
  • the four framework regions largely adopt a beta-sheet conformation and the CDRs form loops connecting, and in some cases comprising part of, the beta-sheet structure.
  • the CDRs are held in close proximity by the framework regions and, with the CDRs from the other domain, contribute to the formation of the antigen binding site.
  • hybridomas have the properties of antibody production derived from spleen cells and of continous growth derived from the myeloma cells.
  • Each hybridoma synthesizes and secretes a single antibody to a particular determinant of the original antigen. To ensure that all cells in a culture are identical, i.e.
  • the hybridomas resulting from cell fusion are cloned and subcloned. In this way, the cloned hybridomas produce homogeneous or monoclonal antibodies.
  • chimeric antibodies When, for example, murine (or ratine) monoclonal antibodies are used in human therapy, the induction of an human anti-mouse antibody response is due to the murine origin of the constant domains and four framework regions. This problem has therefore been addressed by the development of antibodies of two basic types.
  • the first type referred to as chimeric antibodies, is where the murine constant domains only are replaced by equivalent domains of human origin (Morrison et al, P.N.A.S., 1984, 81, 6851-6855; Boulianne et al, Nature, 1985, 314, 268-270; and Neuberger et al, Nature, 1985, 314, 268-270).
  • the second type is where the murine constant domains and the murine framework regions are all replaced by equivalent domains and regions of human origin.
  • This second type of antibody is referred to as a humanised or CDR-grafted antibody (Jones et al, Nature, 1986, 321, 522-525; and Riechmann et al, Nature, 1988, 332, 323-327).
  • a human antibody would of course avoid the need for “humanisation”, however cell lines which secrete human antibodies are very unstable and have generally proven unsuitable for commercial scale production.
  • dhfr dihydrofolate reductase
  • CHO Chinese hamster ovary
  • a process has now been developed that enables the balanced expression of the light and heavy chains of an antibody from CHO cells. Balanced expression is desirable given that the light and heavy chains are linked together in the antibody molecule in equimolar proportions. This process allows the antibody to be obtained in functional form and to be secreted in good yields. Thus the process enables sufficient quantities of functional antibody to be obtained for use in the immunotherapy of pathological disorders.
  • the invention therefore provides a CHO cell line capable of producing antibody, the cell line having been co-transfected with a vector capable of expressing the light chain of the antibody and a vector capable of expressing the heavy chain of the antibody wherein the vectors contain independently selectable markers.
  • the present invention further provides a CHO cell line capable of producing a human antibody or an altered antibody, the cell line having been co-transfected with a vector containing cDNA encoding the light chain of the antibody and a vector containing cDNA encoding the heavy chain of the antibody said vectors capable of expressing the light and heavy chains of the antibody.
  • the vectors may advantageously contain independently selectable markers. Hereafter, reference to the markers includes the singular and vice versa.
  • the cell line of the present invention is capable of producing all kinds of antibodies that generally comprise equimolar proportions of light and heavy chains.
  • the invention therefore includes human antibodies wherein the amino acid sequences of the heavy and light chains are homologous with those sequences of antibodies produced by human lymphocytes in vivo or in vitro by hybridomas.
  • altered antibodies such as hybrid antibodies in which the heavy and light chains are homologous to a natural antibody but are combined in a way that would not occur naturally.
  • a bispecific antibody has antigen binding sites specific to more than one antigen.
  • the constant region of the antibody may relate to one or other of the antigen binding regions or may be from a further antibody.
  • chimaeric antibodies have variable regions from one antibody and constant regions from another.
  • chimaeric antibodies may be species/species chimaeras or class/class chimaeras.
  • Such chimaeric antibodies may have one or more further modifications to improve antigen binding ability or to alter effector functioning.
  • Another form of altered antibody is a humanised or CDR-grafted antibody including a composite antibody, wherein parts of the hypervariable regions in additon to the CDRs are transferred to the human framework. Additional amino acids in the framework or constant regions of such antibodies may be altered. Included in the definition of altered antibody are Fab fragments which are roughly equivalent to the Y branch portions of the heavy and light chains; these may be included incomplete fragments or fragments including part of the Fc region. Thus, within the scope of the invention is included, any altered antibody in which the amino acid sequence is not one which exists in nature.
  • the cell line of the invention is preferentially employed for the production of altered antibodies most preferably chimaeric antibodies or CDR-grafted antibodies.
  • Particular examples of these include antibodies against T cell markers such as CD2, CD3, CD4, CD5, CD7, CD8, CD11a, CD11b, CD18, CD19, CD25, CD45 and CDw52 and especially CDR grafted antibodies against the CDw52 antigen, such as Campath-1H (Campath is a Trademark of the Wellcome Foundation Ltd) described in EP 328404
  • Further examples include CDR-grafted antibodies against various cancer cell marker antigens such as CD33 and CD38.
  • the resulting colonies may be selected using both markers. Colonies exhibiting the dual phenotype are generally capable of co-expressing both the light and heavy chains.
  • the selectable markers may or may not be of a dominant nature. Examples of selectable markers for use in co-transfection include adenosine deaminase (Kaufman et al, P.N.A.S, 1989, 83, 3136-40) asparagine synthetase (Cartier et al, Mol. Cell Biol., 1987, 7, 1623-28), E.
  • Coli trpB gene and Salmonella hisD gene (Hartman et al, P.N.A.S., 1988, 85, 8407-51), M2 mouse ribonucleotide reductase (Thelander et al, EMBO J, 1989, 8, 2475-79), human multidrug resistance gene (Kate et al, Gene, 1989, 84, 439-446), glutamine synthetase (Bebbington et al, DNA Cloning, Vol III, 1987, Ed. D. M.
  • dhfr which is usually employed with a parental CHO cell line of the dhfr ⁇ phenotype (Urlaub et al, P.N.A.S. 1980, 77, 4216-4220).
  • Successfully co-transfected CHO cells will possess the dhfr + phenotype and can readily be selected by culturing the colonies on media devoid of thymidine and hypoxanthine and optionally containing methotrexate (MTX).
  • a preferred selectable marker for use with the other of the vectors is a dominant resistance marker, such as neomycin (neo).
  • CHO cells successfully transfected with this marker can readily be selected by culturing the colonies on media containing the antibiotic, G418, otherwise known as Geneticin.
  • a second preferred system of selection and amplification is provided by the glutamine synthetase selectable marker or (GS system) which is described in WO87/04462.
  • GS system glutamine synthetase selectable marker
  • CHO cells which have been successfully transfected with the gene encoding the GS enzyme and the desired antibody gene can be selected by culturing colonies in media devoid of glutamine as described in PCT published application number WO87/04462.
  • At least one of the selectable markers preferably also provides the basis upon which the genes encoding the light and heavy chains may be amplified.
  • the vector DNAs are often integrated into the chromosome of the cell at the same locus.
  • One particularly preferred selectable marker for use in this way is dhfr which enables the desired amplification to be obtained through the use of increasing concentrations of MTX.
  • a second preferred selectable marker is GS which allows amplification by the addition of methionine sulphoximine (MSX).
  • the selectable markers are of course under the control of regulatory elements of DNA so as to provide for their expression.
  • the regulatory elements are preferably of a vital source, such as from DNA tumour viruses.
  • Particularly preferred are the use of an SV40 or adenovirus major late promoter. It is particularly advantageous in this regard to remove the enhancer element from the promoter thus effectively “crippling” it. This modification allows for increased levels of gene amplification at each concentration of methotrexate selection than would otherwise occur if a strong promoter was used.
  • an example of a suitable promoter is the mouse metallothionein promoter.
  • the light and heavy chain genes may constitute genomic DNA or, preferably, cDNA, and are cloned using procedures known in the art (Molecular Cloning: A Laboratory Manual, Second Edition, Maniatis et al, Cold Spring Harbor).
  • the genes are also under the control of regulatory elements of DNA so as to provide for their expression.
  • the use of the same regulatory elements for both chains is preferred so that their expression is substantially balanced.
  • the regulatory elements may be of vital origin and examples include those mentioned above in conjunction with the expression of dhfr as a selectable marker. Another example is the use of the ⁇ -actin promoter and cognate ⁇ -actin polyadenylation signal.
  • One or both of the vectors may also contain an SV40 origin of replication to allow for the vector constructs to be checked by rapid transient assay.
  • Construction of the expression vectors may be carried out in accordance with procedures known in the art (Molecular Cloning: A Laboratory Manual, Second Edition, Maniatis et al, Cold Spring Harbor).
  • Co-transfection of the CHO cell line with the expression vectors may be carried out simply by using equimolar quantities of both vectors and standard transfection procedures, such as calcium phosphate precipitation or lipofectin. Selection of the desired co-transfected cell line may be carried out in accordance with standard procedures known for the particular selectable markers.
  • the present invention also provides a process for the production of an antibody which comprises culturing a CHO cell line of the present invention.
  • Culture of the CHO cell line may be carried out in serum-containing or preferably serum and protein free media.
  • the medium preferably lacks hypoxanthine and/or thymidine and optionally contains MTX.
  • a selectable marker is glutamine synthetase the medium preferably lacks glutamine and optionally contains MSX.
  • Expression of both chains in substantially equimolar proportions enables optimum yields of functional antibody to be obtained.
  • the two chains assemble within the cell and are then secreted into the culture medium as functional antibody.
  • the resulting antibody may be purified and formulated in accordance with standard procedures.
  • Antibodies are glycoproteins containing between 3 and 12% carbohydrate.
  • the carbohydrate units are transferred to acceptor sites on the antibody chains after the heavy and light chains have combined.
  • the major carbohydrate units are attached to amino acid residues of the constant region of the antibody.
  • Carbohydrate is also known to attach to the antigen binding sites of some antibodies and may affect the antibody-binding characteristics by limiting access of the antigen to the antibody binding site. There are a number of roles associated with the carbohydrate units. They may affect overall solubility and the rate of catabolism of the antibody. It is also known that carbohydrate is necessary for cellular secretion of some antibody chains.
  • glycosylation of the constant region plays a vital role in the effector functioning of an antibody; without this glycosylation in its correct configuration, the antibody may be able to bind to the antigen but may not be able to bind for example to macrophages, helper and suppressor cells or complement, to carry out its role of blocking or lysing the cell to which it is bound.
  • the invention therefore provides an antibody having CHO glycosylation.
  • Such antibodies may be natural, such as human antibodies, altered antibodies for example hybrid antibodies or bispecific antibodies, chimaeric or CDR-grafted antibodies, including Fab fragments.
  • the CHO glycosylation may be associated with the antigen binding site or other parts of the variable domain. It may alternatively or additionally be associated with the constant region.
  • the glycosylated antibody is prepared by expression of the antibody genes in a suitably engineered CHO cell followed by recovery and if necessary, purification of the antibody from the cell culture medium.
  • CHO glycosylated antibodies are useful in medical therapy for treating numerous human disorders, generally as immunosuppressives more particularly for example T-cell mediated disorders including severe vasculitis, rheumatoid arthritis, systemic lupis, also autoimmune disorders such as multiple sclerosis, graft vs host disease, psoriarsis, Juvenile onset diabetes, Sjogrens' disease, thyroid disease, myasthenia gravis, transplant rejection and asthma.
  • T-cell mediated disorders including severe vasculitis, rheumatoid arthritis, systemic lupis, also autoimmune disorders such as multiple sclerosis, graft vs host disease, psoriarsis, Juvenile onset diabetes, Sjogrens' disease, thyroid disease, myasthenia gravis, transplant rejection and asthma.
  • These antibodies are also useful in treating cancer such as Non-Hodgkins lymphoma, multiple myeloma, and infectious diseases such as HIV and herpes.
  • the invention therefore provides the use of CHO glycosylated antibodies in the manufacture of a medicament for the treatment of any of the aforementioned disorders. Also provided is a method of treating a human being having any such a disorder comprising administering to said individual a therapeutically effective amount of a CHO glycosylated antibody.
  • the dosages of such antibodies will vary with the condition being treated and the recipient of the treatment, but will be in the range 1 to about 100 mg for an adult patient preferably 1-10 mg usually administered daily for a period between 1 and 30 days.
  • a two part dosing regime may be preferable wherein 1-5 mg are administered for 5-10 days followed by 6-15 mg for a further 5-10 days.
  • formulations containing CHO glycosylated antibody preferably include, in addition to antibody, a physiologically acceptable diluent or carrier possibly in admixture with other agents such as other antibodies or an antibiotic.
  • Suitable carriers include but are not limited to physiological saline, phosphate buffered saline, phosphate buffered saline glucose and buffered saline.
  • the antibody may be lyophilised (freeze dried) and reconstituted for use when needed by the addition of an aqueous buffered solution as described above.
  • Routes of administration are routinely parenteral including intravenous, intramuscular, subcutaneous and intraperitoneal injection or delivery.
  • the small box with the dashed arrow is the weakened SV40 promoter; the larger dotted box with an arrow is the ⁇ -actin promoter; polyA refers to respectively sourced polyadenylation and termination signals; the small box with ori contains the SV40 origin of replication;
  • the box with an arrow and MT refers to the mouse metallothionein promoter. Restriction sites indicated are: -H, HindIII; Bg, BglII; B, BamHI; R1, EcoR1.
  • the myeloma cell line TF57 (Hale et al, ibid,) was used to generate size selected cDNA fractions of 0.9-1.2 kb and 1.4-1.7 kb for the light and heavy chain cDNAs respectively. These were used to make EcoR1 Tinkered cDNA libraries in ⁇ gt10. All procedures were as described by Huynh et al ( DNA Cloning, Vol I: A Practical Approach, 1984, Glover, D(Editor), IRL Press, Oxford). The libraries were screened using [ 32 P] nick translated probes specific for the variable regions to isolate full length cDNA clones.
  • the 5′ untranslated leader was removed up to position ⁇ 32 using Bal-31 exonuclease and a HindIII linker added.
  • a unique SacI site 47 bp upstream of the stop codon was used to regenerate this sequence and position the HindIII site immediately after the stop codon.
  • the unique NcoI site overlapping the ATG start codon was used to re-build a 29 bp untranslated leader, identical to that of the light chain, using a HindIII-NcoI oligonucleotide pair.
  • the unique NaeI site 12 bp downstream of the stop codon was converted into a HindIII site using linkers.
  • the human ⁇ .-actin promoter was excised from pH ⁇ APr-3-neo (which corresponds to pH ⁇ APr-1-neo (Gunning et al, P.N.A.S., 1987, 84, 483-35) except that the SV40 polyadenylation/termination signal has been replaced with the respective human ⁇ -actin signals) as a 2860 bp PvuII-HindIII fragment, in which the PvulI site was subsequently converted to a BglII site using linkers.
  • an SphI site 1.4 kb downstream of the unique HindIII site was converted to a BamHI site using linkers.
  • the basal dhfr vector called p104 was constructed as follows The SDhI site at position ⁇ 128 in the SV40 promoter in pSV2dhfr (Subramani et al, Mol. Cell. Biol., 1981, 1, 54-864) was converted into a SalI site to remove all enhancer elements from the promoter. The weakened dhfr expression unit was then subcloned as a SalI-BamHI fragment into the homologous sites in pSVOd (Mellon et al., Cell, 1981, 27, 279-288).
  • the p104 vector was digested with BamHI, phosphatased, and ligated with three other fragments consisting of the BglII-HindIII ⁇ -actin promoter, the HindIII Campath-1H light chain cDNA and the HindIII-BamHI ⁇ -actin polyA/termination signals.
  • the construct pdBPV-MMTneo (Law et al, Mol. Cell. Biol., 1993, 3, 2110-2115) was digested with BamHI, phosphatased, and the fragment containing the neomycin gene isolated following separation on an agarose gel. This was ligated to the two ⁇ actin fragments and the Campath-1H heavy chain cDNA.
  • the constructs, pLD9 and pNH316 are depicted in FIG. 1.
  • the dhfr ⁇ CHO cell line DUK-B11 (Urlaub et al, P.N.A.S., 1980, 77, 4216-4220) was grown in Iscove's MEM supplemented with 10% fetal bovine serum, and 4 ⁇ g/ml each of hypoxanthine and thymidine.
  • Each pooled cell population was then cultured in the presence of 10 ⁇ 7 M MTX, and after two weeks, resistant colonies were again pooled and titred for Campath-1H production. There was a considerable increase in yield of up to 80-fold (Table 1). These cells were dilution cloned, screened for Campath-1H yield, and two high producer lines isolated, called A37 and 3D9 (Table 1). These were both amplified further in the presence of 10-6 M MTX, then dilution cloned and screened as above.
  • the co-transfection vectors pLD9 and pNH316 were further employed to evaluate an alternative amplification strategy to the one described above.
  • the dhfr ⁇ CHO cells were co-transfected as usual, and two days later split directly into a series of flasks containing G418 (for neomycin selection) and increasing concentrations of MTX ranging from 3 ⁇ 10 ⁇ 9 M to 10 ⁇ 7 M. Following two weeks of this selection, the number of resistant colonies were counted and pooled for each flask. When the cell populations had stabilized, they were assayed for Campath-1H antibody titres and the results are shown in Table 2.
  • Campath-1H ELISA assay Microtiter plates were coated with anti-human IgG and incubated with the assay sample (in culture medium). Antibody detection was visualized by using an anti-human gamma chain specific peroxidase conjugate.
  • the complement lysis assay is a measure of antibody function expressed as specific activity, determined by the ability of a CHO-glycosylated antibody of known concentration to bind to a pre-determined number of cells and effect cell lysis.
  • the assay is carried out on Campath 1H from Example 4 using Karpas 422 cells (established from B-cell non-Hodgkin lymphoma cell line—Dyer et al., (1990) Blood, 75 704-714) expressing Campath antigen on the cell surface. 1.2 ⁇ 10 7 cells were loaded with radiolabel by incubating for 2 hours at 37° C. in a CO 2 incubator in the presence of 600 ⁇ Ci of 51 Cr (sodium chromate).
  • the cDNA encoding the humanised CD4 light chain was cloned into pLD9 [Page and Sydenham, M. A. 1991 Biotechnology 9 64-68].
  • the resulting plasmid was designated p2110.
  • the humanised CD4 heavy chain was sequenced and cloned into a modified version of plasmid p342-12 [Law M-F., Byrne, J. C. and Hinley, P. M. 1983 Mol. Cell. Biol. 3 2110-2115).
  • Plasmid p342-12 was digested with BamH1 to remove the 7.4 kbp fragment containing part of the BPV-1 genome.
  • the backbone containing the ⁇ -lactamase gene and the neomycin resistance gene under the control of the mouse metallothionine promoter was purified and religated at the BamH1 site.
  • This plasmid was digested with HindIII, incubated with the large fragment of DNA polymerase I to remove the HindIll site and then religated.
  • Plasmid pBan1 therefore, consisted of the neomycin resistance gene, the ⁇ -lactamase gene and the ⁇ -actin expression cassette containing the unique HinDIII site.
  • the cDNA encoding the humanised heavy chain was cloned into this site and the resulting plasmid containing the correctly orientated insert was designated pBanCD4H.
  • p2110 and pBanCD4H contained a different selectable marker and co-transfection into recipient dhfr ⁇ CHO cells would permit the direct selection and isolation of dhfr + /neo r colonies. Cells exhibiting this phenotype should express functional antiCD4 antibody and could be amplified to elevate the antibody titres.
  • the dhfr ⁇ CHO line DUK-B11 [Urlaub, G. and Chasin, L. A. 1980 Proc. Natl. Acad. Sci. USA 77 4216-4220] was propagated in Iscoves MEM medium supplemented with 10% foetal bovine serum and 4 ⁇ g each of hypoxanthine and thymidine (all Flow). After transfection, transformants were selected in the medium described above except that the hypoxanthine/thymidine were omitted and dialysed foetal bovine serum was used. In addition, G418 was included at 500 ⁇ g/ml. To induce spontaneous amplification of sequences containing and flanking the dhfr gene, MTX was added to a concentration of 0.1 ⁇ M.
  • the dhfr ⁇ CHO cell line DUK-B11 was co-transfected with 5 ⁇ g of p2110 and 5 ⁇ g of pBanCD4H using the transfectant reagent under the conditions recommended by the manufacturer. Transformants were selected for the dhfr + /neo r phenotype as described above. Several hundreds of transformants were observed and pooled. Initital titres indicated that the first round basal transformants were secreting about 0.1 ⁇ g/ml/day. This pooled population was then cultured in the presence of 0.1 ⁇ M MTX for about 14 days. Resistant colonies were again pooled and assayed.
  • RNA and DNA was prepared from the various stages of amplification as described by Maniatis et al. [1982 Molecular Cloning. A Laboratory Manual. Cold Spring Harbour Laboratory, Cold Spring Harbour, N.Y.]. After fixing onto nitrocellulose filters, the nucleic acids were probed with [ 32 -P]- ⁇ ATP labelled DNA sequences of the heavy chain, the dhfr gene and the ⁇ -actin gene as a control “housekeeping” gene to eliminate artifacts due to loading errors.
  • the uncloned 0.1 ⁇ M MTX amplified pool was compared to the first round unamplified transformants and the untransformed parental B11 cells, with the probes described. Accordingly, no DNA signal was detected in the parental line when probed with the heavy chain but a weak signal was detected for dhfr. This is due to the single, non-functional dhfr allele in the B11 cell line. As a result, no RNA signal was detected with either probe. In contrast, a strong signal was detected with both probes on RNA and DNA in the primary transformants which reflects the start of expression.
  • the clonal D419 line was labelled with 35 S-methionine and cysteine and the intracellular and secreted antibody extracted by immunoprecipitation with appropriate antibodies. Following electrophoresis on reducing SDS-PAGE gels, the gels were dried and the signal detected by autoradiography.

Abstract

The invention relates to a CHO cell-line capable of producing antibody, the cell-line having been co-transfected with a vector capable of expressing the light chain of the antibody and a vector capable of expressing the heavy chain of the antibody wherein the vectors contain independently selectable markers; also included is a CHO cell-line capable of producing a human antibody or an altered antibody, the cell-line having been transfected with a vector capable of expressing the light chain of the antibody and the heavy chain of the antibody; process for the production of antibody using a CHO cell-line and antibody having CHO glycosylation.

Description

  • The present invention relates to Chinese hamster ovary (CHO) cell lines, to the production of proteins, in particular antibodies from such cell lines, also to antibodies having CHO glycosylation. [0001]
  • Antibodies or immunoglobulins are proteinaceous bifunctional molecules. One region which is highly variable between different antibodies is responsible for binding to an antigen (Fab region), for example the many different infectious agents that the body may encounter, whilst the second, constant region (or Fc region) is responsible for binding to the Fc receptors of cells and also activates complement. In this way, antibodies represent a vital component of the immune response of mammals in destroying foreign microorganisms and viruses. [0002]
  • An antibody molecule is composed of two light chains and two heavy chains that are held together by interchain disulphide bonds. Each light chain is linked to a heavy chain by disulphide bonds and the two heavy chains are linked to each other by disulphide bonds. Each heavy chain has at one end a variable domain followed by a number of constant domains, and each light chain has a variable domain at one end and a constant domain at the other end. The light chain variable domain is aligned with the variable domain of the heavy chain. The light chain constant domain is aligned with the first constant domain of the heavy chain. The remaining constant domains of the heavy chains are aligned with each other. The constant domains in the light and heavy chains are not involved directly in binding the antibody to the antigen. [0003]
  • The variable domains of each pair of light and heavy chains form the antigen binding site. They have the same general structure with each domain comprising a framework of four regions, whose sequences are relatively conserved, connected by three complementarity determining regions (CDRs). The four framework regions largely adopt a beta-sheet conformation and the CDRs form loops connecting, and in some cases comprising part of, the beta-sheet structure. The CDRs are held in close proximity by the framework regions and, with the CDRs from the other domain, contribute to the formation of the antigen binding site. [0004]
  • The immunisation of an animal with an antigen results in the production of different antibodies with different specificities and affinities. An antiserum obtained from the immunised animal will, therefore, be heterogeneous and contain a pool of antibodies produced by many different lymphocyte clones. Antibodies thus obtained are referred to as polyclonal antibodies and this polyclonal nature has been a major drawback in the use of antibodies in diagnostic assays and in therapeutic applications. [0005]
  • A major step forward occurred in 1975 when Kohler and Milstein ([0006] Nature, 1975, 256, 495-497) reported the successful fusion of spleen cells from mice immunized with an antigen with cells of a murine myeloma line. The resulting hybrid cells, termed hybridomas, have the properties of antibody production derived from spleen cells and of continous growth derived from the myeloma cells. Each hybridoma synthesizes and secretes a single antibody to a particular determinant of the original antigen. To ensure that all cells in a culture are identical, i.e. that they contain the genetic information required for the synthesis of a unique antibody species, the hybridomas resulting from cell fusion are cloned and subcloned. In this way, the cloned hybridomas produce homogeneous or monoclonal antibodies.
  • The advantages of hybridoma technology are profound. Because many hybrids arising from each spleen are screened for their potential to produce antibodies to the antigen of interest and only a few are selected, it is possible to immunize with impure antigens and yet obtain specific antibodies. The immortality of the cell line assures that an unlimited supply of a homogeneous, well-characterised antibody is available for use in a variety of applications including in particular diagnosis and immunotherapy of pathological disorders. Unfortunately, the usefulness of such monoclonal antibodies in a clinical setting can be severely hampered by the development of human anti-mouse antibodies—an anti-globulin response—which may interfere with therapy or cause allergic or immune complex hypersensitivity. [0007]
  • When, for example, murine (or ratine) monoclonal antibodies are used in human therapy, the induction of an human anti-mouse antibody response is due to the murine origin of the constant domains and four framework regions. This problem has therefore been addressed by the development of antibodies of two basic types. The first type, referred to as chimeric antibodies, is where the murine constant domains only are replaced by equivalent domains of human origin (Morrison et al, [0008] P.N.A.S., 1984, 81, 6851-6855; Boulianne et al, Nature, 1985, 314, 268-270; and Neuberger et al, Nature, 1985, 314, 268-270). The second type is where the murine constant domains and the murine framework regions are all replaced by equivalent domains and regions of human origin. This second type of antibody is referred to as a humanised or CDR-grafted antibody (Jones et al, Nature, 1986, 321, 522-525; and Riechmann et al, Nature, 1988, 332, 323-327). A human antibody would of course avoid the need for “humanisation”, however cell lines which secrete human antibodies are very unstable and have generally proven unsuitable for commercial scale production.
  • To generate sufficient quantities of antibody for full clinical use it is desirable to employ an efficient recombinant expression system. Since myeloma cells represent a natural host specialized for antibody production and secretion, cell lines derived from these have been used for the expression of recombinant antibodies. Often, complex vector design, based around immunoglobulin gene regulatory elements, is required, and final expression levels have been reported which are highly variable (Winter et al, [0009] Nature, 1988, 332, 323-327; Weidle et al, Gene, 1987, 60, 205-216; Nakatani et al, Bio/Technology, 1989, 7, 805-810; and Gillies et al, Bio/Technology, 1989, 7, 799-804).
  • An alternative mammalian expression system is that offered by the use of dihydrofolate reductase (dhfr) deficient Chinese hamster ovary (CHO) cells. The use of these cells has enabled the production of large quantities of several therapeutic proteins for research and clinical use (Kaufman et al, [0010] Mol. Cell. Biol, 1985, 5, 1750-1759; and Zettlmeissl et al, Bio/Technology, 1987, 5, 720-725). There are, however, very few instances of the use of these cells for the expression of antibodies and the levels of expression of murine antibodies reported to date are low—of the order of 0.01-0.1 μg/ml (Weidle et al, Gene, 1987, 51, 21-29; and Feys et al, Int. J. Cancer, 1988, 2, 26-27).
  • A process has now been developed that enables the balanced expression of the light and heavy chains of an antibody from CHO cells. Balanced expression is desirable given that the light and heavy chains are linked together in the antibody molecule in equimolar proportions. This process allows the antibody to be obtained in functional form and to be secreted in good yields. Thus the process enables sufficient quantities of functional antibody to be obtained for use in the immunotherapy of pathological disorders. [0011]
  • The invention therefore provides a CHO cell line capable of producing antibody, the cell line having been co-transfected with a vector capable of expressing the light chain of the antibody and a vector capable of expressing the heavy chain of the antibody wherein the vectors contain independently selectable markers. [0012]
  • The present invention further provides a CHO cell line capable of producing a human antibody or an altered antibody, the cell line having been co-transfected with a vector containing cDNA encoding the light chain of the antibody and a vector containing cDNA encoding the heavy chain of the antibody said vectors capable of expressing the light and heavy chains of the antibody. The vectors may advantageously contain independently selectable markers. Hereafter, reference to the markers includes the singular and vice versa. [0013]
  • The cell line of the present invention is capable of producing all kinds of antibodies that generally comprise equimolar proportions of light and heavy chains. The invention therefore includes human antibodies wherein the amino acid sequences of the heavy and light chains are homologous with those sequences of antibodies produced by human lymphocytes in vivo or in vitro by hybridomas. Also included in the invention are altered antibodies such as hybrid antibodies in which the heavy and light chains are homologous to a natural antibody but are combined in a way that would not occur naturally. For example, a bispecific antibody has antigen binding sites specific to more than one antigen. The constant region of the antibody may relate to one or other of the antigen binding regions or may be from a further antibody. Altered antibodies, such as chimaeric antibodies have variable regions from one antibody and constant regions from another. Thus, chimaeric antibodies may be species/species chimaeras or class/class chimaeras. Such chimaeric antibodies may have one or more further modifications to improve antigen binding ability or to alter effector functioning. Another form of altered antibody is a humanised or CDR-grafted antibody including a composite antibody, wherein parts of the hypervariable regions in additon to the CDRs are transferred to the human framework. Additional amino acids in the framework or constant regions of such antibodies may be altered. Included in the definition of altered antibody are Fab fragments which are roughly equivalent to the Y branch portions of the heavy and light chains; these may be included incomplete fragments or fragments including part of the Fc region. Thus, within the scope of the invention is included, any altered antibody in which the amino acid sequence is not one which exists in nature. [0014]
  • The cell line of the invention is preferentially employed for the production of altered antibodies most preferably chimaeric antibodies or CDR-grafted antibodies. Particular examples of these include antibodies against T cell markers such as CD2, CD3, CD4, CD5, CD7, CD8, CD11a, CD11b, CD18, CD19, CD25, CD45 and CDw52 and especially CDR grafted antibodies against the CDw52 antigen, such as Campath-1H (Campath is a Trademark of the Wellcome Foundation Ltd) described in EP 328404 Further examples include CDR-grafted antibodies against various cancer cell marker antigens such as CD33 and CD38. [0015]
  • After co-transfection into recipient CHO cells, the resulting colonies may be selected using both markers. Colonies exhibiting the dual phenotype are generally capable of co-expressing both the light and heavy chains. The selectable markers may or may not be of a dominant nature. Examples of selectable markers for use in co-transfection include adenosine deaminase (Kaufman et al, [0016] P.N.A.S, 1989, 83, 3136-40) asparagine synthetase (Cartier et al, Mol. Cell Biol., 1987, 7, 1623-28), E. Coli trpB gene and Salmonella hisD gene (Hartman et al, P.N.A.S., 1988, 85, 8407-51), M2 mouse ribonucleotide reductase (Thelander et al, EMBO J, 1989, 8, 2475-79), human multidrug resistance gene (Kate et al, Gene, 1989, 84, 439-446), glutamine synthetase (Bebbington et al, DNA Cloning, Vol III, 1987, Ed. D. M. Glover, 163-188, IRL Press), xanthine guanine phosphoribosyl transferase (gpt) (Mulligan et al, Science, 1980, 209, 1422-27), hygromycin B (Santerre et al, Gene, 1984, 30, 147-156), neomycin gene (Southern et al, J. Mol. Appl. Genet., 1982, 1, 327-341), and dihydrofolate reductase (Subramani et al, Mol. Cell Biol., 1981, 1, 854-864). One particularly preferred selectable marker is dhfr which is usually employed with a parental CHO cell line of the dhfr phenotype (Urlaub et al, P.N.A.S.1980, 77, 4216-4220). Successfully co-transfected CHO cells will possess the dhfr+ phenotype and can readily be selected by culturing the colonies on media devoid of thymidine and hypoxanthine and optionally containing methotrexate (MTX). A preferred selectable marker for use with the other of the vectors is a dominant resistance marker, such as neomycin (neo). CHO cells successfully transfected with this marker can readily be selected by culturing the colonies on media containing the antibiotic, G418, otherwise known as Geneticin.
  • A second preferred system of selection and amplification is provided by the glutamine synthetase selectable marker or (GS system) which is described in WO87/04462. CHO cells which have been successfully transfected with the gene encoding the GS enzyme and the desired antibody gene can be selected by culturing colonies in media devoid of glutamine as described in PCT published application number WO87/04462. [0017]
  • At least one of the selectable markers preferably also provides the basis upon which the genes encoding the light and heavy chains may be amplified. In co-transfection of a CHO cell line, the vector DNAs are often integrated into the chromosome of the cell at the same locus. Thus, the use of only one of the selectable markers as the basis for amplification normally results in a parallel increase in the copy number of both genes. One particularly preferred selectable marker for use in this way is dhfr which enables the desired amplification to be obtained through the use of increasing concentrations of MTX. A second preferred selectable marker is GS which allows amplification by the addition of methionine sulphoximine (MSX). [0018]
  • The selectable markers are of course under the control of regulatory elements of DNA so as to provide for their expression. In the case of the use of dhfr as a selectable marker, the regulatory elements are preferably of a vital source, such as from DNA tumour viruses. Particularly preferred are the use of an SV40 or adenovirus major late promoter. It is particularly advantageous in this regard to remove the enhancer element from the promoter thus effectively “crippling” it. This modification allows for increased levels of gene amplification at each concentration of methotrexate selection than would otherwise occur if a strong promoter was used. In the case of the use of neo as a selectable marker, an example of a suitable promoter is the mouse metallothionein promoter. [0019]
  • The light and heavy chain genes may constitute genomic DNA or, preferably, cDNA, and are cloned using procedures known in the art (Molecular Cloning: A Laboratory Manual, Second Edition, Maniatis et al, Cold Spring Harbor). The genes are also under the control of regulatory elements of DNA so as to provide for their expression. The use of the same regulatory elements for both chains is preferred so that their expression is substantially balanced. The regulatory elements may be of vital origin and examples include those mentioned above in conjunction with the expression of dhfr as a selectable marker. Another example is the use of the β-actin promoter and cognate β-actin polyadenylation signal. [0020]
  • One or both of the vectors may also contain an SV40 origin of replication to allow for the vector constructs to be checked by rapid transient assay. [0021]
  • Construction of the expression vectors may be carried out in accordance with procedures known in the art (Molecular Cloning: A Laboratory Manual, Second Edition, Maniatis et al, Cold Spring Harbor). [0022]
  • Co-transfection of the CHO cell line with the expression vectors may be carried out simply by using equimolar quantities of both vectors and standard transfection procedures, such as calcium phosphate precipitation or lipofectin. Selection of the desired co-transfected cell line may be carried out in accordance with standard procedures known for the particular selectable markers. [0023]
  • The present invention also provides a process for the production of an antibody which comprises culturing a CHO cell line of the present invention. Culture of the CHO cell line may be carried out in serum-containing or preferably serum and protein free media. In one preferred instance where the CHO cell line is a dhfr[0024] + transformant, the medium preferably lacks hypoxanthine and/or thymidine and optionally contains MTX. Where a selectable marker is glutamine synthetase the medium preferably lacks glutamine and optionally contains MSX. Expression of both chains in substantially equimolar proportions enables optimum yields of functional antibody to be obtained. The two chains assemble within the cell and are then secreted into the culture medium as functional antibody. The resulting antibody may be purified and formulated in accordance with standard procedures.
  • Antibodies are glycoproteins containing between 3 and 12% carbohydrate. The carbohydrate units are transferred to acceptor sites on the antibody chains after the heavy and light chains have combined. The major carbohydrate units are attached to amino acid residues of the constant region of the antibody. Carbohydrate is also known to attach to the antigen binding sites of some antibodies and may affect the antibody-binding characteristics by limiting access of the antigen to the antibody binding site. There are a number of roles associated with the carbohydrate units. They may affect overall solubility and the rate of catabolism of the antibody. It is also known that carbohydrate is necessary for cellular secretion of some antibody chains. It has been demonstrated that glycosylation of the constant region plays a vital role in the effector functioning of an antibody; without this glycosylation in its correct configuration, the antibody may be able to bind to the antigen but may not be able to bind for example to macrophages, helper and suppressor cells or complement, to carry out its role of blocking or lysing the cell to which it is bound. [0025]
  • It has now been found that antibody glycosylated by CHO cells maintains antigen binding capability and effector functionality. This has been demonstrated in vitro complement lysis assays and in vivo in a human patient. [0026]
  • The invention therefore provides an antibody having CHO glycosylation. Such antibodies may be natural, such as human antibodies, altered antibodies for example hybrid antibodies or bispecific antibodies, chimaeric or CDR-grafted antibodies, including Fab fragments. [0027]
  • The CHO glycosylation may be associated with the antigen binding site or other parts of the variable domain. It may alternatively or additionally be associated with the constant region. The glycosylated antibody is prepared by expression of the antibody genes in a suitably engineered CHO cell followed by recovery and if necessary, purification of the antibody from the cell culture medium. [0028]
  • CHO glycosylated antibodies are useful in medical therapy for treating numerous human disorders, generally as immunosuppressives more particularly for example T-cell mediated disorders including severe vasculitis, rheumatoid arthritis, systemic lupis, also autoimmune disorders such as multiple sclerosis, graft vs host disease, psoriarsis, Juvenile onset diabetes, Sjogrens' disease, thyroid disease, myasthenia gravis, transplant rejection and asthma. These antibodies are also useful in treating cancer such as Non-Hodgkins lymphoma, multiple myeloma, and infectious diseases such as HIV and herpes. [0029]
  • The invention therefore provides the use of CHO glycosylated antibodies in the manufacture of a medicament for the treatment of any of the aforementioned disorders. Also provided is a method of treating a human being having any such a disorder comprising administering to said individual a therapeutically effective amount of a CHO glycosylated antibody. [0030]
  • The dosages of such antibodies will vary with the condition being treated and the recipient of the treatment, but will be in the [0031] range 1 to about 100 mg for an adult patient preferably 1-10 mg usually administered daily for a period between 1 and 30 days. A two part dosing regime may be preferable wherein 1-5 mg are administered for 5-10 days followed by 6-15 mg for a further 5-10 days.
  • Also included within the invention are formulations containing CHO glycosylated antibody. Such formulations preferably include, in addition to antibody, a physiologically acceptable diluent or carrier possibly in admixture with other agents such as other antibodies or an antibiotic. Suitable carriers include but are not limited to physiological saline, phosphate buffered saline, phosphate buffered saline glucose and buffered saline. Alternatively, the antibody may be lyophilised (freeze dried) and reconstituted for use when needed by the addition of an aqueous buffered solution as described above. Routes of administration are routinely parenteral including intravenous, intramuscular, subcutaneous and intraperitoneal injection or delivery.[0032]
  • The accompanying drawings show: [0033]
  • FIG. 1[0034]
  • ([0035] a) the pLD9 construct containing expression cassettes for the ‘crippled’ dhfr selection/amplification marker and the Campath-1H light chain cDNA. The small box with the dashed arrow is the weakened SV40 promoter; the larger dotted box with an arrow is the β-actin promoter; polyA refers to respectively sourced polyadenylation and termination signals; the small box with ori contains the SV40 origin of replication;
  • ([0036] b) the pNH316 construct containing expression cassettes for the neomycin selection marker and the Campath-1H heavy chain cDNA. The box with an arrow and MT refers to the mouse metallothionein promoter. Restriction sites indicated are: -H, HindIII; Bg, BglII; B, BamHI; R1, EcoR1.
  • FIG. 2[0037]
  • Comparative determinations of the rate of Campath-1H synthesis in confluent A39 cells over 4 consecutive days. Following the [[0038] 35S] methionine pulse period, equal aliquots of cells (C) and culture medium (M) were Immuno-precipitated and separated by SDS-PAGE. The position of the Campath-1H heavy and light chains are indicated (H and L arrows). There was some loss of material for the day 3 cell sample.
  • FIG. 3[0039]
  • A pulse-chase experiment to determine the rate of secretion and distribution of radiolabelled Campath-1H in A39 cells. Confluent cells were pulsed with [[0040] 35S] methionine for 6 hours, then fresh medium containing an excess of unlabelled methionine was added. Equal aliquots of cells and culture medium were taken at the indicated time points (in hours following the end of the pulse period) and treated as described in the legend of FIG. 2. The samples for the 48 and 72 hour medium time points were run on a different gel to the 6 and 24 hour points, and the tracks are only lined up relative to the position of the heavy (H) chain.
  • FIG. 4[0041]
  • Shows growth of C1H 3D11* 44 in WCM5 (protein-free medium) in a 1 liter fermenter measured as cell count/ml over 90 days. [0042]
  • FIG. 5[0043]
  • Shows antibody production from C1H 3D* 44 cells in WCM5 in a 1 liter fermenter measured as micrograms of antibody/ml over 80 days.[0044]
  • The following Examples are provided purely for illustration of the present invention. [0045]
  • EXAMPLE 1 Cloning of the Heaving and Light Chain cDNAs for Campath-1H
  • The complementarity determining regions from the rat Campath-1G monoclonal were originally grafted directly into genomic human heavy and light chain frameworks (Winter et al, [0046] Nature, 1988, 322, 323-327). These constructs were engineered for expression in the myeloma cell line YO and resulted in yields of Campath-1H of up to 5 μg/ml following 10-14 days in culture (Hale et al, Tissue Antigens, 1990, 35, 118-127 and Winter et al, Nature, 1988, 325, 323-327). The myeloma cell line TF57 (Hale et al, ibid,) was used to generate size selected cDNA fractions of 0.9-1.2 kb and 1.4-1.7 kb for the light and heavy chain cDNAs respectively. These were used to make EcoR1 Tinkered cDNA libraries in λgt10. All procedures were as described by Huynh et al (DNA Cloning, Vol I: A Practical Approach, 1984, Glover, D(Editor), IRL Press, Oxford). The libraries were screened using [32P] nick translated probes specific for the variable regions to isolate full length cDNA clones. For the light chain cDNA, the 5′ untranslated leader was removed up to position −32 using Bal-31 exonuclease and a HindIII linker added. For the 3′ end, use was made of a unique SacI site 47 bp upstream of the stop codon. A SacI-HindIII oligonucleotide pair was used to regenerate this sequence and position the HindIII site immediately after the stop codon. For the 5′ end of the heavy chain cDNA, the unique NcoI site overlapping the ATG start codon was used to re-build a 29 bp untranslated leader, identical to that of the light chain, using a HindIII-NcoI oligonucleotide pair. At the 3′ end, the unique NaeI site 12 bp downstream of the stop codon was converted into a HindIII site using linkers.
  • EXAMPLE 2 Construction of Vectors
  • The human β.-actin promoter was excised from pHβAPr-3-neo (which corresponds to pHβAPr-1-neo (Gunning et al, [0047] P.N.A.S., 1987, 84, 483-35) except that the SV40 polyadenylation/termination signal has been replaced with the respective human β-actin signals) as a 2860 bp PvuII-HindIII fragment, in which the PvulI site was subsequently converted to a BglII site using linkers. To isolate the human β-actin polyadenylation and termination signals from pHβAPr-3-neo, an SphI site 1.4 kb downstream of the unique HindIII site was converted to a BamHI site using linkers. The basal dhfr vector called p104, was constructed as follows The SDhI site at position −128 in the SV40 promoter in pSV2dhfr (Subramani et al, Mol. Cell. Biol., 1981, 1, 54-864) was converted into a SalI site to remove all enhancer elements from the promoter. The weakened dhfr expression unit was then subcloned as a SalI-BamHI fragment into the homologous sites in pSVOd (Mellon et al., Cell, 1981, 27, 279-288).
  • To construct pLD9, the p104 vector was digested with BamHI, phosphatased, and ligated with three other fragments consisting of the BglII-HindIII β-actin promoter, the HindIII Campath-1H light chain cDNA and the HindIII-BamHI β-actin polyA/termination signals. To construct pNH316, the construct pdBPV-MMTneo (Law et al, [0048] Mol. Cell. Biol., 1993, 3, 2110-2115) was digested with BamHI, phosphatased, and the fragment containing the neomycin gene isolated following separation on an agarose gel. This was ligated to the two βactin fragments and the Campath-1H heavy chain cDNA. The constructs, pLD9 and pNH316 are depicted in FIG. 1.
  • EXAMPLE 3 Expression of Campath-1H in CHO Cells
  • The dhfr[0049] CHO cell line DUK-B11 (Urlaub et al, P.N.A.S., 1980, 77, 4216-4220) was grown in Iscove's MEM supplemented with 10% fetal bovine serum, and 4 μg/ml each of hypoxanthine and thymidine. 10 μg of pLD9 and pNH316 was co-precipitated onto cells using the calcium phosphate method, (Gorman et al, DNA Cloning, 1985, Vol II, 143-190, Academic Press, N.Y.) and selected for the double phenotype of dhfr+/neo resistance by using the medium above except that 10% dialysed serum was used, the hypoxanthine/thymidine were omitted, and G418 (Gibco) was included at 500 μg/ml. In some experiments MTX was included directly in the first round selection for dhfr+ transformants. Several hundred resistant colonies were pooled and assayed for the production of Campath-1H antibody in the culture medium. The average yield was 0.5 μg/ml for non-amplified first round transformants.
  • Each pooled cell population was then cultured in the presence of 10[0050] −7 M MTX, and after two weeks, resistant colonies were again pooled and titred for Campath-1H production. There was a considerable increase in yield of up to 80-fold (Table 1). These cells were dilution cloned, screened for Campath-1H yield, and two high producer lines isolated, called A37 and 3D9 (Table 1). These were both amplified further in the presence of 10-6 M MTX, then dilution cloned and screened as above. The increase in expression at this second, and final, amplification stage was not so dramatic as seen previously; nevertheless, when re-fed at confluence and left for a further 4 days, the cell lines A39 and 3D11 were capable of producing up to 200 μg/ml of Campath-1H.
    TABLE 1
    Expression Levels of Campath-1H using Stepwise Amplification
    Accumulated
    Campath-1H
    Construct Selection stage (μg/ml)
    pLD9 + pNH316 dhfr+/neo basal pool  0.5
    10−7 M MTX amplified pool  18-40
    Cell lines A37 and 3D9  40
    10−6 M MTX amplified pool  60-90
    Cell line A39 100
    Cell line 3D11 150-200
  • Legend to Table [0051]
  • Cells were allowed to reach confluence in a T175 tissue culture flask, then refed with fresh 50 ml of tissue culture medium and left for a further 4 days. The Campath1H antibody that had accumulated in the medium during this period was measured by ELISA. Total cell counts on the day of assay were usually 2.5×10[0052] 7. The yield from the 3D11 cell line reflects a productivity of 100 μg/106 cells/day.
  • The co-transfection vectors pLD9 and pNH316 were further employed to evaluate an alternative amplification strategy to the one described above. The dhfr[0053] CHO cells were co-transfected as usual, and two days later split directly into a series of flasks containing G418 (for neomycin selection) and increasing concentrations of MTX ranging from 3×10−9 M to 10−7 M. Following two weeks of this selection, the number of resistant colonies were counted and pooled for each flask. When the cell populations had stabilized, they were assayed for Campath-1H antibody titres and the results are shown in Table 2. As the MTX level was increased, there was a marked decrease in the number of surviving dhfr+ colonies, but they expressed proportionately more Campath-1H. Thus, in a one step direct selection at high concentrations of MTX, it is possible to isolate cell populations which produce up to 60-fold increase in antibody yield compared to cell populations selected for basal dhfr levels.
    TABLE 2
    Expression Levels of Campath-1H using Direct Selection
    Selection (M MTX) Accumulated
    (pg/ml) dhfr+ colonies Campath-1H
    No MTX 500  0.5
     3 × 10−9 40 2
    10−8  5 7
     3 × 10−8  5 30
    10−7
  • Legend to Table [0054]
  • Colonies at each MTX selection stage were pooled and assayed as described in the legend of Table 1. [0055]
  • This selection procedure was repeated following another co-transfection of cells, and in this instance, the entire population was selected in medium containing G418 and 3×10[0056] −8 M MTX. This generated a larger pool of resistant colonies which were subsequently pooled and re-amplified twice more using MTX concentrations of 6×10−7 M, then 3×10−6 M. At this stage, the cells were dilution cloned and screened for Campath-1H levels. The two highest producer cell lines isolated were capable of producing antibody levels up to 100-150 μg/ml and were designated as lines 4F11 and 5E10.
  • The growth rates of these cell lines, and the A39/3D11 lines described above, were considerably slower than the parental non-transformed dhfr[0057] CHO cells. This is usually a common feature of these cells once they have been engineered to express high quantities of a product gene. The yields from the 5E10 and 4F11 cell lines proved to be quite variable over time, and the latter appeared to have only a limited passage life lasting about 3 weeks before entering crisis and death. This instability was not evident at all in the other cell lines, although in general, the lines isolated from the second amplification procedure, including 5E10, were usually more fickle to culture. Of all the lines, the 3D11 coupled good growth and stability with high Campath-1H yields. To ensure the propagation of these features, the 3D11 cell line was dilution cloned once more to generate the 3D11* line and this similarly produced Campath-1H yields up to 200 μg/ml.
  • EXAMPLE 4 Growth of and Production from C1H 3D11* 44 in WCM4
  • a) C1H 3D11* cells growing as a monolayer in Iscoves +10% FBS Flow, non-essential amino acids, 10[0058] −6 M Methotrexate and antibiotics were approximately 90% confluent. These cells were removed from the plastic with trypsin/versene, washed in Iscoves medium without supplements, centrifuged and resuspended at 5×104 /ml in WCM4 medium Table 3+0.25% peptone +0.1% polyethylene glycol (PEG) 10,000+0.5% fetal boine serum (FBS) without methotrexate (MTX).
    TABLE 3
    Formulation for medium WCM4.
    Iscoves modification of DMEM without BSA, transferrin and
    lecithin. Available from GIBCO Ltd., Unit 4, Cowley Mill
    Td. Est., Uxbridge UBS 27G. Similar to published medium
    (Iscoves and Melcher (1978) J. Exp. Med. 1. 47, 923)
    without the bovine serum albumin, pure human tranferrin,
    or soyabean lecithin.
    + 5 ml/liter 200 mM L glutamine
    + 50 mg/liter L proline
    + 50 mg/liter L threonine
    + 50 mg/liter L methionine
    + 50 mg/liter L cysteine
    + 50 mg/liter L tyrosine
    + 25 mg/liter ascorbic acid
    + 0.062 mg/liter vitamin B6
    + 1.36 mg/liter vitamin B12
    + 0.2 mg/liter lipoic acid
    + 0.088 mg/liter methyl linoleate
    + 1 μM methotrexate
    + 1 mg/liter FeSO4
    + 1 mg/liter ZnSO4
    + 0.0025 mg/liter CuSO4
    + 5 mg/liter recombinant
    insulin (Nucellin)
    + 50,000 Iu/liter polymyxin
    + 20,000 Iu/liter neomycin
    + 0.16 mg/liter putrescine-2 HCL.
  • Three 25 cm[0059] 2 flasks were set up with 10 ml of cell suspension+hypoxanthine (H), thymidine (T) or HT. These flasks were incubated at 36.5° C. in 5% CO2 incubator.
  • After six days, the flasks were pooled and added to an equal volume of WCM4+MTX without peptone or PEG, and were transferred to a 75 cm[0060] 2 flask.
  • These cells were used to seed a 500 ml Techner spinner, incubated at 36.5° C. spinning at 40 rpm. Cells continued growing serum free for a period of over five months and although it was found that the cells needed a period of adaptation, the growth rate and viability steadily improved. The population doubling time was calculated to be 73.1 hours over approximately 7 weeks; this decreased to 47.4 hours over the subsequent 20 days then stabilised. Antibody secretion remained high at levels in excess of 60 μg/ml. It was determined that the gene copy number in these cells did not decrease according to band intensity using Northern blot analysis. [0061]
  • In fermenters, these cells produced antibody in excess of 70 μg/ml and regularly achieve levels of 100 μg/ml or more. These cells are denoted C1H 3D11* 44. [0062]
  • b) Cells from a) above which had been growing serum-free for over 2 months were transferred to a [0063] SGi 1 liter fermenter with a stainless steel angled paddle turning at 70 rpm. The temperature was set at 37° C., dO2 at 10% and pH control to 7-7.2. The fermenter was seeded on day 0 with 0.22×106 cells/ml in WCM4 (Table 3) with 0.1% polyethylene glycol (PEG) 10,000 and 0.25% soy peptone, and was top gassed with O2. The cells were routinely passaged using fresh medium and a split rate typically between 1 to 2 and 1 to 4.
  • On day 33 the top gassing was replaced with deep sparging which is can be expected to cause more physical damage to the cells. [0064]
  • On [0065] day 50 onwards WCM5 (Table 4) was used together with peptone and PEG instead of WCM4.
    TABLE 4
    Formulation for Medium WCM5
    Iscoves modification of DMEM without BSA,
    transferrin or lecithin (see Table 3).
    + 5 ml/liter 200 mM L glutamine
    + 50 mg/liter L proline
    + 50 mg/liter L threonine
    + 50 mg/liter L methionine
    + 50 mg/liter L cysteine
    + 50 mg/liter L tyrosine
    + 25 mg/liter L ascorbic acid
    + 0.062 mg/liter Vitamin B6
    + 1.36 mg/liter Vitamin B12
    + 2 mg/liter Ferric citrate
    + 1 mg/liter Zinc sulphate
    + 0.0025 mg/liter Copper sulphate
    + 50,000 IU/liter Polymyxin
    + 20,000 IU/liter Neomycin
    + 3 μl/liter Ethanolamine
    + 0.16 mg/liter Putrescine
    + 5 mg/liter Recombinant
    Insulin (Nucellin)
  • On day 53 the PEG was replaced with 0.1% pluronic F68. The resulting growth and antibody levels achieved are shown the the attached graphs (FIGS. 4 and 5), and demonstrate the capacity of the invention to allow protein-free production of antibody in excess of 100 μg/ml in fermenters. [0066]
  • EXAMPLE 5 Analysis of the Rate of Campath-1H Synthesis and Secretion from CHO Cells
  • During the course of culturing the Campath-1H producing CHO cells of Example 3, it became clear that even when they reached confluence, antibody levels continued to accumulate, with time, in the culture medium. To determine whether this was possibly a consequence of intracellular accumulation coupled to slow secretion, the rates of Campath-1H synthesis and secretion were measured using A39 cells. These analyses were performed over 3-4 consecutive days on cells which were either in growth phase, or confluent stationary phase. For the cells in either growth state, the results were identical, and data is presented only for the immuno-precipitated radiolabelled Campath-1H produced from stationary cells. [0067]
  • The rate of antibody synthesis was measured by pulsing the cells for a short period with [S[0068] 35]-methionine on each of four consecutive days, and then examining the quantity, and distribution, of immuno-precipitated material. In FIG. 2, it is clear that the rate of synthesis is equally high at all time points measured. Furthermore, even by the end of this short pulse, in each case, more than half of the newly synthesized Campath-1H is already present in the medium suggesting rapid secretion. This was confirmed by the data shown in FIG. 3, in which parallel cells were similarly pulsed, and the distribution of the radiolabelled Campath-1H chased over a three day period. Within 24 hours, virtually all of the cellular radiolabelled antibody has been chased into the medium, where it remained stable for the duration of the experiment. This demonstrates that even when the recombinant CHO cells remain stationary for long periods, the rates of Campath-1H synthesis and secretion are not diminished.
  • Campath-1H ELISA assay. Microtiter plates were coated with anti-human IgG and incubated with the assay sample (in culture medium). Antibody detection was visualized by using an anti-human gamma chain specific peroxidase conjugate. [0069]
  • Analysis of rates of Campath-1H synthesis and secretion. Cells from Example 3 were grown to confluence in 3 cm tissue culture wells, then incubated for 30 minutes in methionine-free Dulbeccos's MEM containing 10% fetal calf serum. Following this, the cells were labelled in the presence of 120 μCi/ml [[0070] 35S] methionine (>800 Ci/mmol: Amersham) for the appropriate time period, then either harvested and lysed in 500 μl of NP-40 lysis buffer, or incubated further in normal growth medium. Then 125 μl aliquots of cell lysate or culture medium were immuno-precipitated using goat anti-human IgG (heavy chain specific; Sigma) and 10% protein-A Sepharose (Pharmacia). Samples were then separated on 10% SDS-PAGE reducing gels according to Laemmli and the signals amplified with Enhance (NEN-Dupont). The dried gels were then autoradiographed overnight.
  • Biological Assays for Functional CHO-[0071] Glycosylated Campath 1H
  • Complement Lysis Assay for [0072] Campath 1H
  • The complement lysis assay is a measure of antibody function expressed as specific activity, determined by the ability of a CHO-glycosylated antibody of known concentration to bind to a pre-determined number of cells and effect cell lysis. [0073]
  • The assay is carried out on [0074] Campath 1H from Example 4 using Karpas 422 cells (established from B-cell non-Hodgkin lymphoma cell line—Dyer et al., (1990) Blood, 75 704-714) expressing Campath antigen on the cell surface. 1.2×107 cells were loaded with radiolabel by incubating for 2 hours at 37° C. in a CO2 incubator in the presence of 600 μCi of 51 Cr (sodium chromate).
  • 5.3 ml of the loaded cells in medium (total volume 23.5 ml), were added to 12.5 ml of normal human serum and 150 μl of the mixture were pipetted into the wells of a microtitre plate. [0075]
  • 50 μl samples of the final eluate from three purification runs were mixed with the cells and incubated for 30 minutes at 4° C. followed by 90 minutes at 37° C. The culture was centrifuged at 2000 rpm for 5 minutes and the radioactivity in 100 μl of cell supernatant was counted on a gamma counter. Complement lysis activity in Kilo Units/ml was calculated from a standard curve of a reference preparation (1000 Units/ml). [0076]
  • The results are set out in Table 5. [0077]
  • The concentration of [0078] Campath 1H in the 50 μl samples of final eluate was estimated using samples in PBS pH 7.2 read on a spectrophotometer at 280 hm. The results are expressed in Table 3 as optical density in mg/ml.
  • From this data the specific activity: KU/ml OD is determined. [0079]
    TABLE 5
    Sample
    Specific Complement lysis Protein Conc
    Activity Kilo Units/ml mg/ml
    A 1.0 11.2 11.1
    B 1.0 14.8 14.2
    C 1.0 13.7 13.6
  • The results indicate that CHO-glycosylated [0080] Campath 1H is functional.
  • Treatment of an Individual with CHO-[0081] Glycosylated Campath 1H
  • An individual diagnosed as having severe T-cell mediated inflammation of the joints (immobilising polyarthritis, pleuritis, abdominal pains) over five years requiring long periods of hospitalisation was treated with CHO derived [0082] Campath 1H from Example 4 using the following regime:
  • 2 mg per day over 6 days by intravenous injection 10 mg per day over subsequent 6 days by intravenous injection. [0083]
  • During the second 6 day treatment there was a significant symptomatic improvement. By the end of the second period the joint inflammation was much improved and a skin abscess had cleared with antibiotic treatment. Thirty days after the end of the treatment the individual was discharged. [0084]
  • Approximately 9 months after the initial treatment, the individual suffered a relapse with multiple joint involvement. After initial testing for sensitivity with a low dose, the individual was given a further course of treatment with 10 mg/[0085] day Campath 1H for 10 days with significant improvement.
  • EXAMPLE 6
  • Expression of Humanised Anti-CD4 Antibody from CHO Cells [0086]
  • Construction of the Expression Vector PBanl: Modification of P342-12 [0087]
  • The complementarily determining regions from a rat IgG2b raised against human CD4 (The New England Journal of Medicine 1990 323: 250-254) were grafted onto human heavy and light chain frameworks (Winter et al, [0088] Nature, 1988, 322 323-327).
  • The cDNA encoding the humanised CD4 light chain was cloned into pLD9 [Page and Sydenham, M. A. 1991 Biotechnology 9 64-68]. The resulting plasmid was designated p2110. The humanised CD4 heavy chain was sequenced and cloned into a modified version of plasmid p342-12 [Law M-F., Byrne, J. C. and Hinley, P. M. 1983 Mol. Cell. Biol. 3 2110-2115). Plasmid p342-12 was digested with BamH1 to remove the 7.4 kbp fragment containing part of the BPV-1 genome. The backbone containing the β-lactamase gene and the neomycin resistance gene under the control of the mouse metallothionine promoter was purified and religated at the BamH1 site. This plasmid was digested with HindIII, incubated with the large fragment of DNA polymerase I to remove the HindIll site and then religated. The β-actin expression cassette, containing the β-actin promoter immediately upstream of a unique HindIII site followed by the polyadenylation signal, was cloned into the BamHI site of the modified p342-12 plasmid to generate pBanl. [0089]
  • Plasmid pBan1, therefore, consisted of the neomycin resistance gene, the β-lactamase gene and the β-actin expression cassette containing the unique HinDIII site. The cDNA encoding the humanised heavy chain was cloned into this site and the resulting plasmid containing the correctly orientated insert was designated pBanCD4H. Thus, p2110 and pBanCD4H contained a different selectable marker and co-transfection into recipient dhfr− CHO cells would permit the direct selection and isolation of dhfr[0090] +/neor colonies. Cells exhibiting this phenotype should express functional antiCD4 antibody and could be amplified to elevate the antibody titres.
  • Expression of Anti-CD4 Antibody in CHO Cells [0091]
  • a) Cell Culture Methods. [0092]
  • The dhfr− CHO line DUK-B11 [Urlaub, G. and Chasin, L. A. 1980 Proc. Natl. Acad. Sci. USA 77 4216-4220] was propagated in Iscoves MEM medium supplemented with 10% foetal bovine serum and 4 μg each of hypoxanthine and thymidine (all Flow). After transfection, transformants were selected in the medium described above except that the hypoxanthine/thymidine were omitted and dialysed foetal bovine serum was used. In addition, G418 was included at 500 μg/ml. To induce spontaneous amplification of sequences containing and flanking the dhfr gene, MTX was added to a concentration of 0.1 μM. [0093]
  • b) Transfection and Amplification [0094]
  • The dhfr− CHO cell line DUK-B11 was co-transfected with 5 μg of p2110 and 5 μg of pBanCD4H using the transfectant reagent under the conditions recommended by the manufacturer. Transformants were selected for the dhfr[0095] +/neor phenotype as described above. Several hundreds of transformants were observed and pooled. Initital titres indicated that the first round basal transformants were secreting about 0.1 μg/ml/day. This pooled population was then cultured in the presence of 0.1 μM MTX for about 14 days. Resistant colonies were again pooled and assayed. Expression had increased some 100 fold, the pooled, amplified colonies producing about 10-12 μg/ml/day. In order to obtain stable, clonal cell lines giving high antibody titres, the resistant pools were cloned by limiting dilution in 96-well plates. Fifty single colonies were identified and assayed and the four lines giving the highest titres propagated. This process of identifying highly expressing clones within the resistant population produced a line designated D419 which expressed the anti-CD4 antibody at about 20 μg/ml/day.
  • Characterisation of dhfr[0096] +/neor Cell Lines
  • i) Determination of copy number and steady state transcription levels by slot blot analysis of DNA and RNA. [0097]
  • Whole cell RNA and DNA was prepared from the various stages of amplification as described by Maniatis et al. [1982 Molecular Cloning. A Laboratory Manual. Cold Spring Harbour Laboratory, Cold Spring Harbour, N.Y.]. After fixing onto nitrocellulose filters, the nucleic acids were probed with [[0098] 32-P]-αATP labelled DNA sequences of the heavy chain, the dhfr gene and the β-actin gene as a control “housekeeping” gene to eliminate artifacts due to loading errors.
  • Inititally, the uncloned 0.1 μM MTX amplified pool was compared to the first round unamplified transformants and the untransformed parental B11 cells, with the probes described. Accordingly, no DNA signal was detected in the parental line when probed with the heavy chain but a weak signal was detected for dhfr. This is due to the single, non-functional dhfr allele in the B11 cell line. As a result, no RNA signal was detected with either probe. In contrast, a strong signal was detected with both probes on RNA and DNA in the primary transformants which reflects the start of expression. A very significant increase in copy number and steady state levels of RNA of heavy chain and dhfr is observed in the uncloned amplified pool. This accurately correlates with the observed increase in expression. Steady state levels of β-actin RNA were consistent in all three lines examined. [0099]
  • A similar comparison was made between the four highest expressing cloned cell lines. A strong signal was detected on both the RNA and the DNA blots. However, although the DA19 line was expressing twice as much antibody as a line designated D423, this difference was not in either the copy number or steady state levels of RNA. There are two possible explanations for this observation; the first is that the DNA in the DA19 line has integrated at a site in the genome at which it is under the influence of an enhancer. However, this presumably would be reflected in elevated levels of RNA. The more likely explanation is that in the replication and duplication of the tandem arrays in the line D423, some of the copies of the dhfr/antibody cassette have undergone re-arrangement and are non-functional and truncated. This is not uncommon since the site of integration of heterologous genes is often at breakpoints in the chromosomes such as telomeres which are known to be “hot spots” for such re-arrangements. This could be resolved by Northern and Southern analysis. [0100]
  • ii) Protein synthesis and secretion of anti-CD4 antibody in the D419 line [0101]
  • The clonal D419 line was labelled with [0102] 35S-methionine and cysteine and the intracellular and secreted antibody extracted by immunoprecipitation with appropriate antibodies. Following electrophoresis on reducing SDS-PAGE gels, the gels were dried and the signal detected by autoradiography.
  • It was clear from the result that both heavy and light chain are efficiently synthesised. Intracellularly, there need not be sto chiometry between heavy and light chains since the two associate as they pass through the secretory organelles. However, close stochiometry is observed in the secreted material. [0103]

Claims (38)

1. An immunotherapy method of treating a human suffering from a disease or disorder, which method comprises the steps of:
(i) constructing a first recombinant expression vector encoding a light chain of a therapeutically effective antibody and constructing a second expression vector encoding a heavy chain of said therapeutically effective antibody;
(ii) introducing said vectors of step (i) into a Chinese hamster ovary (CHO) cell;
(iii) culturing said CHO cell in a culture medium so that said light and heavy chains are produced and a CHO glycosylated therapeutically effective recombinant antibody is thereby produced;
(iv) recovering said therapeutically effective recombinant antibody of step (iii);
(v) administering the recombinant antibody of step (iv) in a therapeutically effective amount to said human.
2. The method of claim 1 wherein the recombinant antibody is a human, chimeric, CDR-grafted or bi-specific antibody.
3. The method of claim 1 wherein the human is afflicted with a T-cell disorder.
4. The method of claim 3 wherein the T-cell disorder is severe vasculitis, rheumatoid arthritis or systemic lupus.
5. The method of claim 1 wherein the human is afflicted with an autoimmune disease.
6. The method of claim 5 wherein the autoimmune disease is multiple sclerosis, graft vs host disease, psoriasis, Juvenile onset diabetes, Sjogrens disease, thyroid disease, myasthenia gravis, transplant rejection or asthma.
7. The method of claim 1 wherein the human is afflicted with cancer.
8. The method of claim 7 wherein the cancer is non-Hodgkins lymphoma or multiple myeloma.
9. The method of claim 1 wherein the human is afflicted with an infectious disease.
10. The method of claim 9 wherein the infectious disease is HIV or herpes.
11. An immunotherapy method of treating a human suffering from a disease or disorder, which method comprises the steps of:
(i) transforming a Chinese hamster ovary (CHO) cell with a recombinant expression vector such that said cell can express a recombinant antibody;
(ii) culturing said CHO cell in serum-free medium so that a CHO glycosylated therapeutically effective recombinant antibody is thereby produced;
(iii) recovering said therapeutically effective recombinant antibody of step (ii);
(iv) administering the recombinant antibody of step (iii) in a therapeutically effective amount to said human.
12. The method of claim 11 wherein the recombinant antibody is a human, chimaeric, CDR-grafted or bi-specific antibody.
13. The method of claim 11 wherein the human is afflicted with a T-cell disorder.
14. The method of claim 13, wherein the T-cell disorder is severe vasculitis, rheumatoid arthritis or systemic lupus.
15. The method of claim 11 wherein the human is afflicted with an autoimmune disease.
16. The method of claim 15 wherein the autoimmune disease is multiple sclerosis, graft vs host disease, psoriasis, Juvenile onset diabetes, Sjogrens disease, thyroid disease, myasthenia gravis, transplant rejection or asthma.
17. The method of claim 11 wherein the human is afflicted with cancer.
18. The method of claim 17 wherein the cancer is non-Hodgkins lymphoma or multiple myeloma.
19. The method of claim 11 wherein the human is afflicted with an infectious disease.
20. The method of claim 19 wherein the infectious disease is HIV or herpes.
21. The method of claim 11 wherein the cell is cultured in said serum-free medium for greater than two months.
22. The method of claim 21 wherein the cell is cultured in said serum free medium for greater than five months.
23. The method of claim 11 wherein the culture undergoes multiple passage.
24. The method of claim 11 wherein the serum free medium comprises water, an osmolarity regulator, a buffer, an energy source, L-glutamine and at least one additional amino acid, an inorganic iron source and a recombinant growth factor wherein each component of said medium is obtained from a source other than directly from an animal source.
25. The medium of claim 11 wherein the medium is devoid of bovine serum albumin, pure human transferrin and soyabean lecithin.
26. The method of claim 11 wherein the growth factor is recombinant insulin.
27. The method of claim 11 wherein the basal medium component of said serum-free medium is an Iscove modification of DMEM.
28. The method of claim 11 wherein the serum-free medium consists essentially of the medium of WCM4.
29. The method of claim 11 wherein the serum-free medium consists essentially of the medium of WCM 5.
30. The method of claim 11 wherein the serium-free medium consists of the medium WCM 4.
31. The method of claim 11 wherein the serum-free medium consists of the medium WCM 5.
32. An immunotherapy method of treating a human suffering from a disease or disorder, which method comprises the steps of:
administering to a human suffering from a disease or disorder, a therapeutically effective amount of a therapeutically effective recombinant human antibody or recombinant altered antibody, said recombinant antibody comprising two light chains and two heavy chains and having an Fc region and a Fab region, wherein said recombinant antibody is expressed in a Chinese hamster ovary (CHO) cell and glycosylated in said Fc region by said CHO cell.
33. The method of claim 32, wherein said antibody has complement lysis activity in an in vitro assay.
34. The method of claim 32, wherein when said antibody is bound to a target cell in said human, the cell to which it is bound is lysed.
35. The method of claim 32, wherein said antibody contains human constant domains.
36. The method of claim 8, wherein the cancer is non-Hodgkins lymphoma.
37. The method of claim 18, wherein the cancer is non-Hodgkins lymphoma.
38. The method of claim 32, wherein the cancer is non-Hodgkins lymphoma.
US10/145,992 1990-10-17 2002-05-16 Glycosylated antibody Abandoned US20030035799A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US10/145,992 US20030035799A1 (en) 1990-10-17 2002-05-16 Glycosylated antibody
US12/122,123 US20080279852A1 (en) 1990-10-17 2008-07-28 Methods of treatment with glycosylated antibodies

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
GB9022543.4 1990-10-17
GB909022543A GB9022543D0 (en) 1990-10-17 1990-10-17 Antibody production
US77773091A 1991-10-16 1991-10-16
US94314692A 1992-09-10 1992-09-10
US4689393A 1993-04-15 1993-04-15
US08/155,864 US5545403A (en) 1990-10-17 1993-11-23 Method for treating a mammal by administering a CHO-glycosylated antibody
US47560795A 1995-06-07 1995-06-07
US49586100A 2000-02-02 2000-02-02
US10/145,992 US20030035799A1 (en) 1990-10-17 2002-05-16 Glycosylated antibody

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US49586100A Continuation 1990-10-17 2000-02-02

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/122,123 Division US20080279852A1 (en) 1990-10-17 2008-07-28 Methods of treatment with glycosylated antibodies

Publications (1)

Publication Number Publication Date
US20030035799A1 true US20030035799A1 (en) 2003-02-20

Family

ID=10683862

Family Applications (7)

Application Number Title Priority Date Filing Date
US08/155,864 Expired - Lifetime US5545403A (en) 1990-10-17 1993-11-23 Method for treating a mammal by administering a CHO-glycosylated antibody
US08/335,400 Expired - Lifetime US5545404A (en) 1990-10-17 1994-11-03 Method for treating a mammal suffering from a T-cell medicated disorder with a CHO-Glycosylated antibody
US08/335,401 Expired - Lifetime US5545405A (en) 1990-10-17 1994-11-03 Method for treating a mammal suffering from cancer with a cho-glycosylated antibody
US10/145,992 Abandoned US20030035799A1 (en) 1990-10-17 2002-05-16 Glycosylated antibody
US10/145,712 Abandoned US20020182208A1 (en) 1990-10-17 2002-05-16 Glycosylated antibody
US10/765,067 Abandoned US20040228857A1 (en) 1990-10-17 2004-01-28 Glycosylated antibody
US12/122,123 Abandoned US20080279852A1 (en) 1990-10-17 2008-07-28 Methods of treatment with glycosylated antibodies

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US08/155,864 Expired - Lifetime US5545403A (en) 1990-10-17 1993-11-23 Method for treating a mammal by administering a CHO-glycosylated antibody
US08/335,400 Expired - Lifetime US5545404A (en) 1990-10-17 1994-11-03 Method for treating a mammal suffering from a T-cell medicated disorder with a CHO-Glycosylated antibody
US08/335,401 Expired - Lifetime US5545405A (en) 1990-10-17 1994-11-03 Method for treating a mammal suffering from cancer with a cho-glycosylated antibody

Family Applications After (3)

Application Number Title Priority Date Filing Date
US10/145,712 Abandoned US20020182208A1 (en) 1990-10-17 2002-05-16 Glycosylated antibody
US10/765,067 Abandoned US20040228857A1 (en) 1990-10-17 2004-01-28 Glycosylated antibody
US12/122,123 Abandoned US20080279852A1 (en) 1990-10-17 2008-07-28 Methods of treatment with glycosylated antibodies

Country Status (13)

Country Link
US (7) US5545403A (en)
EP (5) EP1484402A3 (en)
JP (1) JPH0690752A (en)
AT (2) ATE297988T1 (en)
CA (1) CA2053585C (en)
DE (2) DE69133472T2 (en)
DK (1) DK0481790T3 (en)
ES (2) ES2131507T3 (en)
GB (1) GB9022543D0 (en)
GR (1) GR3030289T3 (en)
IE (1) IE913558A1 (en)
NZ (1) NZ240249A (en)
ZA (1) ZA918248B (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060115474A1 (en) * 1999-07-14 2006-06-01 Jacquemin Marc G Ligands for use in therapeutic compositions for the treatment of hemostasis disorders
US8277805B2 (en) 2003-08-14 2012-10-02 Life Sciences Research Partners Methods for treating or inhibiting thromboembolic disorders or for inhibiting coagulation
EP2971040B1 (en) 2013-03-14 2018-09-19 Momenta Pharmaceuticals, Inc. Methods of cell culture

Families Citing this family (153)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US6893625B1 (en) 1986-10-27 2005-05-17 Royalty Pharma Finance Trust Chimeric antibody with specificity to human B cell surface antigen
IL85035A0 (en) * 1987-01-08 1988-06-30 Int Genetic Eng Polynucleotide molecule,a chimeric antibody with specificity for human b cell surface antigen,a process for the preparation and methods utilizing the same
US7247475B2 (en) 1989-07-28 2007-07-24 Wyeth Method for producing monoclonal antibodies
US6475787B1 (en) 1989-07-28 2002-11-05 Wyeth Method for producing monoclonal antibodies
GB9020282D0 (en) 1990-09-17 1990-10-31 Gorman Scott D Altered antibodies and their preparation
GB9022545D0 (en) 1990-10-17 1990-11-28 Wellcome Found Culture medium
GB9022543D0 (en) * 1990-10-17 1990-11-28 Wellcome Found Antibody production
US6800738B1 (en) * 1991-06-14 2004-10-05 Genentech, Inc. Method for making humanized antibodies
WO1992022653A1 (en) 1991-06-14 1992-12-23 Genentech, Inc. Method for making humanized antibodies
GB9122820D0 (en) * 1991-10-28 1991-12-11 Wellcome Found Stabilised antibodies
GB9125768D0 (en) * 1991-12-04 1992-02-05 Hale Geoffrey Therapeutic method
DE4314556A1 (en) * 1993-05-04 1994-11-10 Behringwerke Ag Modified antibody-enzyme conjugates and fusion proteins and their use in tumor-selective therapy
GB9424449D0 (en) * 1994-12-02 1995-01-18 Wellcome Found Antibodies
JPH11506336A (en) 1995-06-07 1999-06-08 コノート ラボラトリーズ リミテッド Chimeric antibodies for antigen delivery to selected cells of the immune system
CA2223577A1 (en) 1995-06-07 1996-12-19 Connaught Laboratories Limited Chimeric antibodies for delivery of antigens to selected cells of the immune system
CA2257357C (en) * 1996-06-07 2010-04-13 Neorx Corporation Humanized antibodies with modified glycosylation
US6475725B1 (en) 1997-06-20 2002-11-05 Baxter Aktiengesellschaft Recombinant cell clones having increased stability and methods of making and using the same
US20020029391A1 (en) * 1998-04-15 2002-03-07 Claude Geoffrey Davis Epitope-driven human antibody production and gene expression profiling
US20020102208A1 (en) 1999-03-01 2002-08-01 Paul Chinn Radiolabeling kit and binding assay
MY133346A (en) 1999-03-01 2007-11-30 Biogen Inc Kit for radiolabeling ligands with yttrium-90
JP2003521528A (en) * 2000-02-01 2003-07-15 ステヴィア,エイプス Substance used in the preparation of a dietary supplement or a medicament for the treatment of non-insulin dependent diabetes, hypertension and / or metabolic syndrome
US6573096B1 (en) * 2000-04-01 2003-06-03 The Research Foundation At State University Of New York Compositions and methods for inhibition of cancer invasion and angiogenesis
CN101632669B (en) 2000-08-04 2011-05-18 Dmi生物科学公司 Method of using diketopiperazines and composition containing them
AU2002251913A1 (en) * 2001-02-02 2002-08-19 Millennium Pharmaceuticals, Inc. Hybrid antibodies and uses thereof
US6972324B2 (en) 2001-05-18 2005-12-06 Boehringer Ingelheim Pharmaceuticals, Inc. Antibodies specific for CD44v6
GB0123098D0 (en) * 2001-09-26 2001-11-14 Lonza Biologics Plc Use of aminoglycoside resistance gene
US7092957B2 (en) * 2002-01-18 2006-08-15 Boundary Solutions Incorporated Computerized national online parcel-level map data portal
EP2316922B1 (en) 2002-05-24 2013-05-22 Merck Sharp & Dohme Corp. Neutralizing human anti-IGFR antibody
USRE47770E1 (en) 2002-07-18 2019-12-17 Merus N.V. Recombinant production of mixtures of antibodies
ES2442615T5 (en) 2002-07-18 2023-03-16 Merus Nv Recombinant production of antibody mixtures
JP4674317B2 (en) * 2002-10-02 2011-04-20 ディエムアイ アクイジション コーポレイション Disease determination and monitoring
EP2314604A3 (en) 2002-10-15 2011-05-25 Intercell AG Nucleic acids coding for adhesion factors of group B streptococcus, adhesion factors of group B streptococcus and further uses thereof
SI2891666T1 (en) 2002-10-16 2017-11-30 Purdue Pharma L.P. Antibodies that bind cell-associated CA 125/O722P and methods of use thereof
EA013225B1 (en) 2003-01-07 2010-04-30 Симфоген А/С Method for manufacturing cell line producing recombinant polyclonal protein, method for manufacture of polyclonal protein, cell line producing recombinant polyclonal protein, library of vectors, cells population
US7790399B2 (en) 2003-04-03 2010-09-07 Laboratoire Francais du Fractionnement et des Biotechnolgies (LFB) Preparation of human, humanized or chimaeric antibodies or polypeptides having different binding profiles to Fcgamma receptors
WO2004101764A2 (en) 2003-05-13 2004-11-25 Chiron Corporation Methods of modulating metastasis and skeletal related events resulting from metastases
JP4422430B2 (en) 2003-05-14 2010-02-24 帝國製薬株式会社 External patch containing estrogen and / or progestogen
KR20150080004A (en) 2003-05-15 2015-07-08 앰피오 파마슈티컬스 인코퍼레이티드 Treatment of t-cell mediated diseases
ES2408582T3 (en) 2003-05-30 2013-06-21 Merus B.V. Fab library for the preparation of a mixture of antibodies
US20100069614A1 (en) 2008-06-27 2010-03-18 Merus B.V. Antibody producing non-human mammals
CA2544368C (en) 2003-11-04 2014-04-01 Chiron Corporation Methods of therapy for b cell-related cancers
DK1694360T3 (en) 2003-11-04 2010-10-18 Novartis Vaccines & Diagnostic Use of antagonist anti-CD40 antibodies to treat autoimmune and inflammatory diseases and organ transplant rejection
ATE476991T1 (en) 2003-11-04 2010-08-15 Novartis Vaccines & Diagnostic METHOD FOR TREATING SOLID TUMORS WITH EXPRESSION OF CD40 CELL SURFACE ANTIGEN
AU2004287480B2 (en) 2003-11-04 2011-09-15 Novartis Vaccines And Diagnostics, Inc. Use of antagonist anti-CD40 antibodies for treatment of chronic lymphocytic leukemia
PL1684805T3 (en) 2003-11-04 2010-12-31 Novartis Vaccines & Diagnostics Inc Use of antagonist anti-cd40 monoclonal antibodies for treatment of multiple myeloma
JP5912211B2 (en) 2004-01-20 2016-04-27 メルス ビー.ヴィー. Mixture of binding proteins
US8263746B2 (en) 2004-02-06 2012-09-11 Morphosys Ag Anti-CD38 human antibodies and uses thereof
US9200061B2 (en) 2004-02-06 2015-12-01 Morpho Sys AG Generation and profiling of fully human HuCAL gold®-derived therapeutic antibodies specific for human CD3i
ES2426816T3 (en) 2004-08-04 2013-10-25 Mentrik Biotech, Llc Fc regions variants
PT1784426E (en) 2004-09-03 2012-03-06 Genentech Inc Humanized anti-beta7 antagonists and uses therefor
US8137907B2 (en) 2005-01-03 2012-03-20 Cold Spring Harbor Laboratory Orthotopic and genetically tractable non-human animal model for liver cancer and the uses thereof
US7589182B1 (en) 2005-01-07 2009-09-15 Los Alamos National Security, Llc Anti-sulfotyrosine antibodies
WO2006078776A2 (en) * 2005-01-19 2006-07-27 The Trustees Of The University Of Pennsylvania Inhibitors and methods of treatment of cardiovascular diseases, and methods for identifying inhibitors
CA2597924C (en) * 2005-02-15 2018-10-02 Duke University Anti-cd19 antibodies and uses in oncology
US8444973B2 (en) 2005-02-15 2013-05-21 Duke University Anti-CD19 antibodies and uses in B cell disorders
US7595380B2 (en) 2005-04-27 2009-09-29 Tripath Imaging, Inc. Monoclonal antibodies and methods for their use in the detection of cervical disease
AU2006244445B2 (en) * 2005-05-05 2013-04-18 Duke University Anti-CD19 antibody therapy for autoimmune disease
ES2429564T3 (en) 2005-05-18 2013-11-15 Novartis Ag Procedures for the diagnosis and treatment of diseases that have an autoimmune and / or inflammatory component
US20090123950A1 (en) * 2005-05-24 2009-05-14 Morphosys Ag Generation And Profiling Of Fully Human Hucal Gold®-Derived Therapeutic Antibodies Specific For Human CD38
CA2610265A1 (en) 2005-05-31 2007-05-10 Cold Spring Harbor Laboratory Methods for producing micrornas
JP2008546647A (en) * 2005-06-08 2008-12-25 デューク ユニバーシティ Anti-CD19 antibody treatment for transplantation
LT2860192T (en) 2005-10-12 2017-12-11 Morphosys Ag Generation and profiling of fully human HuCAL GOLD-derived therapeutic antibodies specific for human CD38
WO2007046439A1 (en) * 2005-10-18 2007-04-26 National Institute Of Agrobiological Sciences Transgenic silkworm capable of producing antibody and method for production thereof
US7632498B2 (en) 2005-12-19 2009-12-15 Tripath Imaging, Inc. MCM6 and MCM7 monoclonal antibodies and methods for their use in the detection of cervical disease
EP1806365A1 (en) 2006-01-05 2007-07-11 Boehringer Ingelheim International GmbH Antibody molecules specific for fibroblast activation protein and immunoconjugates containing them
US20080014203A1 (en) * 2006-04-11 2008-01-17 Silke Hansen Antibodies against insulin-like growth factor I receptor and uses thereof
TW200813231A (en) 2006-04-13 2008-03-16 Novartis Vaccines & Diagnostic Methods of treating, diagnosing or detecting cancer
US8945564B2 (en) 2006-04-21 2015-02-03 Novartis Ag Antagonist anti-CD40 antibody pharmaceutical compositions
EP3026123A1 (en) 2006-04-27 2016-06-01 Klaritos, Inc. Method and kit for predicting antibody therapy
ES2402591T3 (en) 2006-08-14 2013-05-07 Xencor Inc. Optimized antibodies that target CD19
ATE551071T1 (en) 2006-09-08 2012-04-15 Medimmune Llc HUMANIZED ANTI-CD19 ANTIBODIES AND THEIR USE FOR THE TREATMENT OF CANCER, TRANSPLANTATION AND AUTOIMMUNE DISEASES
NZ575328A (en) 2006-09-13 2012-06-29 Abbott Lab Cell culture improvements
US8911964B2 (en) 2006-09-13 2014-12-16 Abbvie Inc. Fed-batch method of making human anti-TNF-alpha antibody
WO2008043018A1 (en) 2006-10-04 2008-04-10 Dana-Farber Cancer Institute, Inc. Tumor immunity
WO2008112988A2 (en) 2007-03-14 2008-09-18 Novartis Ag Apcdd1 inhibitors for treating, diagnosing or detecting cancer
DE602008002593D1 (en) * 2007-05-25 2010-10-28 Symphogen As METHOD FOR PRODUCING A RECOMBINANT POLYCLONAL PROTEIN
US8394763B2 (en) 2007-09-26 2013-03-12 Oregon Health & Science University Cyclic undecapeptides and derivatives as multiple sclerosis therapies
WO2009146320A1 (en) 2008-05-27 2009-12-03 Dmi Life Sciences, Inc. Therapeutic methods and compounds
WO2010045321A2 (en) * 2008-10-15 2010-04-22 Baxter International Inc. Pegylation of recombinant blood coagulation factors in the presence of bound antibodies
WO2010102167A1 (en) 2009-03-05 2010-09-10 Becton, Dickinson And Company Matrix metalloproteinase-7 (mmp-7) monoclonal antibodies and methods for their use in the detection of ovarian cancer
EP2403875A1 (en) 2009-03-06 2012-01-11 Tripath Imaging, Inc. Glycodelin monoclonal antibodies and methods for their use in the detection of ovarian cancer
CA2755133A1 (en) 2009-03-20 2010-09-23 Amgen Inc. Selective and potent peptide inhibitors of kv1.3
KR20170113704A (en) 2009-05-13 2017-10-12 젠자임 코포레이션 Anti-human cd52 immunoglobulins
CN102438654A (en) 2009-05-13 2012-05-02 基酶有限公司 Methods and compositions for treating lupus
JP4525863B1 (en) 2009-06-05 2010-08-18 東洋紡績株式会社 Expression vector for establishment of high-productivity cells and high-productivity cells
EP2470566A1 (en) 2009-08-24 2012-07-04 St. Jude Children's Research Hospital Compositions and methods for potentiating interleukin-35
WO2011063198A2 (en) 2009-11-20 2011-05-26 St. Jude Children's Research Hospital Methods and compositions for modulating the activity of the interleukin-35 receptor complex
US20120231006A1 (en) 2009-11-20 2012-09-13 Amgen Inc. Anti-orai1 antigen binding proteins and uses thereof
EP2325322A1 (en) * 2009-11-23 2011-05-25 4-Antibody AG Retroviral vector particles and methods for their generation and use
US8383793B2 (en) 2010-04-15 2013-02-26 St. Jude Children's Research Hospital Methods and compositions for the diagnosis and treatment of cancer resistant to anaplastic lymphoma kinase (ALK) kinase inhibitors
MX2013001632A (en) 2010-08-10 2013-06-05 Amgen Inc Dual function in vitro target binding assay for the detection of neutralizing antibodies against target antibodies.
CA2810844C (en) 2010-09-07 2017-03-21 Dmi Acquisition Corp. Diketopiperazine compositions for the treatment of metabolic syndrome and related conditions
MX360946B (en) 2010-09-22 2018-10-29 Amgen Inc Star Carrier immunoglobulins and uses thereof.
US9005907B2 (en) 2010-10-01 2015-04-14 St. Jude Children's Research Hospital Methods and compositions for typing molecular subgroups of medulloblastoma
WO2012075111A1 (en) 2010-11-30 2012-06-07 Novartis Ag Uses of anti-cd40 antibodies in combination therapy for b cell-related cancers
JP2012125197A (en) * 2010-12-16 2012-07-05 Kyowa Hakko Kirin Co Ltd Method for producing protein
AU2012207366A1 (en) 2011-01-18 2013-07-11 Amgen Inc. Nav1.7 knockout mice and uses thereof
WO2012142233A1 (en) 2011-04-14 2012-10-18 St. Jude Children's Research Hospital Methods and compositions for detecting and modulating a novel mtor complex
US9062106B2 (en) 2011-04-27 2015-06-23 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
RU2608957C2 (en) * 2011-05-26 2017-01-27 Юниверсити Оф Вашингтон Cell and gene methods for improvement of cardiac function
EP2760471B9 (en) 2011-09-30 2017-07-19 Dana-Farber Cancer Institute, Inc. Therapeutic peptides
CA2846464A1 (en) 2011-10-10 2013-04-18 Ampio Pharmaceuticals, Inc. Implantable medical devices with increased immune tolerance, and methods for making and implanting
BR112014007675A2 (en) 2011-10-10 2017-04-18 Ampio Pharmaceuticals Inc degenerative joint disease treatment
CN104203982B (en) 2011-10-28 2018-08-31 特瓦制药澳大利亚私人有限公司 Polypeptide construct and application thereof
EP2771007B1 (en) 2011-10-28 2018-04-04 Ampio Pharmaceuticals, Inc. Treatment of rhinitis
CA2864702A1 (en) 2012-02-22 2013-08-29 Amgen Inc. Autologous mammalian models derived from induced pluripotent stem cells and related methods
US9895437B2 (en) 2012-04-18 2018-02-20 Valneva Austria Gmbh Aluminum compounds for use in therapeutics and vaccines
WO2013158279A1 (en) 2012-04-20 2013-10-24 Abbvie Inc. Protein purification methods to reduce acidic species
PL2838918T3 (en) 2012-04-20 2019-11-29 Merus Nv Methods and means for the production of heterodimeric ig-like molecules
US9181572B2 (en) 2012-04-20 2015-11-10 Abbvie, Inc. Methods to modulate lysine variant distribution
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
US9505833B2 (en) 2012-04-20 2016-11-29 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
WO2013176754A1 (en) 2012-05-24 2013-11-28 Abbvie Inc. Novel purification of antibodies using hydrophobic interaction chromatography
WO2014006217A1 (en) 2012-07-06 2014-01-09 Genmab B.V. Dimeric protein with triple mutations
US9206390B2 (en) 2012-09-02 2015-12-08 Abbvie, Inc. Methods to control protein heterogeneity
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
DK2900694T3 (en) 2012-09-27 2018-11-19 Merus Nv BISPECIFIC IGG ANTIBODIES AS T-CELL ACTIVATORS
DK2722341T3 (en) 2012-10-22 2018-03-12 Fountain Biopharma Inc Interleukin-6 antibodies and their applications
WO2014159579A1 (en) 2013-03-14 2014-10-02 Abbvie Inc. MUTATED ANTI-TNFα ANTIBODIES AND METHODS OF THEIR USE
US9017687B1 (en) 2013-10-18 2015-04-28 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
US9499614B2 (en) 2013-03-14 2016-11-22 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides
AU2014233198B2 (en) 2013-03-15 2019-06-27 Sutter West Bay Hospitals Falz for use as a target for therapies to treat cancer
AR095199A1 (en) 2013-03-15 2015-09-30 Genzyme Corp ANTI-CD52 ANTIBODIES
US9808454B2 (en) 2013-03-15 2017-11-07 Ampio Pharmaceuticals, Inc. Compositions for the mobilization, homing, expansion and differentiation of stem cells and methods of using the same
AU2014228502A1 (en) 2013-03-15 2015-08-20 Dana-Farber Cancer Institute, Inc. Therapeutic peptides
MX370377B (en) 2013-04-29 2019-12-11 Teva Pharmaceuticals Australia Pty Ltd Anti-cd38 antibodies and fusions to attenuated interferon alpha-2b.
US11117975B2 (en) 2013-04-29 2021-09-14 Teva Pharmaceuticals Australia Pty Ltd Anti-CD38 antibodies and fusions to attenuated interferon alpha-2B
US9598667B2 (en) 2013-10-04 2017-03-21 Abbvie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US8946395B1 (en) 2013-10-18 2015-02-03 Abbvie Inc. Purification of proteins using hydrophobic interaction chromatography
US9085618B2 (en) 2013-10-18 2015-07-21 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9181337B2 (en) 2013-10-18 2015-11-10 Abbvie, Inc. Modulated lysine variant species compositions and methods for producing and using the same
WO2015073884A2 (en) 2013-11-15 2015-05-21 Abbvie, Inc. Glycoengineered binding protein compositions
KR20160090904A (en) 2013-12-06 2016-08-01 다나-파버 캔서 인스티튜트 인크. Therapeutic peptides
EP3110849B9 (en) 2014-02-28 2021-04-07 Merus N.V. Antibody that binds erbb-2 and erbb-3
EP3786186A1 (en) 2014-02-28 2021-03-03 Merus N.V. Antibodies that bind egfr and erbb3
US10279021B2 (en) 2014-03-14 2019-05-07 Dana-Faber Cancer Institute, Inc. Vaccine compositions and methods for restoring NKG2D pathway function against cancers
UA119352C2 (en) 2014-05-01 2019-06-10 Тева Фармасьютикалз Острейліа Пті Лтд Combination of lenalidomide or pomalidomide and cd38 antibody-attenuated interferon-alpha constructs, and the use thereof
WO2015179627A1 (en) 2014-05-21 2015-11-26 Dana-Farber Cancer Institute, Inc. Methods for treating cancer with anti bip or anti mica antibodies
AU2015305611B2 (en) 2014-08-18 2020-04-23 Ampio Pharmaceuticals, Inc. Treatment of joint conditions
AU2015332577B2 (en) 2014-10-15 2021-12-23 Amgen Inc. Promoter and regulatory elements for improved expression of heterologous genes in host cells
MX2017005481A (en) 2014-10-29 2017-10-26 Teva Pharmaceuticals Australia Pty Ltd Interferon a2b variants.
WO2016209969A1 (en) 2015-06-22 2016-12-29 Ampio Pharmaceuticals, Inc. Use of low molecular weight fractions of human serum albumin in treating diseases
CA2991880A1 (en) 2015-07-10 2017-01-19 Merus N.V. Human cd3 binding antibody
JP7296728B2 (en) 2015-10-23 2023-06-23 メルス ナムローゼ フェンノートシャップ Binding molecules that suppress cancer growth
WO2018182422A1 (en) 2017-03-31 2018-10-04 Merus N.V. Erbb-2 and erbb3 binding bispecific antibodies for use in the treatment f cells that have an nrg1 fusion gene
KR20200042485A (en) 2017-08-09 2020-04-23 메뤼스 엔.페. Antibodies that bind EGFR and cMET
US20200317783A1 (en) 2017-10-06 2020-10-08 Ono Pharmaceutical Co., Ltd. Bispecific antibody
TWI804572B (en) 2018-02-09 2023-06-11 日商小野藥品工業股份有限公司 Bispecific antibody
TW202102544A (en) 2019-04-04 2021-01-16 日商小野藥品工業股份有限公司 Bispecific antibody
US20220372148A1 (en) 2019-07-05 2022-11-24 Ono Pharmaceutical Co., Ltd. A pharmaceutical composition for treating hematological cancer
EP4008348A4 (en) 2019-07-30 2023-11-15 ONO Pharmaceutical Co., Ltd. Bispecific antibody
US20220332825A1 (en) 2019-08-08 2022-10-20 Ono Pharmaceutical Co., Ltd. Bispecific protein

Citations (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4205126A (en) * 1978-01-01 1980-05-27 Cartaya Oscar A Serum-free cell culture media
US4399216A (en) * 1980-02-25 1983-08-16 The Trustees Of Columbia University Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
US4456748A (en) * 1981-02-23 1984-06-26 Genentech, Inc. Hybrid human leukocyte interferons
US4608337A (en) * 1980-11-07 1986-08-26 The Wistar Institute Human hybridomas and the production of human monoclonal antibodies by human hybridomas
US4657866A (en) * 1982-12-21 1987-04-14 Sudhir Kumar Serum-free, synthetic, completely chemically defined tissue culture media
US4767704A (en) * 1983-10-07 1988-08-30 Columbia University In The City Of New York Protein-free culture medium
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4929706A (en) * 1988-11-02 1990-05-29 W. R. Grace & Co.-Conn. Cell growth enhancers and/or antibody production stimulators comprising chemically modified hydrophilic polyurea-urethane prepolymers and polymers
US5001065A (en) * 1987-05-27 1991-03-19 Cetus Corporation Human cell line and triomas, antibodies, and transformants derived therefrom
US5019499A (en) * 1986-04-14 1991-05-28 Daiichi Seiyaku Co., Ltd. Method of producing peptides by transforming myeloma cells with a recombinant plasmid
US5045468A (en) * 1986-12-12 1991-09-03 Cell Enterprises, Inc. Protein-free culture medium which promotes hybridoma growth
US5063157A (en) * 1988-01-18 1991-11-05 Boehringer Mannheim Gmbh Serum-free culture medium for mammalian cells
US5081235A (en) * 1989-07-26 1992-01-14 City Of Hope Chimeric anti-cea antibody
US5089397A (en) * 1985-02-13 1992-02-18 California Biotechnology Inc. Superior mammalian expression system
US5098833A (en) * 1989-02-23 1992-03-24 Genentech, Inc. DNA sequence encoding a functional domain of a lymphocyte homing receptor
US5116964A (en) * 1989-02-23 1992-05-26 Genentech, Inc. Hybrid immunoglobulins
US5219996A (en) * 1987-09-04 1993-06-15 Celltech Limited Recombinant antibodies and methods for their production in which surface residues are altered to cysteine residues for attachment of effector or receptor molecules
US5225538A (en) * 1989-02-23 1993-07-06 Genentech, Inc. Lymphocyte homing receptor/immunoglobulin fusion proteins
US5232848A (en) * 1987-03-24 1993-08-03 W. R. Grace & Co.-Conn. Basal nutrient medium for cell culture
US5316938A (en) * 1990-10-17 1994-05-31 Burroughs Wellcome Co. Defined media for serum-free tissue culture
US5336603A (en) * 1987-10-02 1994-08-09 Genentech, Inc. CD4 adheson variants
US5417970A (en) * 1988-10-21 1995-05-23 Sanofi Drugs containing a glycosylated interleukin-2
US5460811A (en) * 1980-09-25 1995-10-24 Genentech, Inc. Mature human fibroblast interferon
US5500362A (en) * 1987-01-08 1996-03-19 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
US5545405A (en) * 1990-10-17 1996-08-13 Burroughs Wellcome Co. Method for treating a mammal suffering from cancer with a cho-glycosylated antibody
US5605689A (en) * 1991-05-31 1997-02-25 Genentech, Inc. Treatment of HIV-associated immune thrombocytopenic purpura
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
US5846534A (en) * 1988-02-12 1998-12-08 British Technology Group Limited Antibodies to the antigen campath-1
US5876961A (en) * 1991-07-15 1999-03-02 Glaxo Wellcome Inc. Production of antibodies
US5877293A (en) * 1990-07-05 1999-03-02 Celltech Therapeutics Limited CDR grafted anti-CEA antibodies and their production
US5985279A (en) * 1991-07-16 1999-11-16 Waldmann; Herman Humanized antibody against CD18
US6010902A (en) * 1988-04-04 2000-01-04 Bristol-Meyers Squibb Company Antibody heteroconjugates and bispecific antibodies for use in regulation of lymphocyte activity
US6020153A (en) * 1988-01-05 2000-02-01 Ciba-Geigy Corporation Chimeric antibodies
US6291158B1 (en) * 1989-05-16 2001-09-18 Scripps Research Institute Method for tapping the immunological repertoire

Family Cites Families (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1A (en) * 1836-07-13 John Ruggles Locomotive steam-engine for rail and other roads
JPS6147500A (en) * 1984-08-15 1986-03-07 Res Dev Corp Of Japan Chimera monoclonal antibody and its preparation
GB8601597D0 (en) 1986-01-23 1986-02-26 Wilson R H Nucleotide sequences
CA1341235C (en) 1987-07-24 2001-05-22 Randy R. Robinson Modular assembly of antibody genes, antibodies prepared thereby and use
GB8719963D0 (en) * 1987-08-24 1987-09-30 Cattaneo A Recombinant dna products
ZA89430B (en) * 1988-01-22 1989-10-25 Gen Hospital Corp Cloned genes encoding ig-cd4 fusion proteins and the use thereof
AU618989B2 (en) 1988-02-12 1992-01-16 British Technology Group Limited Improvements in or relating to antibodies
US4975369A (en) * 1988-04-21 1990-12-04 Eli Lilly And Company Recombinant and chimeric KS1/4 antibodies directed against a human adenocarcinoma antigen
EP0365209A3 (en) * 1988-10-17 1990-07-25 Becton, Dickinson and Company Anti-leu 3a amino acid sequence
FR2639232A1 (en) * 1988-10-21 1990-05-25 Sanofi Sa DRUGS CONTAINING A GLYCOSYLATED INTERLEUKIN-2
US5530101A (en) * 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
JP2751325B2 (en) * 1989-02-22 1998-05-18 三菱化学株式会社 Method for producing protein
GB8905669D0 (en) * 1989-03-13 1989-04-26 Celltech Ltd Modified antibodies
GB8928874D0 (en) * 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
US5122469A (en) * 1990-10-03 1992-06-16 Genentech, Inc. Method for culturing Chinese hamster ovary cells to improve production of recombinant proteins
CA2120911A1 (en) * 1991-10-15 1993-04-29 Geoffrey Hale Medicaments
GB9125768D0 (en) * 1991-12-04 1992-02-05 Hale Geoffrey Therapeutic method
US5483047A (en) * 1994-03-15 1996-01-09 Inter Bold Automated teller machine
US7775598B2 (en) * 2007-07-03 2010-08-17 Keiper Gmbh & Co. Kg Fitting for a vehicle seat

Patent Citations (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4205126A (en) * 1978-01-01 1980-05-27 Cartaya Oscar A Serum-free cell culture media
US4399216A (en) * 1980-02-25 1983-08-16 The Trustees Of Columbia University Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
US5460811A (en) * 1980-09-25 1995-10-24 Genentech, Inc. Mature human fibroblast interferon
US4608337A (en) * 1980-11-07 1986-08-26 The Wistar Institute Human hybridomas and the production of human monoclonal antibodies by human hybridomas
US4456748A (en) * 1981-02-23 1984-06-26 Genentech, Inc. Hybrid human leukocyte interferons
US4657866A (en) * 1982-12-21 1987-04-14 Sudhir Kumar Serum-free, synthetic, completely chemically defined tissue culture media
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US6331415B1 (en) * 1983-04-08 2001-12-18 Genentech, Inc. Methods of producing immunoglobulins, vectors and transformed host cells for use therein
US4767704A (en) * 1983-10-07 1988-08-30 Columbia University In The City Of New York Protein-free culture medium
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
US5089397A (en) * 1985-02-13 1992-02-18 California Biotechnology Inc. Superior mammalian expression system
US5019499A (en) * 1986-04-14 1991-05-28 Daiichi Seiyaku Co., Ltd. Method of producing peptides by transforming myeloma cells with a recombinant plasmid
US6120767A (en) * 1986-10-27 2000-09-19 Pharmaceutical Royalties, L.L.C. Chimeric antibody with specificity to human B cell surface antigen
US5045468A (en) * 1986-12-12 1991-09-03 Cell Enterprises, Inc. Protein-free culture medium which promotes hybridoma growth
US5721108A (en) * 1987-01-08 1998-02-24 Xoma Corporation 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
US5232848A (en) * 1987-03-24 1993-08-03 W. R. Grace & Co.-Conn. Basal nutrient medium for cell culture
US5001065A (en) * 1987-05-27 1991-03-19 Cetus Corporation Human cell line and triomas, antibodies, and transformants derived therefrom
US5219996A (en) * 1987-09-04 1993-06-15 Celltech Limited Recombinant antibodies and methods for their production in which surface residues are altered to cysteine residues for attachment of effector or receptor molecules
US5336603A (en) * 1987-10-02 1994-08-09 Genentech, Inc. CD4 adheson variants
US6020153A (en) * 1988-01-05 2000-02-01 Ciba-Geigy Corporation Chimeric antibodies
US5063157A (en) * 1988-01-18 1991-11-05 Boehringer Mannheim Gmbh Serum-free culture medium for mammalian cells
US5846534A (en) * 1988-02-12 1998-12-08 British Technology Group Limited Antibodies to the antigen campath-1
US6010902A (en) * 1988-04-04 2000-01-04 Bristol-Meyers Squibb Company Antibody heteroconjugates and bispecific antibodies for use in regulation of lymphocyte activity
US5417970A (en) * 1988-10-21 1995-05-23 Sanofi Drugs containing a glycosylated interleukin-2
US4929706A (en) * 1988-11-02 1990-05-29 W. R. Grace & Co.-Conn. Cell growth enhancers and/or antibody production stimulators comprising chemically modified hydrophilic polyurea-urethane prepolymers and polymers
US5225538A (en) * 1989-02-23 1993-07-06 Genentech, Inc. Lymphocyte homing receptor/immunoglobulin fusion proteins
US5514582A (en) * 1989-02-23 1996-05-07 Genentech, Inc. Recombinant DNA encoding hybrid immunoglobulins
US5098833A (en) * 1989-02-23 1992-03-24 Genentech, Inc. DNA sequence encoding a functional domain of a lymphocyte homing receptor
US5455165A (en) * 1989-02-23 1995-10-03 Genentech, Inc. Expression vector encoding hybrid immunoglobulins
US5428130A (en) * 1989-02-23 1995-06-27 Genentech, Inc. Hybrid immunoglobulins
US5116964A (en) * 1989-02-23 1992-05-26 Genentech, Inc. Hybrid immunoglobulins
US5135866A (en) * 1989-03-03 1992-08-04 W. R. Grace & Co.-Conn. Very low protein nutrient medium for cell culture
US6291158B1 (en) * 1989-05-16 2001-09-18 Scripps Research Institute Method for tapping the immunological repertoire
US5081235A (en) * 1989-07-26 1992-01-14 City Of Hope Chimeric anti-cea antibody
US5877293A (en) * 1990-07-05 1999-03-02 Celltech Therapeutics Limited CDR grafted anti-CEA antibodies and their production
US5633162A (en) * 1990-10-17 1997-05-27 Glaxo Wellcome Inc. Method for culturing Chinese hamster ovary cells
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
US5545403A (en) * 1990-10-17 1996-08-13 Burroughs Wellcome Co. Method for treating a mammal by administering 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
US5316938A (en) * 1990-10-17 1994-05-31 Burroughs Wellcome Co. Defined media for serum-free tissue culture
US5605689A (en) * 1991-05-31 1997-02-25 Genentech, Inc. Treatment of HIV-associated immune thrombocytopenic purpura
US5876961A (en) * 1991-07-15 1999-03-02 Glaxo Wellcome Inc. Production of antibodies
US5985279A (en) * 1991-07-16 1999-11-16 Waldmann; Herman Humanized antibody against CD18

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060115474A1 (en) * 1999-07-14 2006-06-01 Jacquemin Marc G Ligands for use in therapeutic compositions for the treatment of hemostasis disorders
US7829085B2 (en) 1999-07-14 2010-11-09 Life Sciences Research Partners Vzw Methods of treating hemostasis disorders using antibodies binding the C1 domain of factor VIII
US20100285021A1 (en) * 1999-07-14 2010-11-11 Jacquemin Marc G Ligands for use in therapeutic compositions for the treatment of hemostasis disorders
US8309086B2 (en) 1999-07-14 2012-11-13 Life Sciences Research Partners Vzw Antibodies which bind factor VIII
US8784816B2 (en) 1999-07-14 2014-07-22 Life Sciences Research Partners Vzw Antibodies which bind factor VIII
US8277805B2 (en) 2003-08-14 2012-10-02 Life Sciences Research Partners Methods for treating or inhibiting thromboembolic disorders or for inhibiting coagulation
US8282923B2 (en) 2003-08-14 2012-10-09 Life Sciences Research Partners Antibodies which bind and inhibit blood factor VIII
US8293234B2 (en) 2003-08-14 2012-10-23 Life Sciences Research Partners Antibodies which bind and inhibit blood factor VIII
EP2971040B1 (en) 2013-03-14 2018-09-19 Momenta Pharmaceuticals, Inc. Methods of cell culture

Also Published As

Publication number Publication date
EP1950301A3 (en) 2009-05-27
EP1247865A2 (en) 2002-10-09
EP0481790B1 (en) 1999-02-24
EP1247865B1 (en) 2005-06-15
ES2131507T3 (en) 1999-08-01
DE69133472D1 (en) 2005-07-21
EP1950301A2 (en) 2008-07-30
ATE297988T1 (en) 2005-07-15
ZA918248B (en) 1993-04-16
EP0481790A3 (en) 1993-04-14
GB9022543D0 (en) 1990-11-28
DE69130912T2 (en) 1999-09-02
DE69133472T2 (en) 2006-05-11
US20020182208A1 (en) 2002-12-05
AU8591491A (en) 1992-04-30
US5545403A (en) 1996-08-13
US20080279852A1 (en) 2008-11-13
EP0822255A2 (en) 1998-02-04
US5545404A (en) 1996-08-13
US20040228857A1 (en) 2004-11-18
DE69130912D1 (en) 1999-04-01
US5545405A (en) 1996-08-13
NZ240249A (en) 1994-03-25
CA2053585C (en) 2006-08-29
ES2242822T3 (en) 2005-11-16
ATE176926T1 (en) 1999-03-15
JPH0690752A (en) 1994-04-05
GR3030289T3 (en) 1999-09-30
EP1484402A2 (en) 2004-12-08
DK0481790T3 (en) 1999-09-27
EP0481790A2 (en) 1992-04-22
EP1484402A3 (en) 2005-12-21
IE913558A1 (en) 1992-04-22
CA2053585A1 (en) 1992-04-18
EP0822255A3 (en) 1999-10-27
EP1247865A3 (en) 2003-06-04
AU645355B2 (en) 1994-01-13

Similar Documents

Publication Publication Date Title
US5545404A (en) Method for treating a mammal suffering from a T-cell medicated disorder with a CHO-Glycosylated antibody
ES2242823T3 (en) AEROSOL COMPOSITION PREPARATION PROCEDURE.
US5644036A (en) Purified immunoglobulin
Wright et al. Effect of altered CH2-associated carbohydrate structure on the functional properties and in vivo fate of chimeric mouse-human immunoglobulin G1.
KR100191152B1 (en) Cd4 specific recombinant antibody
EP0918797B2 (en) Humanized immunoglobulin reactive with alpha4beta7 integrin
KR100842132B1 (en) Remedies for infant chronic arthritis-relating diseases
EP4186926A1 (en) Ccr8 antibody and application thereof
US20190247498A1 (en) Neutralizing anti-tl1a monoclonal antibodies
IE84931B1 (en) Purified IgG Antibodies
EA015009B1 (en) Antigen binding molecules with increased fc receptor binding affinity and effector function
JP2004000249A (en) Preparation of antibody
EA013677B1 (en) Human monoclonal antibodies against cd25 and use thereof
MX2011008611A (en) Humanized anti-cd20 antibodies and methods of use.
US5786176A (en) Recombinant CDw52 antigen
CN115768477A (en) Antibody specifically recognizing CD40 and application thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: SMITHKLINE BEECHAM CORPORATION, PENNSYLVANIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PAGE, MARTIN J.;CROWE, JAMES S.;RAPSON, NICHOLAS T.;AND OTHERS;REEL/FRAME:013263/0668;SIGNING DATES FROM 20020801 TO 20020823

STCB Information on status: application discontinuation

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