CA2053585C - Antibody production in chinese hamster ovary cells - Google Patents

Antibody production in chinese hamster ovary cells Download PDF

Info

Publication number
CA2053585C
CA2053585C CA002053585A CA2053585A CA2053585C CA 2053585 C CA2053585 C CA 2053585C CA 002053585 A CA002053585 A CA 002053585A CA 2053585 A CA2053585 A CA 2053585A CA 2053585 C CA2053585 C CA 2053585C
Authority
CA
Canada
Prior art keywords
antibody
cell
cells
immunoglobulin
antibodies
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.)
Expired - Lifetime
Application number
CA002053585A
Other languages
French (fr)
Other versions
CA2053585A1 (en
Inventor
Martin John Page
James Scott Crowe
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.)
Wellcome Foundation Ltd
Original Assignee
Wellcome Foundation Ltd
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=CA2053585(C) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Wellcome Foundation Ltd filed Critical Wellcome Foundation Ltd
Publication of CA2053585A1 publication Critical patent/CA2053585A1/en
Application granted granted Critical
Publication of CA2053585C publication Critical patent/CA2053585C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

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

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

ANTIBODY PRODUCTION IN CHINESE HAMSTER OVARY CELLS
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.
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, 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.
The variable domains of each pair of light and heavy chains form the antigen binding site. They havo 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.
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.
A major step forward occurred in 1975 when Kohler and Milstein (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.
NJBM/LB/30th September, 1991.
- 3 ~ PA1195 Unfortunately, the usefulness of such monoclonal antibodies in a clinical setting can be severely hampered by the development of htunan anti-mouse antibodies - an anti-globulin response - which may interfere with therapy or cause allergic or immune complex hypersensitivity.
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 htunanised or CDR-grafted antibody (Jones et al, Nature, 1986, 321, 522-525; and Riechmann et ~1, Nature, 1988, 332, 323-327). A
human antibody would of course avoid the need for "htunanisation", however cell lines which secrete hwnan 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, Nature, 1988, 332, 323-327; Weidle et al, Gene, 1987, 00, 205-216; Nakatani et al, Bio/Technnlogy, 1989, Z, 805-810; and Gillies et al, BioL echnolo~;x, 1989, 7, 799-804).
NJBM/LB/30th September, 1991.
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, Mol. Cell. Biol., 1985, 5, 1750-1759; and Zettlmeissl et al, Bio/Tehnolo~y, 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 marine antibodies reported to date are low - of the order of 0.01-O.lpg/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.
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 also provides a therapeutically useful CHO cell glycosylated antibody capable of fixing complement for human immunotherapeutic treatment of cancer, severe vasculitis, systemic lupus, an infectious disease or an autoimmune disorder selected from the group consisting of multiple sclerosis, Graft versus host disease (GVHD), psoriasis, juvenile onset diabetes, Sjorgren's disease, thyroid disease, myasthenia gravis, transplant rejection and asthma.

- 4 a-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.

2~~3~8~

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.
The cell line of the invention is preferen.'.ially employed for the production. of altered antibodies most preferably chimaerie antibodies or CDR-grafted antibodies. Particular examples of these include antibodies against T cell markers such as CD2, CD3, CD4, CDS, CD7, CD8, CDlla, CDllb, CD18, CD19, CD25, CD45 and CDw52 and especially CDR
grafted antibodies against the CDw52 antigen, such as Campath-1H
NJBM/LB/30th September, 1991.

~~0~~~~8~
(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.
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~nco-transfection include adenosine deaminase (Kaufman et al, P.N.A.S., 1989, 83, 3136-40) ,~sparagine synthetase (Cartier et. al, Mol.Cel1 Biol., 1987, Z,, 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 (Kane et al, Gene, 1989, 84, 439-446), glutamine synthetase (Bebbington et al, DNA Clonine, Vol III, 1987, Ed. D.M.
Glover, 163-188, IRL Press), xanthine guanine phosphoribosyl transferase (gpt) (Mulligan et a1, Science, 1980, 209, 1422-27), hygromycin B (Santerre et al, Gene, 1984, 30, 147-156), neomycin gene (Southern et al, J. Mol. A~~pl.Genet.., 1982, 1, 327-341), and dihydrofolate reductase (Subramani et al, Mol.Cel1 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, 6418, otherwise known as Geneticin.
NJBM/LB/30th September, 1991.

~0~~5~
- 7 - PA7.195 A second preferred system of selection and amplification is provided by the glutamine synthetase selectable marker or (GS system) which is described in W087/04462. 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 W087/04462.
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. 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).
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 viral 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 suitably promoter is the mouse metallothioneiw 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 NJBM/LB/30th September, 1991.

2~~3~~~
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 prefexred so that their expression is substantially balanced. The regulatory elements may be of viral 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. In one preferred instance where the CHO cell line is a dhfr+ 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 NJBM/LB/30th September, 1991.

20~~~~~
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 xegion 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.
It has now been found that antibody glycosylated by CHO cells maintains antigen binding capability and effector functionality. This has been demonstrated in in vitro complement lysis assays and in vivo in a human patient.
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, chimaericor. CDR-grafted antibodies, including Fab fragments.
NJ~M/LB/30th September, 1991.
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, Sjorgren's 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.
More particularly, the invention provides a method for the production of a pharmaceutical composition comprising an immunoglobulin effective in the therapy of human disease, which immunoglobulin is capable of activating complement, said method comprises the steps o~
(a) culturing in suspension a recombinantly transformed CHO cell in a serum free media which cell secretes into said media, a glycosylated immunoglobulin comprising a light and heavy chain;
(b) recovering said immunoglobulin of step (a);
(c) incorporating said immunoglobulin of step (b) into said composition.

. , -l0a-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 35 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 40 diluent or Garner 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 axe routinely parenteral including intravenous, intramuscular, subcutaneous and intraperitoneal injection or delivery.
The accompanying drawings show:
Figure 1 (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 ~-actin promoter; polyA refers to respectively sourced polyadenylation and termination signals; the small box with on contains the SV40 origin of replication;
(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, EcoRl.
Figure 2 Comparative determinations of the- rate of Campath-1H synthesis in confluent A39 cells over 4 consecutive days. Following the [35SJ
methionine pulse period, equal-aliquots of cells (C) and culture medium (M) were immuno-precipitated and separated by SDS-PAGE. The NJBM/LB/30th September, 1991.
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.
Fi,.rure 3 A pulse-chase experiment to determine the rate of secretion and distribution of radiolabelled Campath-1H in A39 cells. Confluent cells were pulsed with [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 pexiod) and treated as described in-the legend of Figure 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.
F~ure 4 Shows growth of C1H 3D11* 44 in WCMS (protein-free medium) in a 1 litre fermenter measured as cell count/ml over 90 days.
Figure 5 Shows antibody production from C1H 3D* 44 cells in WCMS in a 1 litre fermenter measured as micrograms of antibody/ml over 80 days.
The following Examples are provided,..purely for illustration of the present invention.
ERAMPLE 1:1: Cloni~ of the Heavr~ 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 a , Nature, 1988, 322, 323-327).
These constructs were engineered for expression in the myeloma cell NJBM/LB/30th September, 1991.

_, 20~3~~~
line YO and resulted in yields of Campath-1H of up to 5~g/ml following 10-14 days in culture (Hale et al, Tissue Antigens, 1990, 35, 118-127 and Winter et al, Nature, 1988, 322, 323-327). The myeloma cell line TF57 (Hale et al, ibid,) was used to generate size selected cDNA
fractions of 0.9-l.2kb and 1.4-l.7kb for the light and heavy chain cDNAs respectively. These were used to make EcoRl linkered cDNA
libraries in agtl0. All procedures were as described by Huynh et al (DNA Clonine, Vol I: A Practical Approach, 1984,.G1over,D(Editor), IRL
Press, Oxford). The libraries were screened using (32P] nick translated probes specific for the variable regions to isolate full length eDNA 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 47bp upstream of the stop codon. A Sacl-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 29bp untranslated leader, identical to that of the light chain, using a HindIII-NcoI oligonucleotide pair. At the 3' end, the unique NaeI site l2bp downstream of the stop codon was converted into a HindIII site using linkers.
ERAMPLE 2: Construction of Vectors:
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 by PvuII-HindIII fragment, in which the PvuII site was subsequently converted to a BglII site using linkers. To isolate the human ~-actin polyadenylation and termination signals from pH,BAPr-3-neo, an SphI
site l.4kb 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 Sphl site at position -128 in the. SV40 promoter in pSV2dhfr (Subramani et al, Mol.Cell.Biol., 1981, 1, NJBM/LB/30th September, 1991.
854-864) was converted into a Sall 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 (Melton 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 ~-actin promoter, the HindIII Campath-1H light chain cDNA and the HindIII-BamHI ~-actin polyA/termination signals. To construct pNH316, the construct pdBPV-MMTneo (Law et al, Mal.Cell.Bio1-., 1983, 3, 2110-2115) was di-Bested with ~~BamHI, phnsphatased, 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 Figure 1.
ERAMPLE 3: Expression of Campath-1H in CH0 Cells 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~sg/ml each of hypoxanthine and thymidine. l0ug of pLD9 and pNH316 was ca-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 6418 (Gibco) was included at 500pg/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 O.Spg/ml for non-amplified first round transformants.
Each pooled cell population was then cultured in the presence of 10 7M
MTX, and after two weeks, resistant colonies wars again pooled and NJBM/LB/30th September, 1991.

~a5~~8~
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 6M 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 far a further 4 days, the cell lines A39 and 3D11 were capable of producing up to 200~sg/ml of Campath-1H.
_ TABLE 1 Expression Levels of Camvath-1H using Stepwise Amplification Accumulated Construct Selection stage Campath-1H (pg/ml) pLD9 + pNH316 dhfr+/neo basal pool 0.5 7M MTX amplified pool 18-40 Cell lines A37 and 3D9 40 10 6M-MTX amplified pool 60-90 Cell line A39 i00 Cell line 3D11 150-200 NJBM/LB/30th September, 1991.

2~~~~~~
Legend to Table Cells were allowed to reach confluence in a T-175 tissue culture flask, then re-fed with fresh 50m1 of tissue culture medium and left for a further 4 days. The Campath-1H 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 X
107, The yield from-the 3D11 cell line reflects a productivity of 100~ag/106 cells/day.
The co-transfection vectors~pLD9 and-. pNH316.were further employed to evaluaCe 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 6418 (for neomycin selection) and increasing concentrations of MTX ranging from 3 X 10 9M to 10 ~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.

Expression Levels of Campath-1H using Direct Selection Accumulated Selection (M MTX) dhfr+ colonies Campath-1H (~g/ml) No MTX 500 0.5 NJBM/LB/30th September, 1991.

~~~~5~~
3 X 10 9 4o z 10 ~ - -Legend to Table Colonies at each MTX selection stage were-pooled and assayed as described in the legend of Table 1.
This selection procedure was repeated following another co-transfection of oells, and in this instance, the entire population was selected in medium containing 6418 and 3 X 10 8M MTX. This generated a larger pool of resistant colonies which were subsequently pooled and re-amplified twice more using MTX concentrations of.6 X
10 ~M, then 3 X ZO 6M. 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~cg/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 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 ~rariable 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 avident 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 NJBM/LB/30th September, 1991.
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~cg/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 6M Methotrexate and antib-iotics 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 x 104/ml in WCM4 medium Table 3 + 0.25% peptone + 0.1% polyethylene glycol (PEG) 10,000 + 0.5%
fetal bovine serum(FBS) without methotrexate (MTX).

Formulation for medium WCM4.
Iscoves modification of DMEM without BSA, transferrin and lecithin. Available from GIBCO Ltd., Unit 4, Cowley Mill Td.
Est., Uxbridge UB8 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/litre 200mM L glutamine + 50 mg/litre L proline + 50 mg/litre L threonine + 50 mg/litre L methionine + 50 mg/litre L cysteine + 50 mg/litre L tyrosine + 25 mg. litre ascorbic acid + 0.062 mg.litre vitamin B6 + 1.36 mg.litre vitamin B12 + 0.2 mg/litre lipoic acid + 0.088 mg/litre methyl linoleate + 1~M methotrexate + 1 mg/litre FeS04 + 1 mg/litre ZnS04 + 0.0025 mg/litre CuS04 + 5 mg/litre recombinant insulin (Nucellin) + 50,000 Iu/litre polymyxin + 20,000 Iu/litre neomycin + 0.16 mg/litre putrescine-2 HCL.

Three 25cm2 flasks were up with lOml of cell suspension set +

hypoxa nthine (H), thymidineor HT. These flasks were incubated (T) at 36.5oC in 5~ C02 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 75cm2 flask.
These cells were used to seed a SOOml 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 ~g/ml. It was determined that the gene copy number in these cells did not decrease according to band intensity using Northern blot analysis.
In fermenters, these cells produced antibody in excess of 70~g/ml and regularly achieve levels of l~O~g/ml or more. These cells are denoted C1H 3D11 44.
b) Cells from a) above which had been growing serum-free for over 2 months were transferred to a SGi 1 litre fermenter with a stainless * Trade-mark steel angled paddle turning at 70rpm. The temperature was set at 37oC, d0~ at 10~ and pH control to 7-7.2. The fermenter was seeded on day 0 with 0.22 x 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 02. 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.
- On-day 50 onwards G~CMS-(Table 4)-was used together with:peptone and PEG instead of.WCM~f.

Formulation for Medium WCMS
Iscoves modification of DMEM without BSA, transferrin or lecithin (see Table 3).
+ 5 ml/litre 200mM L glutamine + 50 mg/litre L proline + 50 mg/litre L threonine + 50 mg/litre L methionine + 50 mg/litre L cysteine + 50 mg/litre L tyrosine + 25 mg/litre L ascorbic acid + 0.062 mg.litre Vitamin B6 + 1.36 ~ig.litre Vitamin B12 + 2 mg/litre Ferric citrate + 1 mg/litre zinc_sulphate + 0.0025 mg. lit Copper sulphate + 50,000 IU/litre Polymyxin + 20,000 IU/litre Neomycin + 3 ~al/litre Ethanolamine N,~BM/LB/30th September, 1991.
+ 0,16 mg/litre Putrescine + 5 mg/litre 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~cg/ml in fermenters, EXAMPLE 5: Analysis of tyre 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.
The rate of antibody synthesis was measured by pulsing the cells for a short period with [S35]-methionine on each of four consecutive days, and then ~ examining the quantity, and distribution, of immuno-precipitated material. Tn Figure 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 Figure 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 NJBM/LB/30th September, 1991.
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.
Analysis.o~--rates of Campath-1H synthesis and secretion.-- Ce~ls from Example 3 were grown to confluence in 3cm 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 120pCi/ml [35S) methionine (>800Ci/mmol; Amersham) for the appropriate time period, then either harvested and lysed in 5001 of NP-40*lysis buffer, or incubated further in normal growth medium.
Then 125p1 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 haemmli and the signals amplified with Enhance'~(NEN-Dupont*). The dried gels were then autoradiographed overnight.
Biological assays for functional CHO-g_lyco~lated Campath 1H
Complement lysis assay,for 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.
* Trade-mark 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 x 107 cells were loaded with radiolabel by incubating for 2 hours at 37oC in a C02 incubator in the presence of 600~sCi of 5lCr (sodium chromate).
5.3 ml of the loaded cells in medium (total volume 23.5m1), were added to 12.5m1 of normal human serum and ~.50~1 of the mixture were pipetted into the wells of a microtitre plate.
50q1 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 37oC. The culture was centrifuged at 2000 rpm for 5 minutes and the radioactivity in 100~c1 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).
The results axe set out in Table 5.
The concentration of Campath 1H in the 501 samples of final eluate was estimated using samples in PBS pH 7.2 read on a spectrophotometer at 280nm. The.results are expressed in Table 3 as optical density in mg/ml-.
From this data the specific activity: KU ml is determined.
OD

Sample Complement 1~ Protein ConcSpecific Kilo Units/ml mg(ml Activitv A 11.2 11.1 1.0 B 14.8 14.2 1.0 C 13.7 13.6 1.0 NJBM/LB/30th September, 1991.
- 24 - PA1.195 The results indicate that CHO-glycosylated Campath 1H is functional.
Treatment of an individual with CHO-glycosylated Camypath 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 Campath 1H from Example 4 using the following regime:
2mg per day over 6 days by intravenous injection mg per day over subsequent 6 days by intravenous injection.
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.
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 lOmg/day Campath 1H for 10 days with significant improvement.

Construction of the expression vector ~Banl~ modification of p342-12 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, Nature, 1988, 322 323-327).
NJBM/LB/30th September, 1991.

~~J3~8~
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 BamHl to remove the 7.4kbp 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 BamHl site. This plasmid was digested with HinDIII, incubated with the large fragment of DNA polymerase I to remove the HinDIII site and then religated. The ~-actin expression cassette, containing the ~-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.
Plasmid pBanl, 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. Thus, p2110 and pBanCD4H contained a different selectable marker and ca-transfection into recipient dhfr- CHO cells would permit the direct selection and isolation of dhfr+/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 a) Cell culture methods.
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, NJBM/LB/30th September, 1991.
transformants were selected in the medium described above except that the hypoxanthine/thymidine were omitted and dialysed foetal bovine serum was used. In addition, 6418 was included at 500pg/ml. To induce spontaneous amplification of sequences containing and flanking the dhfr gene, MTX was added to a concentration of O.I~M.
b) Transfection and amplification The dhfr- CHO cell line DUK-B11 was co-transfected with 5~g of p2110 and S~g of pBanCD4H using the transfectam*reagent under the conditions recommended by the manufacturer. Transformants were selected for the dhfr+/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 O.l~g/ml/day. This pooled population was then cultured in the presence of O.luM 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~g/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~g/ml/day.
Characterisation of dhfr+/neor cell lines i) Determination of copy number and steady state transcription levels by slot blot analysis of DNA and RNA.
Whole cell RNA and DNA was prepared from the various stages of amplification as described by Maniatis et al. [1982 Mol_ecu~ar * Trade-mark Cloning. A Laboratory Manual. Cold Spring Harbour Laboratory, Cold Spring Harbour, New York]. After fixing onto nitrocellulose filters, the nucleic acids were probed with [32-P]-oATP 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.
Initially, the uncloned O.IpM 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 an 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 S-actin RNA were consistent in all three lines examined.
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 ~0~~5~~
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.
ii) Protein s~inthesis and secretion of anti-CD4 antibody, in the D419 line The clonal D419 line was labelled with 35S-methionine and cysteine and the intracellular.and secxeted antibody extx~acted_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.
NJBM/LB/30th September, 1991.

Claims (13)

1. A method for the production of a pharmaceutical composition comprising an immunoglobulin effective in the therapy of human disease, which immunoglobulin is capable of activating complement, said method comprises the steps of:
(a) culturing in suspension a recombinantly transformed CHO cell in a serum free media which cell secretes into said media, a glycosylated immunoglobulin comprising a light and heavy chain;
(b) recovering said immunoglobulin of step (a);
(c) incorporating said immunoglobulin of step (b) into said composition.
2. The method of claim 1 wherein said immunoglobulin is an antibody.
3. The method of claim 2 wherein said antibody is a human, chimaeric or CDR - grafted antibody.
4. The method of claim 2 or 3 wherein said antibody binds to a T-cell marker.
5. The method of claim 4 wherein said antibody binds a T-cell marker selected from the group consisting of: CD2, CD3, CD4, CD5, CD7, CD8, CD11a, CD11b, CD18, CD19, CD25, CD45, and CD52.
6. The method of any one of claims 1 to 3 wherein said immunoglobulin is for use in the treatment of a disease or disorder selected from the group consisting from the group consisting of: T-cell mediated disorders, autoimmune disorders, cancer, and infectious diseases.
7. The method of claim 6 wherein said T-cell mediated disorder is selected from the group consisting of: severe vasculitis, rheumatoid arthritis, and systematic lupis.
8. The method of claim 6 wherein said autoimmune disorder is selected from the group consisting of: multiple sclerosis, GVHD, psoriarsis, juvenile onset diabetes, Sjogrens' disease, thyroid disease, myasthenia gravis, transplant rejection, and asthma.
9. The method of claim 6 wherein said cancer is non-Hodgkins lymphoma or multiple myeloma.
10. The method of claim 6 wherein said infectious disease is HIV
or Herpes.
11. The method according to anyone of claims 1 to 10, wherein said composition further comprises a physiologically acceptable diluent.
12. The method of any one of claims 1 to 10, wherein said composition is further lyophilised.
13. The method according to anyone of claims 1 to 12, wherein said cell is transformed with an expression vector encoding said heavy chain and another expression vector encoding said light chain.
CA002053585A 1990-10-17 1991-10-16 Antibody production in chinese hamster ovary cells Expired - Lifetime CA2053585C (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB909022543A GB9022543D0 (en) 1990-10-17 1990-10-17 Antibody production
GB9022543.4 1990-10-17

Publications (2)

Publication Number Publication Date
CA2053585A1 CA2053585A1 (en) 1992-04-18
CA2053585C true CA2053585C (en) 2006-08-29

Family

ID=10683862

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002053585A Expired - Lifetime CA2053585C (en) 1990-10-17 1991-10-16 Antibody production in chinese hamster ovary cells

Country Status (13)

Country Link
US (7) US5545403A (en)
EP (5) EP0822255A3 (en)
JP (1) JPH0690752A (en)
AT (2) ATE297988T1 (en)
CA (1) CA2053585C (en)
DE (2) DE69130912T2 (en)
DK (1) DK0481790T3 (en)
ES (2) ES2242822T3 (en)
GB (1) GB9022543D0 (en)
GR (1) GR3030289T3 (en)
IE (1) IE913558A1 (en)
NZ (1) NZ240249A (en)
ZA (1) ZA918248B (en)

Families Citing this family (156)

* 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
US6475787B1 (en) 1989-07-28 2002-11-05 Wyeth Method for producing monoclonal antibodies
US7247475B2 (en) * 1989-07-28 2007-07-24 Wyeth Method for producing monoclonal antibodies
GB9020282D0 (en) * 1990-09-17 1990-10-31 Gorman Scott D Altered antibodies and their preparation
GB9022543D0 (en) * 1990-10-17 1990-11-28 Wellcome Found Antibody production
GB9022545D0 (en) 1990-10-17 1990-11-28 Wellcome Found Culture medium
US6800738B1 (en) 1991-06-14 2004-10-05 Genentech, Inc. Method for making humanized antibodies
DE69233254T2 (en) 1991-06-14 2004-09-16 Genentech, Inc., South San Francisco Humanized Heregulin antibody
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
ATE395422T1 (en) 1995-06-07 2008-05-15 Sanofi Pasteur Ltd CHIMERIC ANTIBODIES FOR DELIVERY OF ANTIGENS TO SELECTED CELLS OF THE IMMUNE SYSTEM
AU6117896A (en) * 1995-06-07 1996-12-30 Connaught Laboratories Limited Chimeric antibodies for delivery of antigens to selected cel ls of the immune system
EP1378525A3 (en) 1996-06-07 2004-01-14 Neorx Corporation Humanized antibodies that bind to the antigen bound by antibody NR-LU-13 and their use in pretargeting methods
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
MY133346A (en) * 1999-03-01 2007-11-30 Biogen Inc Kit for radiolabeling ligands with yttrium-90
US20020102208A1 (en) 1999-03-01 2002-08-01 Paul Chinn Radiolabeling kit and binding assay
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
WO2001056959A1 (en) * 2000-02-01 2001-08-09 Stevia Aps A substance for the use in a dietary supplementation or for the preparation of a medicament for the treatment of non-insulin dependent diabetes mellitus, hypertension and/or the 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
DK1311269T3 (en) 2000-08-04 2012-03-26 Dmi Biosciences Inc PROCEDURE FOR 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
JP4563171B2 (en) 2002-05-24 2010-10-13 シェーリング コーポレイション Neutralizing human anti-IGFR antibody
PT2314629E (en) 2002-07-18 2014-01-22 Merus B V Recombinant production of mixtures of antibodies
USRE47770E1 (en) 2002-07-18 2019-12-17 Merus N.V. Recombinant production of mixtures of antibodies
ATE520981T1 (en) * 2002-10-02 2011-09-15 Dmi Biosciences Inc DIAGNOSIS AND MONITORING OF DISEASES
AU2003274011B2 (en) 2002-10-15 2010-03-04 Intercell Ag Nucleic acids coding for adhesion factor of group B streptococcus, adhesion factors of group B streptococcus and further uses thereof
HUE034378T2 (en) 2002-10-16 2018-02-28 Purdue Pharma Lp Antibodies that bind cell-associated CA 125/O722P and methods of use thereof
BRPI0406678A (en) 2003-01-07 2005-12-20 Symphogen As Method for producing recombinant polyclonal proteins
AU2004226167A1 (en) 2003-04-03 2004-10-14 Institut National De La Sante Et De La Recherche Medicale (Inserm) Antibodies with enhanced ability to immunomodulate cell functions
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
EP2537524B1 (en) 2003-05-15 2016-07-06 Ampio Pharmaceuticals, Inc. Treatment of T-cell mediated diseases
US20100069614A1 (en) 2008-06-27 2010-03-18 Merus B.V. Antibody producing non-human mammals
ES2408582T3 (en) 2003-05-30 2013-06-21 Merus B.V. Fab library for the preparation of a mixture of antibodies
JP4934796B2 (en) 2003-08-14 2012-05-16 トロンボジェニクス・ナムローゼ・フエンノートシャップ Variable antibody
DK1684869T3 (en) 2003-11-04 2011-10-31 Novartis Vaccines & Diagnostic Method of Therapy for B-Cell-Related Cancer Diseases
ATE476448T1 (en) 2003-11-04 2010-08-15 Novartis Vaccines & Diagnostic USE OF ANTAGONIST ANTI-CD40 ANTIBODIES FOR THE TREATMENT OF AUTOIMMUNE DISEASES AND INFLAMMATORY DISEASES AND ORGAN TRANSPLANT REJECTION
PT1682177E (en) 2003-11-04 2010-11-29 Xoma Technology Lt Use of antagonist anti-cd40 antibodies for treatment of chronic lymphocytic leukemia
CA2544852A1 (en) 2003-11-04 2005-05-19 Chiron Corporation Methods of therapy for solid tumors expressing the cd40 cell-surface antigen
WO2005044855A2 (en) 2003-11-04 2005-05-19 Chiron Corporation Use of antagonist anti-cd40 monoclonal antibodies for treatment of multiple myeloma
EP1737971B1 (en) 2004-01-20 2017-08-16 Merus N.V. Mixtures of binding proteins
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
CA2555185C (en) 2004-02-06 2020-03-24 Morphosys Ag Anti-cd38 human antibodies and uses therefor
JP5055603B2 (en) 2004-08-04 2012-10-24 メントリック・バイオテック・リミテッド・ライアビリティ・カンパニー Mutated Fc region
PL2990422T3 (en) 2004-09-03 2018-11-30 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
US8165517B2 (en) * 2005-01-19 2012-04-24 The Trustees Of The University Of Pennsylvania Methods for identifying inhibitors of vascular injury
US20060280738A1 (en) * 2005-06-08 2006-12-14 Tedder Thomas F Anti-CD19 antibody therapy for transplantation
WO2006089133A2 (en) * 2005-02-15 2006-08-24 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
EP2221316A1 (en) * 2005-05-05 2010-08-25 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
CA2625681C (en) 2005-10-12 2016-08-02 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
AU2007238636A1 (en) 2006-04-13 2007-10-25 Novartis Vaccines & Diagnostics, Inc. Methods of treating, diagnosing or detecting cancer
EA018301B1 (en) 2006-04-21 2013-07-30 Новартис Аг Pharmaceutical compositions comprising antagonist monoclonal anti-cd40 antibody and use thereof
CA2650730A1 (en) 2006-04-27 2007-11-08 Pikamab, Inc. Methods and compositions for antibody therapy
DK2383297T5 (en) 2006-08-14 2022-07-04 Xencor Inc Optimized antibodies directed against CD19
AU2007294575B2 (en) 2006-09-08 2013-06-27 Viela Bio, Inc. Humanized anti-CD19 antibodies and their use in treatment of oncology, transplantation and autoimmune disease
US8911964B2 (en) 2006-09-13 2014-12-16 Abbvie Inc. Fed-batch method of making human anti-TNF-alpha antibody
CA2910619A1 (en) 2006-09-13 2008-03-20 Abbvie Inc. Cell culture improvements
US9492518B2 (en) 2006-10-04 2016-11-15 Dana-Farber Cancer Institute, Inc. Tumor immunity
JP2010521180A (en) 2007-03-14 2010-06-24 ノバルティス アーゲー APCDD1 inhibitor for treating, diagnosing or detecting cancer
DK2152872T3 (en) * 2007-05-25 2010-12-06 Symphogen As Process for producing a recombinant polyclonal protein
WO2009042892A1 (en) 2007-09-26 2009-04-02 Oregon Health & Science University Cyclic undecapeptides and derivatives as multiple sclerosis therapies
US8217047B2 (en) * 2008-05-27 2012-07-10 Dmi Acquisition Corp. Therapeutic methods and compounds
CA2738064C (en) * 2008-10-15 2013-03-19 Baxter International Inc. Pegylation of recombinant blood coagulation factors in the presence of bound antibodies
EP2403880A1 (en) 2009-03-05 2012-01-11 Tripath Imaging, Inc. Matrix metalloproteinase-7 (mmp-7) monoclonal antibodies and methods for their use in the detection of ovarian cancer
AU2010221188A1 (en) 2009-03-06 2011-10-20 Tripath Imaging, Inc. Glycodelin monoclonal antibodies and methods for their use in the detection of ovarian cancer
MX2011009797A (en) 2009-03-20 2012-01-12 Amgen Inc Selective and potent peptide inhibitors of kv1.3.
US9617343B2 (en) 2009-05-13 2017-04-11 Genzyme Corporation Methods and compositions for treating lupus
SG176060A1 (en) 2009-05-13 2011-12-29 Genzyme Corp Anti-human cd52 immunoglobulins
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
WO2011028390A1 (en) 2009-08-24 2011-03-10 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
MX2012005864A (en) 2009-11-20 2012-08-31 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
CN103314296A (en) 2010-08-10 2013-09-18 安姆根有限公司 Dual function in vitro target binding assay for the detection of neutralizing antibodies against target antibodies
EP2613786A4 (en) 2010-09-07 2013-10-23 Dmi Acquisition Corp Treatment of diseases
EP2725034B1 (en) 2010-09-22 2019-04-03 Amgen Inc. Carrier immunoglobulins with no specificity for human tissues 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
US20120185956A1 (en) 2011-01-18 2012-07-19 Gingras Jacinthe Global nav1.7 knockout mice and uses
US20140157443A1 (en) 2011-04-14 2014-06-05 St. Jude Children's Research Hospital Methods and compositions for detecting and modulating a novel mtor complex
EP2702077A2 (en) 2011-04-27 2014-03-05 AbbVie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9868937B2 (en) 2011-05-26 2018-01-16 University Of Washington Cell and gene based methods to improve cardiac function
PE20142361A1 (en) 2011-09-30 2015-01-16 Dana Farber Cancer Inst Inc THERAPEUTIC PEPTIDES
CN103841934A (en) 2011-10-10 2014-06-04 安皮奥制药股份有限公司 Implantable medical devices with increased immune tolerance, and methods for making and implanting
US8980834B2 (en) 2011-10-10 2015-03-17 Ampio Pharmaceuticals, Inc. Treatment of degenerative joint disease
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
BR112014008036A2 (en) 2011-10-28 2017-04-11 Ampio Pharmaceuticals Inc rhinitis treatment
AU2013222188A1 (en) 2012-02-22 2014-09-11 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
HUE045944T2 (en) 2012-04-20 2020-02-28 Merus Nv Methods and means for the production of heterodimeric ig-like molecules
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
US9181572B2 (en) 2012-04-20 2015-11-10 Abbvie, Inc. Methods to modulate lysine variant distribution
WO2013176754A1 (en) 2012-05-24 2013-11-28 Abbvie Inc. Novel purification of antibodies using hydrophobic interaction chromatography
US11180572B2 (en) 2012-07-06 2021-11-23 Genmab B.V. Dimeric protein with triple mutations
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
AU2013309506A1 (en) 2012-09-02 2015-03-12 Abbvie Inc. Methods to control protein heterogeneity
WO2014051433A1 (en) 2012-09-27 2014-04-03 Merus B.V. BISPECIFIC IgG ANTIBODIES AS T CELL ENGAGERS
ES2662020T3 (en) 2012-10-22 2018-04-05 Fountain Biopharma Inc. Antibodies against interleukin-6 and its uses
CA2905010A1 (en) 2013-03-12 2014-09-18 Abbvie Inc. Human antibodies that bind human tnf-alpha and methods of preparing the same
US9499614B2 (en) 2013-03-14 2016-11-22 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides
US8921526B2 (en) 2013-03-14 2014-12-30 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
US9217168B2 (en) 2013-03-14 2015-12-22 Momenta Pharmaceuticals, Inc. Methods of cell culture
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
EA201591559A1 (en) 2013-03-15 2016-03-31 Дана-Фарбер Кэнсер Инститьют, Инк. THERAPEUTIC PEPTIDES
JP6566933B2 (en) 2013-03-15 2019-08-28 サッター ベイ ホスピタルズ FALZ for use as a target for therapy to treat cancer
SG11201508923VA (en) 2013-04-29 2015-11-27 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
EP3052640A2 (en) 2013-10-04 2016-08-10 AbbVie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
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
US8946395B1 (en) 2013-10-18 2015-02-03 Abbvie Inc. Purification of proteins using hydrophobic interaction chromatography
WO2015073884A2 (en) 2013-11-15 2015-05-21 Abbvie, Inc. Glycoengineered binding protein compositions
EP3077504B1 (en) 2013-12-06 2019-08-14 Dana-Farber Cancer Institute, Inc. Therapeutic peptides
SG10201913289TA (en) 2014-02-28 2020-02-27 Merus Nv Antibody that binds erbb-2 and erbb-3
JP6771385B2 (en) 2014-02-28 2020-10-21 メルス ナムローゼ フェンノートシャップ Bispecific antibody and pharmaceutical composition
CA3124243A1 (en) 2014-03-14 2015-09-17 Dana-Farber 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
AU2015264114A1 (en) 2014-05-21 2016-11-03 Dana-Farber Cancer Institute, Inc. Methods for treating cancer with anti BiP or anti MICA antibodies
KR20170045274A (en) 2014-08-18 2017-04-26 앰피오 파마슈티컬스 인코퍼레이티드 Treatment of joint conditions
US10202616B2 (en) 2014-10-15 2019-02-12 Amgen Inc. Promoter and regulatory elements for improved expression of heterologous genes in host cells
KR102639037B1 (en) 2014-10-29 2024-02-20 테바 파마슈티컬즈 오스트레일리아 피티와이 엘티디 INTERFERON α2b 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
WO2017010874A1 (en) 2015-07-10 2017-01-19 Merus N.V. Human cd3 binding antibody
US11939394B2 (en) 2015-10-23 2024-03-26 Merus N.V. Binding molecules that inhibit cancer growth
BR112019020508A2 (en) 2017-03-31 2020-08-04 Merus N.V. bispecific antibodies binding to erbb-2 and erbb3 for use in the treatment of cells that have an nrg1 fusion gene
US11773170B2 (en) 2017-08-09 2023-10-03 Merus N.V. 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
JP6881658B2 (en) 2019-07-05 2021-06-02 小野薬品工業株式会社 Blood cancer treatment with PD-1 / CD3 bispecific protein
KR20220039720A (en) 2019-07-30 2022-03-29 오노 야꾸힝 고교 가부시키가이샤 bispecific antibody
WO2021025140A1 (en) 2019-08-08 2021-02-11 小野薬品工業株式会社 Dual-specific protein

Family Cites Families (53)

* 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
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
FR2490675B1 (en) * 1980-09-25 1985-11-15 Genentech Inc MICROBIAL PRODUCTION OF HUMAN FIBROPLASTER 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
US4767704A (en) * 1983-10-07 1988-08-30 Columbia University In The City Of New York Protein-free culture medium
JPS6147500A (en) * 1984-08-15 1986-03-07 Res Dev Corp Of Japan Chimera monoclonal antibody and its preparation
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
ATE89314T1 (en) * 1985-02-13 1993-05-15 Scios Nova Inc HUMAN METALLOTHIONEIN II PROMOTOR IN MAMMALIAN EXPRESSION SYSTEMS.
GB8601597D0 (en) 1986-01-23 1986-02-26 Wilson R H Nucleotide sequences
CA1297434C (en) * 1986-04-14 1992-03-17 Kenji Murakami Method of producing peptides, recombinant plasmid for use in the same and animal cells transformed with the same
US5045468A (en) * 1986-12-12 1991-09-03 Cell Enterprises, Inc. Protein-free culture medium which promotes hybridoma growth
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
EP0283942B1 (en) * 1987-03-24 1992-05-20 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
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
GB8720833D0 (en) * 1987-09-04 1987-10-14 Celltech Ltd Recombinant dna product
US5336603A (en) * 1987-10-02 1994-08-09 Genentech, Inc. CD4 adheson variants
GB8800077D0 (en) * 1988-01-05 1988-02-10 Ciba Geigy Ag Novel chimeric antibodies
DE3801236A1 (en) * 1988-01-18 1989-07-27 Boehringer Mannheim Gmbh PENTOSANE SULFATE MEDIUM
PT89484B (en) * 1988-01-22 1994-03-31 Gen Hospital Corp GENE CLONED CODES OF FUSAO PROTEINS IG-CD4 AND ITS UTILIZATION
US5846534A (en) * 1988-02-12 1998-12-08 British Technology Group Limited Antibodies to the antigen campath-1
CA1339198C (en) * 1988-02-12 1997-08-05 Gregory Paul Winter 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
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
US5417970A (en) * 1988-10-21 1995-05-23 Sanofi Drugs containing a glycosylated interleukin-2
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
US5225538A (en) * 1989-02-23 1993-07-06 Genentech, Inc. Lymphocyte homing receptor/immunoglobulin fusion proteins
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
US5135866A (en) * 1989-03-03 1992-08-04 W. R. Grace & Co.-Conn. Very low protein nutrient medium for cell culture
GB8905669D0 (en) * 1989-03-13 1989-04-26 Celltech Ltd Modified antibodies
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
GB8928874D0 (en) * 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
GB9014932D0 (en) * 1990-07-05 1990-08-22 Celltech Ltd Recombinant dna product and method
US5122469A (en) * 1990-10-03 1992-06-16 Genentech, Inc. Method for culturing Chinese hamster ovary cells to improve production of recombinant proteins
GB9022545D0 (en) * 1990-10-17 1990-11-28 Wellcome Found Culture medium
GB9022543D0 (en) * 1990-10-17 1990-11-28 Wellcome Found Antibody production
EP0636173B1 (en) * 1991-05-31 1998-09-02 Genentech, Inc. Treatment of hiv-associated immune thrombocytopenic purpura
CA2111858A1 (en) * 1991-07-15 1993-02-04 James S. Crowe Production of antibodies
GB9115364D0 (en) * 1991-07-16 1991-08-28 Wellcome Found Antibody
ATE174515T1 (en) * 1991-10-15 1999-01-15 Btg Int Ltd USE OF A CDW52-SPECIFIC ANTIBODY FOR THE TREATMENT OF T-CELL-MEDIATED JOINT INFLAMMATION
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

Also Published As

Publication number Publication date
EP0822255A2 (en) 1998-02-04
EP1950301A2 (en) 2008-07-30
CA2053585A1 (en) 1992-04-18
EP1484402A2 (en) 2004-12-08
EP0481790B1 (en) 1999-02-24
US20030035799A1 (en) 2003-02-20
DE69130912T2 (en) 1999-09-02
AU645355B2 (en) 1994-01-13
ES2242822T3 (en) 2005-11-16
EP1247865A3 (en) 2003-06-04
IE913558A1 (en) 1992-04-22
ZA918248B (en) 1993-04-16
US5545403A (en) 1996-08-13
EP1247865B1 (en) 2005-06-15
EP0481790A2 (en) 1992-04-22
EP1247865A2 (en) 2002-10-09
DE69133472T2 (en) 2006-05-11
US20040228857A1 (en) 2004-11-18
US20020182208A1 (en) 2002-12-05
EP1950301A3 (en) 2009-05-27
DE69133472D1 (en) 2005-07-21
US5545405A (en) 1996-08-13
GB9022543D0 (en) 1990-11-28
GR3030289T3 (en) 1999-09-30
EP0481790A3 (en) 1993-04-14
ATE297988T1 (en) 2005-07-15
ATE176926T1 (en) 1999-03-15
DK0481790T3 (en) 1999-09-27
DE69130912D1 (en) 1999-04-01
JPH0690752A (en) 1994-04-05
ES2131507T3 (en) 1999-08-01
EP1484402A3 (en) 2005-12-21
US20080279852A1 (en) 2008-11-13
NZ240249A (en) 1994-03-25
AU8591491A (en) 1992-04-30
EP0822255A3 (en) 1999-10-27
US5545404A (en) 1996-08-13

Similar Documents

Publication Publication Date Title
CA2053585C (en) Antibody production in chinese hamster ovary cells
ES2242823T3 (en) AEROSOL COMPOSITION PREPARATION PROCEDURE.
US5644036A (en) Purified immunoglobulin
IE84931B1 (en) Purified IgG Antibodies
JP2004000249A (en) Preparation of antibody
JPH11228600A (en) Improvement in or on antibody
MX2011008611A (en) Humanized anti-cd20 antibodies and methods of use.
US5786176A (en) Recombinant CDw52 antigen
AU649078C (en) Purified CDW52-specific antibodies
CN115768477A (en) Antibody specifically recognizing CD40 and application thereof
WO1994015950A1 (en) Purification of immunoglobulin

Legal Events

Date Code Title Description
EEER Examination request
MKEX Expiry