AU768101B2 - Production of a multimeric protein by cell fusion method - Google Patents

Production of a multimeric protein by cell fusion method Download PDF

Info

Publication number
AU768101B2
AU768101B2 AU72174/01A AU7217401A AU768101B2 AU 768101 B2 AU768101 B2 AU 768101B2 AU 72174/01 A AU72174/01 A AU 72174/01A AU 7217401 A AU7217401 A AU 7217401A AU 768101 B2 AU768101 B2 AU 768101B2
Authority
AU
Australia
Prior art keywords
cell
antibody
cells
heavy chain
light chain
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired
Application number
AU72174/01A
Other versions
AU7217401A (en
Inventor
Claude Geoffrey Davis
Larry Green
Nobuaki Hori
Aya Jakobovits
Richard F. Weber
Krisztina M Zsebo
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.)
Japan Tobacco Inc
Amgen Fremont Inc
Original Assignee
Japan Tobacco Inc
Abgenix Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU49898/97A external-priority patent/AU734800B2/en
Application filed by Japan Tobacco Inc, Abgenix Inc filed Critical Japan Tobacco Inc
Priority to AU72174/01A priority Critical patent/AU768101B2/en
Publication of AU7217401A publication Critical patent/AU7217401A/en
Application granted granted Critical
Publication of AU768101B2 publication Critical patent/AU768101B2/en
Priority to AU2004200848A priority patent/AU2004200848A1/en
Anticipated expiration legal-status Critical
Expired legal-status Critical Current

Links

Description

AUSTRALIA
Patents Act 1990 Japan Tobacco, Inc., Abgenix, Inc.
COMPLETE SPECIFICATION STANDARD PATENT Invention Title:
S
0 S 0 0 Production of a multimeric protein by cell fusion method The following statement is a full description of this invention including the best method of performing it known to us:- PRODUCTION OF A MULTIMERTC PROTEIN BY CELL FUSION METHOD FIELD OF THE INVENTION This invention relates generally to methods for use in gene expression and cell fusion techniques, particularly in the production of multi-component proteins.
BACKGROUND OF THE INVENTION Recombinant DNA techniques have been used for production of heterologous proteins in transformed host cells. Generally, the produced proteins are composed of a single amino acid chain or two chains cleaved from a single polypeptide chain. More recently, multichain proteins such as antibodies have been produced by transforming a single host cell with DNA sequences encoding each of the polypeptide chains and expressing the polypeptide chains in the transformed host cell Patent No.
4,816,397).
The basic immunoglobulin (Ig) structural unit in vertebrate systems is composed of 15 two identical "light" polypeptide chains (approximately 23 kDa), and two identical o*o :"heavy" chains (approximately 53 to 70 kDa). The four chains are joined by disulfide Sbonds in a configuration, and the "tail" portions of the two heavy chains are bound by covalent disulfide linkages when the immunoglobulins are generated either by hybridomas or by B cells.
A schematic of the general antibody structure is shown in Fig. 1. The light and heavy chains are each composed of a variable region at the N-terminal end, and a constant region at the C-terminal end. In the light chain, the variable region (termed "VLJL") is the product of the recombination of a VL gene to a JL gene. In the heavy chain, the variable region (VnDHJ) is the product of recombination of first a DH and a JH gene, followed by a S 25 DNJ to V, recombination. The VLJL and VHDHJH regions of the light and heavy chains, respectively, are associated at the tips of the Y to form the antibody's antigen binding domain and together determine antigen binding specificity.
The (CH) region defines the antibody's isotype, its class or subclass.
Antibodies of different isotypes differ significantly in their effector functions, such as the ability to activate complement, bind to specific receptors (Fc receptors) present on a wide variety of cell types, cross mucosal and placental barriers, and form polymers of the basic four-chain IgG molecule.
Antibodies are categorized into "classes" according to the CH type utilized in the immunoglobulin molecule (IgM, IgG, IgD, IgE, or IgA). There are at least five types of CH genes Cy, C8, Ce, and Ca), and some species (including humans) have multiple CH subtypes CY 1
CY
2 Cy 3 and CY 4 in humans). There are a total of nine CH genes in the haploid genome of humans, eight in mouse and rat, and several fewer in many other species. In contrast, there are normally only two types of light chain constant regions kappa (Kc) and lambda and only one of these constant regions is present in a single light chain protein there is only one possible light chain constant region for every VAJ produced). Each heavy chain class can be associated with either of the light chain classes a CHY region can be present in the same antibody as either a K or X light chain).
A process for the immor talization of B cell clones producing antibodies of a single 15 specificity has been developed involving fusing B cells from the spleen of an immunized mouse with immortal myeloma cells. Single clones of fused cells secreting the desired antibody could then be isolated by drug selection followed by immunoassay. These cells were given the name "hybridoma" and their antibody products termed "monoclonal antibodies." 20. The use of monoclonal antibodies as therapeutic agents for human disease requires the ability to produce large quantities of the desired antibody. One approach to increased production was simply to scale up the culture of hybridoma cells. Although this approach is useful;' it is limited to prod .uction of that antibody originally isolated from the mouse. In the case where a hybridoma, cell produces a high affinity monoclonal antibod. with the desired biological activity, but has a low production rate, the gene encoding the antibody can be isolated and transferred to a different cell with a high production rate.
In some cases it is desirable to retain the specificity of the original monoclonal antibody while altering some of its other properties. For example, a problem with using murine antibodies directly for human therapy is that antibodies produced in murine systems may be recognized as "foreign"e proteins by the human immune system, eliciting a response against the antibodies. A human anti-murine antibody (HAMA) response results in antibody neutralization and clearance and/or potentially serious side-effects associated with the anti-antibody immune response. Such murine-derived antibodies thus have limited therapeutic value.
One approach to reducing the immunogenicity of murine antibodies is to replace the constant domains of the heavy and light chains with the corresponding human constant domains, thus generating human-murine chimeric antibodies. Chimeric antibodies are generally produced by cloning the antibody variable regions and/or constant regions, combining the cloned sequences into a single construct encoding all or a portion of a functional chimeric antibody having the desired variable and constant regions, introducing the construct into a cell capable of expressing antibodies, and selecting cells that stably express the chimeric antibody. Examples of methods using recombinant DNA techniques to produce chimeric antibodies are described in PCT Publication No. WO 86/01533 (Neuberger et and in U.S. Patent Nos. 4,816,567 (Cabilly et al.) and 5,202,238 (Fell et al.).
In another approach, complementarity determining region (CDR)-grafted humanized antibodies have been constructed by transplanting the antigen binding site, S rather than the entire variable domain, from a rodent antibody into a human antibody.
.i Transplantation of the hypervariable regions of an antigen-specific mouse antibody into a human heavy chain gene has been shown to result in an antibody retaining antigenspecificity with greatly reduced immunogenicity in humans (Riechmann et al. (1988) 20 Nature 332:323-327; Caron et al. (1992) J. Exp. Med 176:1191-1195).
Another approach in the production of human antibodies has been the generation of S: human B cell hybridomas. Applications of human B cell hybridoma-produced monoclonal antibodies have promising potential in the treatment of cancer, microbial infections, B cell S immunodeficiencies associated with abnormally low antibody production, and other 25 diseases and disorders of the immune system. Obstacles remain in the development of such human monoclonal antibodies. For example, many human tumor antigens may not be immunogenic in humans and thus it may be difficult to isolate anti-tumor antigen antibody-producing human B cells for hybridoma fusion.
For a given disease indication, one antibody isotype is likely to be greatly preferred over another. The preferred isotype may vary from one indication to the next. For example, to treat cancer it may be desirable that the binding of an antibody to a tumor cell result in killing of a tumor cell. In this case, an IgG1 antibody, which mediates both antibody-dependent cellular cytotoxicity and complement fixation, would be the antibody of choice. Alternatively, for treating an autoimmune disease, it may be important that the antibody only block binding of a ligand to a receptor and not cause cell killing. In this case, an IgG4 or IgG2 antibody would be preferred. Thus, even in a situation where a high affinity, antigen-specific, fuly human antibody has been isolated, it may be desirable to re-engineer that antibody and express the new product in a different cell.
The growing use of phage display technology also points to a need for antibody engineering and expression methodologies. Phage display technology is used for producing libraries of antibody variable domains cloned into bacteria. This allows variable domains of desired specificity to be selected and manipulated in vitro. While bacteria offer a great advantage for selecting and producing antibody fragments, they are not~i capable of producing full-size intact antibodies in native configuration, and it is neces sary to reconstitute fragments selected in bacteria into intact antibodies and express them in eucaryotic cells.
15 SUMMARY OF THE INV =~TON The present invention features a method of producing a multimeric protein from a hybrid cell formed from the fusion of two or more cells, each of which cell is engineered to express one component of the multimeric protein, as well as a method for screening for successful fusion of the cells to produce a desired hybrid cell. The methods of the invention are widely applicable to the production of proteins having two or more components.
In one specific application of the method of the invention, the multimeric protein is an antibody composed of antigen-specific heavy and light chains. DNA encoding the desired heavy chain (or a fragment of the heavy chain) is introduced into a first mammalian host cell, while DNA encoding the desired light chain (or a fragment of the light chain) is introduced into a second mammalian host cell. The first transformed host cell and the second transformed host cell are then combined by cell fusion to form a third cell. Prior to fusion of the first and second cells, the transformed cells may be selected for specifically desired characteristics, high levels of expression. After fusion, the resulting hybrid cell contains and expresses both the DNA encoding the desired heavy chain and the DNA encoding the desired light chain, resulting in production of the multimeric antibody.
In one aspect the invention features the multimeric protein produced by the method of the invention. in one embodiment, the invention includes an antibody produced by the method of the invention.
In another aspect the invention features a method for screening for successful fusion of a first cell containing a first nucleotide sequence encoding a desired antibody heavy chain and a second cell containing a second nucleotide sequence encoding a desired antibody light chain, the method comprising including a nucleotide sequence encoding a first marker gene in the first cell, including a nucleotide sequence encoding a second marker gene in the second cell, fusing the first and second cells to produce a fused cell and assaying for the presence of the first and second marker genes in the fused cell.
One advantage of the method of the invention is that cells expressing a single component of the final multi-component protein can be individually selected for one or more desired characteristics, such as a high rate of production.
Another advantage is that the method generates a cell which produces an antibody at a multiplication high rate through the fusion of two kinds of cells which are each selected prior to fusion for high production of the desired heavy or light chains.
Another advantage is that the final multi-component protein is not expressed until all the cells expressing the individual components of the multi-component protein are fused into a single hybrid cell.
Other aspects, features, and advantages of the invention will become apparent from the following detailed description, and the claims.
BRIEF_ DECiTo FTEFGURES9 Figure 1 is a schematic showing the basic immunoglobulin structure.
Figure 2 is a flow chart showing one embodiment of the method of invention when mammnalian cells are separately transformed with the desired light and heavy chain DNA, then fused to form the hybrid cell expressing both chains.
Figure 3 illustrates a specific embodiment of the invention in which a mammalian cell expressing an irrelevant light chain is transformed with the desired heavy chain DNA, a second mammnalian cell is transformed with the desired light chain DNA, and the desired hybrid cell formed from fusion of the transformed host cells is selected which expresses the desired antibody product.
Figure 4 is a schematic illustrating a specific embodiment of the invention in which DHFR CHO cells are independently transfected with pManuGamma#6, a human heavy chain Ig construct and (ii) pManuKappa#14, a human light chain Ig construct. The independent cell lines are selected, amplified, fused, and selected to yield a hybrid cell containing the human heavy chain Ig construct and the human light chain Ig construct.
Figure 5 is a schematic diagram of a fusion method in accordance with the present invention demonstrating the use of HPRT and LacZ marker genes for the initial determination of the success of a fusion process.
Figure 6 is a schematic diagram of the IgK expression vector (pLS413) and the IgH expression vector (pLS421).
Figure 7 is a graph showing the results of an ELISA assay to determine the IL-8 binding affinity of antibodies produced by the cell fusion products of Example 2. Open circles, F-2 fusion clone; open squares, F-5 fusion clone; open triangles, F-13 fusion clone; open diamonds, F-15 fusion clone; closed bar, F-16 fusion clone; closed stars, F-17 15 fusion clone; closed circles, negative control human IgG antibody (no binding affinity for IL-8); and closed squares, D39.2 anti-Il-8 antibody (starting antibody).
Figure 8 is a schematic representation of a construct used for transfection of cells with heavy or light chain Ig cassettes and selectable marker cassettes.
Figure 9 is a restriction map of the human heavy chain construct H/Pur/pEE12.1 20 containing the puromycin resistance gene.
Figure 10 is a restriction map of the human kappa light chain construct K/Hyg/pEE12.1 containing the hygromycin resistance gene.
DETALED
DESCRIPTION
Before the methods and compositions of the present invention are described and 25 disclosed it is to be understood that this invention is not limited to the particular methods and compositions described as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting since the scope of the present invention will be limited only by the appended claims.
It must be noted that as used in this specification and the appended claims, the singular forms "an" and "the" include plural references unless the context clearly dictates otherwise. Thus, for example, reference to "a DNA sequence" includes a plurality of DNA sequences and different types of DNA sequences.
Unless defined otherwise all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any materials or methods similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods and materials are now described. All publications mentioned herein are incorporated herein by reference fro the purpose of describing and disclosing the particular information for which the publication was cited. The publications discussed above are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the inventor is not entitled to antedate such disclosure by virtue of prior invention.
Any discussion of documents, acts, materials, devices, articles or the like which has been included in the present specification is solely for the purpose of providing a context for the present invention. It is not to be taken as an admission that any or all of these matters form part of the prior art base or were common general knowledge in the field relevant to the present invention as it existed before the priority date of each claim of this application.
Definitions Throughout this specification the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated element, integer or step, or group of elements, integers or steps, but not the exclusion of any other element, integer or step, or group of elements, integers or steps.
.ooo.i o By the term "nucleotide sequence" is meant any DNA fragment of interest which may be introduced into a cell, including an intact gene or fragment of a gene.
When the method of the invention is used to generate an antibody, the nucleotide sequence of interest will be all or part of either the constant region and/or variable region of the light or heavy chains, and may include all, part, or none of the regulatory nucleotide sequences that control expression of the light or heavy chains. The nucleotide sequence of interest for heavy chains includes but is not limited to all or a portion of the V, D, J, and switch regions (including intervening sequences) and flanking sequences. For light chains, the nucleotide sequence of interest includes but is not limited to the V and J regions, and flanking and intervening sequences. The nucleotide sequence may be a naturally occurring sequence, synthetic, or partially natural and partially synthetic. The sequence may also be a non-naturally occurring or modified naturally-occurring sequence. The DNA sequence includes sequences taken 7A from different sources, different species. For example, when the method is used to produce an antibody, the DNA chain may encode a chimeric (for example, humanmouse) immunoglobin chain, or it may be a CDR-grafted DNA sequence having a human immunoglobin sequence with antigen-specific murine CDR sequences. The DNA of the nucleotide sequence may encode a fully human antibody. B-cells obtained from non-human animals immunized with an antigen and also hybridoma, trioma, and quadromas derived from such B-cells can also provide the nucleotide sequence introduced into the host cells. B-cells and hybridomas producing any kind of monoclonal antibody may be used as a source of the nucleotide sequence, including cells producing, for example, fully mouse monoclonal antibodies, fully human monoclonal antibodies, CDR-grafted monoclonal antibodies, chimeric monoclonal antibodies, and F(ab) 2 By the terms "multi-component", "multichain", or "multimeric" protein is meant a protein composed of two or more proteins or polypeptides. The method of the invention is useful for producing a multimeric protein by the fusion of two or more cells each expressing a single component of the multimeric protein. For example, in one embodiment the multi-component protein is an antibody generated from two heavy chains encoded by DNA transfected into a first cell and two light chains encoded by DNA transfected into a second cell, where the final multimeric antibody is produced by a hybrid cell formed from the fusion of the first and second cells. "Multi-component," "multichain," and "multimeric" protein is meant to include any heterodimeric or heterooligomeric protein BMP2/BMP7 heterodimeric osteogenic protein, ICE (interleukin- 15 1 converting protein), receptors of the nucleus retinoid receptors), heterodimeric cell surface receptors T cell receptors), integrins cell adhesion molecules, P,integrins, (see, Hynes, 1987 Cell 48:549-554; Hynes 1992 Cell 60:11-25), tumor necrosis factor (TNF) receptor, and soluble and membrane-bound forms of class I and class II MHC (major histocompatibility complex proteins). Where the multimeric protein 20 is a receptor, "multimeric protein" is meant to encompass soluble and membrane forms of the receptor.
By the term "introducing" a nucleotide sequence into a cell means inserting an exogenous piece of DNA into a cell, including but not limited to transfection or S transduction with a vector, such that all or part of the exogenous nucleotide sequence is 25 stably maintained in the cell, and the resulting transformed cell expresses the introduced nucleotide sequence.
By the term "fusing" or "fusion" of two or more cells is meant a method in which two or more cells are combined to form a single hybrid cell which contains all or part of at least the nucleic acid content of each individual cell. Fusion may be accomplished by any method of combining cells under fuseogenic conditions well known in the art (See, for example, Harlow Lane (1988) in Antibodies, Cold Spring Harbor Press, New York).
Known methods for fusing cells includes by use with polyethylene glycol (PEG) or Sendai virus.
By the term "hybrid cell" is meant a cell formed by combining two or more cells, by fusion. In the method of the invention, hybrid cells are formed from the fusion of one or more transformed cells each expressing a single component of a multimeric protein.
The term "irleat as in, an irrelevant light chain" means a light chain which does not contribute to the binding of the antigen of interest and is not a component of the multimeric protein produced by the hybrid cell of the invention.
By the term "desired" component, desired heavy chain, or desired light chain, is meant an immunoglobulin chain which recognizes the antigen of interest.
Generation of a Hybrid Cell Producing a Heterologous Multimeric Protei n The present invention provides a method for generating a hybrid cell producing a multi-component protein from two or more transformed cells each of which cells produces a single component of the niultimeric protein. This method features several important advantages relative to conventional methods for protein production. For example, the method of the present invention allows separately transformed cells to be individually selected for optimal expression of each component of the multi-component protein.' This selection occurs prior to fusion of cells forming the hybrid cell and prior to production of the final multimeric protein. The method of the invention results in a final multicomponent protein product which is not expressed until a single hybrid cell is produced from the fusion of each cell expressing a component of the final protein product.
Generally, when the multi-component protein to be produced is an antibody, the method of the invention involves generation of a cell expressing a desired heavy chain, generation of a cell expressing a desired light chain, and fusion of the two cells to form a hybrid cell expressing the final antibody protein (Fig. Generation of a cell expressing the desired heavy chain involves the following steps: identifying and cloning and/or synthesizing the gene, gene fragment, or nucleotide sequence encoding the variable segment or antigen-binding sequences of the heavy chain. The nucleotide sequence may be obtained from either a cDNA or genomic source, or synthesized de novo; cloning the nucleotide sequence encoding the desired constant regions of the heavy chain; (3) ligating the variable region with the constant region so that the complete nucleotide sequence can be transcribed and translated to express the desired heavy chain polypeptide; ligating the construct into a vector containing a selectable marker and appropriate gene control regions; amplifying the construct in bacteria; introducing the vector into eukaryotic cells; selecting the cells expressing the selectable marker; and screening the cell supernatants or lysates for the expressed heavy chain. Similarly, a cell expressing a desired light chain construct is generated as outlined above.
Alternatively, the process of generating a cell expressing a desired heavy or light chain may involve construction of a Ig chain DNA sequence containing a signal sequence, the gene, gene fragment, or nucleotide sequence encoding the variable region or antigen-binding sequences, and the nucleotide sequence encoding the desired constant region of the Ig chain, followed by PCR amplification of the Ig construction, insertion of the construct into eukaryotic cells, selecting the cells expressing the selectable marker, and screening the cells for the expressed Ig chain. Optionally, the cells expressing the desired heavy chain or the desired light chain can be further selected for desirable characteristics, such as heavy or light chain production rate or level, ability of the expressed heavy or light chain to combine with another light or heavy chain, respectively, to provide an antibody having a desired antigen binding affinity, and/or other characteristics desirable for heavy or light chain production or function in an antibody.
Transformed cells expressing or capable of expressing the desired component of the multimeric protein are fused by methods known in the art to form a hybrid cell expressing the multimeric protein. When the multimeric protein is an antibody, the DNA sequences encoding the desired immunoglobulin may be composed entirely of sequences originating from a single species, fully human or fully murine, or may be contain sequences originating from more than one species, a human-mouse chimera or CDR-grafted humanized antibody. The hybrid cell produced antibody product may also contain a 25 desired antigen binding site (variable region) linked to a desired constant region. Thus, a specifically designed antibody may be generated with a desired antigenicity combined with the desired isotype.
Prior art methods for independently expressing the light and heavy chains in a single host cells are known, for example, U.S. Patent No. 4,816,397, European *patent application publication No. 88,994, PCT published patent application WO 93/19172, U.S. Patent No. 4,816,567, U.S. Patent No. 4,975,369, U.S. Patent No.
5,202,238, PCT published patent application WO 86/01533, PCT published patent application WO 94/02602, and European published patent application No. 273,889.
Vector constructs The vectors of the invention are recombinant DNA vectors including, but not limited to, plasmids, phages, phagemids, cosmids, viruses, retroviruses, and the like, which insert a nucleotide sequence into a cell.
Methods for introducing an exogenous nucleotide sequence of interest into a cell, including into antibody-producing cells, are known in the art. These methods typically include use of a DNA vector to introduce the nucleotide sequence into the genome or a cell or cells, and then growing the cells to generate a suitable population. Nucleotide sequences may also be introduced directly into a cell by methods known in the art.
In a preferred embodiment, nucleotide sequences are introduced into mammalian cells according to the electroporation transfer procedure described by Neumann et. al.
(1982) EMBO J. 1:841, herein specifically incorporated by reference. In another S 15 preferred embodiment, nucleotide sequences are introduced into mammalian cells according to the liposome-mediated transfer procedure described by Felgner et. al. (1987) PNAS 84:7413, herein specifically incorporated by reference. Lipofection is particularly preferred when the host cells are myeloma cells. Transfection of mammalian cell lines may be accomplished by any of a number of methods known to those skilled in the art, 20 including but not limited to microinjection, CaPO 4 precipitation, RBC ghost fusion, protoplast fusion, and the like.
DNA sequences The nucleotide sequence encoding a component of the desired multi-component protein may be obtained as a cDNA or as a genomic DNA sequence by methods known in 25 the art. For example, messenger RNA coding for a desired component may be isolated from a suitable source employing standard techniques of RNA isolation, and the use of oligo-dT cellulose chromatography to segregate the poly-A mRNA. When the product multi-component protein is an antibody, suitable sources of desired nucleotide sequences may be isolated from mature B cells or a hybridoma culture.
In addition to the nucleotide sequence encoding the desired component of the product multi-component protein, vector constructs can include additional components to facilitate replication in prokaryotic and/or eukaryotic cells, integration of the construct into -11a eukaryotic chromosome, and markers to aid in selection of and/or screening for cells containing the construct the detectable markers and drug resistance genes discussed above for the targeting construct). For eukaryotic expression, the construct should preferably additionally contain a polyadenylation sequence positioned 3' of the gene to be expressed. The polyadenylation signal sequence may be selected from any of a variety of polyadenylation signal sequences known in the art. Preferably, the polyadenylation signal sequence is the SV40 early polyadenylation signal sequence.
Transformation of host cells Antibodies have been expressed in a variety of host cells, including bacterial, yeast, and insect cells. For the production of large, multimeric proteins, mammalian cell expression systems generally provide the highest level of secreted product (Bebbington (1991) Methods: A Companion to Methods Enzymol. 2:136-145). Myeloma cells have been used as fusion partners for splenic cells to generate hybridomas cells expressing antibodies. Transformed myeloma cells may be. used as fusible host cells in the method of 15 the invention.
Host cells Nonlymphoid cells lines have been investigated for use in producing antibodies (Cattaneo Neuberger (1987) EMBO J. 6:2753-2758; Deans et al. (1984) Proc. Natl.
Acad. Sci. 81:1292-1296; Weidle et al. (1987) Gene 51:21-29). The ability of 20 nonlymphoid cell lines to assemble and secrete fully functional antibodies may be exploited for antibody production. For example, Chinese hamster ovary (CHO) cells, Chinese Hamster lung (V79) cells (Elkind et al. (1960) Radiat. Res. 13:556), and COS cells have well-characterized efficient expression systems and have been used for both long-term and transient expression of a variety of proteins (Bebbington (1991) sipra; 25 Rauschenbach et. al. (1995) Eur. J. Pharm. 293:183-90). A method for achieving a high level of expression of DNA sequences encoding a chimeric antibody in transformed NSO myeloma cells has been described (Bebbington et al. (1992) Bio/Technology 10:169-175).
Any mammalian cell line capable of expressing the desired multimeric protein and amenable to fusion is suitable for use in the present invention. For example, where the desired protein is an antibody, the cell line is any mammalian cell capable of expressing a functional antibody. A preferred host cell is a mammalian myeloma cell; most preferably, an non-secreting (NS) myeloma cell a non-secreting (NSO) myeloma). Other -12myeloma cells include mouse derived P3/X63-Ag8.653, P3/NS 1/-Ag4-1(NS-1), P3/X63Ag8.U1 (P3U1), SP2/O-Agl4 (Sp2/O, Sp 2 PAI, FO, and BW5147; rat derived 210RCY3-Ag.2.3; and human derived U-266AR1, GM1500-6TG-A1-2, UC729, CEM- AGR, DIR11, and Selection of transformed cells Detection of transfectants with properly integrated vector sequences can be accomplished in a number of ways, depending on the nature of the integrated sequences.
If the transferred nucleotide sequence includes a selectable marker, the initial screening of the transfected cells is to select those which express the marker. Any of a variety of selectable markers known in the art may be included in the construct, including dihydrofolate reductase (DHFR), guanosine phosphoryl transferase gene (gpt), neomycin resistance gene (Neo), hygromycin resistance gene (Hyg) and hypoxanthine phosphoribosyl transferase (HPRT). For example, when using a drug resistance gene, those transfectants that grow in the selection media containing the drug (which is lethal to cells that do not contain the drug resistance gene) can be identified in the initial screening. It will be e* appreciated that a variety of other positive, as well as negative HSV-TK, cytosine deaminase, and the like), selectable markers that are well known in the art can be utilized in accordance with the present invention for selection of specific cells and transfection or other events. As well, a variety of other marker genes the LacZ reporter gene and the like) can be utilized in similar manners.
After a period of time sufficient to allow selection to occur (in most cases, about 2 S:weeks) the surviving cells are then subjected to a second screening to identify those transfectants which express the desired peptide component of interest. This may be accomplished by, for instance, an immunoassay using antibodies specific for the particular 25 immunoglobulin class.
The protocol for the second screening depends upon the nature of the inserted sequences. For example, where the cell is transformed with a sequence which does not result in a secreted product, selection for the presence of the foreign DNA can be detected by Southern blot using a portion of the exogenous sequence as a probe, or by polymerase chain reaction (PCR) using sequences derived from the exogenous sequence as amplifiers.
The cells having an appropriately integrated sequence can also be identified by detecting expression of a functional product, immuno-detection of the product. Alternatively, -13the expression product can be detected using a bioassay to test for a particular effector function conferred by the exogenous sequence.
Where the first host cell is transfected with DNA encoding heavy chain, the expression of the heavy chain can be tested using any conventional immunological screening method known in the art, for example, ELISA conducted with cell lysate samples (see, for example, Colcher et al. Protein Engineering 1987 1:499-505). The cell can be further selected for additional desirable characteristics such as heavy chain production rate or level, ability of the expressed heavy chain to combine with light chain to provide an antibody of a desired antigen binding affinity, and other characteristics desirable for heavy chain production and heavy chain function in an antibody.
Nonlymphoid cells expressing a desired protein may be transfected in a number of ways known to the art. One example of the method of the invention is described in Example 1 below. A first CHO cell may be transfected with a vector comprising a DNA sequence encoding a desired light chain and a second CHO cell transfected with a vector 15 comprising a DNA sequence encoding a desired heavy chain. Transfected cells are selected and fused. Fused cells are selected for expression of an antibody having the desired light chain Ig and heavy chain Ig.
Similarly, a nonlymphoid cell such as V79 may be used as the host cell in the method outlined above. This example of the method of invention is described in Example 20 2 below.
In one embodiment, a cell expressing an Ig heavy chain gene also expresses an irrelevant Ig light chain gene. In some instances, co-expression of a light chain may be required-for secretion and expression of the Ig heavy chain. Failure of a cell to secrete the heavy chain peptide may make detection of transfectants more difficult since it necessitates 25 assaying the cells themselves by Northern blot analysis or immuno-detection), as opposed to conveniently screening the cell supernatant by ELISA.
In a specific embodiment of the invention, this problem is avoided by transfecting a first host cell expressing an irrelevant light chain with a plasmid bearing the desired heavy chain (Fig. The gene encoding the irrelevant light chain may either be integrated into a chromosome or be present in an episomal vector, such as bovine papilloma virus (BPV) or other episomal vector known in the art. After selection for transformants, expression of -14the heavy chain is easily confirmed by an ELISA assay of the cell lysates for secreted antibody.
Cells expressing the desired heavy chain are then fused with a second cell that has been transfected with the desired light chain under appropriate fuseogenic conditions according to methods well known in the art (see, Harlow Lane, supra). Any combination of cells capable of expressing a desired heavy chain or desired light chain and that can be fused to produce a hybrid cell expressing both heavy and light chains can be used. Thus, the first cell expressing the desired heavy chain) can be of the same or different type as the second cell expressing the desired light chain), the first cell can be a myeloma cell and the second cell can be a non-lymphoid cell. The fusion product cells which are candidates for manufacturing lines will express the desired heavy chain and light chain, but will have lost the irrelevant light chain. During the fusion process, random chromosomes are normally lost. Thus, it is expected that cells lacking the irrelevant Ig light chain will be generated during the fusion process. These hybrid cells can easily be identified by ELISA assay of the supernatants for the presence of the desired chains and absence of the irrelevant chain.
Thus, in one embodiment, the desired light chain of the final antibody product is the K light chain. In such cases, a Ig expressing myeloma cell is transfected with the desired IgH gene. After transfection with a plasmid carrying the desired heavy chain and 20 selection, cells expressing the heavy chain are examined directly for expression of the desired heavy chain, ELISA assay of the supernatants with antibody specific to the heavy chain. The second cell, a non-secreting myeloma cell, is transfected with the K light chain, and transfectants detected through Northern blot analysis or immunodetection with an antibody specific to the K light chain. The cells expressing the light 25 chain can be further selected for desirable characteristics associated with production of a functional light chain, such as light chain production rate or level, ability of the expressed light chain to combine with heavy chain to provide an antibody of a desired antigen binding affinity, and other characteristics desirable for light chain production and heavy chain function in an antibody. The cells are then fused, and the hybrid cell expressing the desired IgH/IgK antibody is selected for the presence of the K light chain and desired heavy chain Cy) and the absence of light chain, by ELISA assay of the culture medium.
When the desired product antibody contains a I light chain, the first cell transfected with DNA encoding the desired heavy chain will express a KC light chain, and final selection of hybrid cells expressing the desired antibody will select for the presence of the A light chain and the absence of the Kc light chain.
In an alternative embodiment of the invention, selection of fused or hybrid cells can be initially determined through the utilization of distinct marker genes in each of the "parental" cells or cell lines. Such technique is shown in Figure 4. There, a parental CHO cell line, that is DHFR, is transfected with a vector (pManu Kappa) that contains the DHFR resistance gene and the hygromycin resistance gene (HYGRO). Another parental CHO cell line, that is DHFR, is transfected with a vector (pManu Gamma) that contains the DHFR resistance gene and the neomycin resistance gene. Each cell line, following transfection, contains distinct selectable markers hygromycin resistance in the first and neomycin/G418 resistance in the second). Thus, upon fusion, resulting "daughter" cells in which fusion has been successful will be resistant to both hygromycin and G418.
The screening technique of the invention is advantageous in that it mitigates the need to determine expression of immunoglobulin molecules in order to determine if a fusion has •been successfully performed.
enscesflypromd Under certain fusion conditions, cells and cell lines can become spontaneously resistant to G418, and, possibly, other selectable markers. Thus, in certain embodiments 20 of the invention, it is preferable to utilize selectable markers to which cells and cell lines are less likely to spontaneously generate resistance. An example of one such marker is the .hypoxanthine phosphoribosyl transferase gene (HPRT) which confers resistance to hypoxanthine aminopterin. Another marker that can be used in tandem with HPRT resistance is the LacZ gene. The LacZ gene is not a selectable marker; but, rather, acts as 25 a marker gene which, when expressed by a cell, stains blue in the presence of 13galactosidase.
Thus, through following a similar scheme as described in connection with Figure 4, a first parental cell line, which is HPRT deficient (such as the P3X, NSO, and NSO-bcl2 myeloma cell lines), is transfected with an antibody gene cassette. The cassette includes, for example, appropriate antibody genes, a gene amplification system, and an HPRT selectable marker. Transfected cells can be selected through HPRT selection and cells producing high levels of antibodies can be picked. A second parental cell line, which is -16- 0:96 sees 09*00 *0 S 00
O
S.
5@
C
also preferably HPRT deficient, is transfected with an antibody gene cassette. The cassette includes, for example, appropriate antibody genes, a gene amplification system, and the LacZ gene. Transfected cells can be selected through staining with P-gal. As will be appreciated, either the first or second parental cell line can include the light chain genes or the heavy chain genes and the other of the first or second parental cell line will contain the other of the light or heavy chain genes. As will also be appreciated, other selectable markers can be included in the cassettes utilized to transfect the cells. Upon fusion of the first and second parental cell lines, successful fusion can be determined through HPRT selection and P-gal staining of daughter cells. Daughter cells can be further selected based upon expression levels of immunoglobulin molecules.
Specific embodiments of this technique is illustrated in Figure 5 in several exemplary schemes. In the Figure, a first parental cell line, exemplified by the myeloma cell line, NSO, which is HPRT deficient, is transfected with a light chain cassette containing a gene amplification system an antibody light chain gene system 15 (VKJKCK), and an HPRT selectable marker (HPRT) (Step A second parental cell line, exemplified by any one of J558L, Ag.1, or NSO, are transfected with a heavy chain cassette containing a gene amplification system an antibody heavy chain gene system (VDHJhy), and the LacZ gene (Step The transfection of J558L cell line is indicated as Step 2a, the transfection of the Ag. cell line is indicated as Step 2b, and the transfection of the NSO cell line is indicated as Step 2c. With respect of each Step 1 and Steps 2a-2c, the success of the transfection can be determined through the use of the selectable marker HPRT in Step 1 and through P-gal staining in connection with each of Steps 2a-2c. Additionally cells can be picked for expression of light chain (Step 1) or heavy chain (Step 2a-2c).
25 Following isolation and generation of parental cell lines incorporating the antibody gene cassettes, fusion between a parental cell line including heavy chain genes and a parental cell line including light chain genes is conducted. Utilizing techniques described herein, the parental cell line resulting from Step 1 is fused with a parental cell line resulting from Steps 2a-2c. This is indicated in the Figure as fusion 1-2a, fusion 1-2b, and fusion 1-2c, which results in fused cells 1-2a, 1-2b, and 1-2c, respectively. Such fused cells can be readily identified through dual marker selection, that is, HPRT selection and *5.S r 0 @0 0 0@0e 0 *00@ e 0S -17- P-gal staining. Cells which have been successfully fused, will be HPRT resistant and will stain positive with P-gal.
As will be appreciated, the parental cell lines utilized in fusions l-2a and 1-2b additionally contain mouse mX and rat yK genes. Thus, daughter cells from fusions 1-2a and 1-2b are preferably selected to ensure that they are mX and yK. Loss of mouse mA genes and rat yK genes will generally occur naturally through recombination events during the fusion process.
EXAMPLES
The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use various constructs and perform the various methods of the present invention and are not intended to limit the scope of what the inventors regard as their invention. Unless indicated otherwise, parts are parts by weight, temperature is in degrees centigrade, and pressure is at or near atmospheric pressure. Efforts have been made to ensure accuracy with respect to numbers used, length of DNA sequences, molecular weights, amounts, particular components, etc.) but some deviations should be accounted for.
Example 1 Generation of Hybrid Cells Containing Light and Heavy g Chains.
The human heavy chain Ig construct (IgH gamma) was ligated into the i. pManugamma#6 vector (Fig. 4; Cell Genesys, Inc., Foster City, CA) containing DHFR 20 and neo marker genes. The human kappa light chain Ig construct was ligated into the pManukappa#14 (Fig. 4; Cell Genesys, Inc.) which contains DHFR and hygromycin Sresistance marker genes. The Ig constructs were derived from a hybridoma which secretes an IL-8 antibody.
Overview of the Cell Fusion Method In general, the experiment proceeds as follows: A first cell is transfected with the pManukappa vector comprising the human kappa light chain transgene, and MTX and hygromycin selection marker genes. A second cell is transfected with the pManugamma vector comprising a human Y 4 heavy chain transgene and Neo and MTX selectable marker genes. After the appropriate selection and amplification, the selected first and second cells are fused to form the hybrid cell of the invention expressing a human antibody.
-18- Cell Transfection Chinese hamster ovary (CHO) cells are transfected by electroporation as follows: DHFR-deficient CHO cells in exponential growth are fed with growth medium 4 hours prior to electroporation [growth medium: DMEM/Ham's F12 (50:50 mixture; JRH BioSciences, Woodland, CA), 10% FBS, 2 mM glutamine, non-essential amino acids (NEAA) plus glycine, hypoxanthine and thymidine Cells are collected, washed in PBS, and resuspended in PBS to a concentration of 5 x 106 cells per 0.8 ml. The cell suspension is aliquoted into 0.4 cm electroporation cuvettes (0.8 ml per cuvette) and 5-20 pg linearized DNA added. The suspension is mixed and left on ice for 10 min. Each cuvette is electroporated at 260 V and 960 Each cuvette is place on ice for 10 min, the cells resuspended in 20 ml growth medium, then plated onto 2 10 cm cell culture plates. After 48 hrs, cells from each culture plate are replated in 10 culture plates in the presence of selective media [DMEM, 4.5 g/l glucose (JRH Biosciences), 10% dialyzed FBS (Life Technologies, Bethesda, MD), 5 mM glutamine, NEAA, 0.6 mg/ml G418].
Selection of Transfectants Cells transfected with the kappa light chain transgene were selected in the presence of methotrexate (MTX) and hygromycin. Cells were plated 48 hr post-electroporation into 10 plates in DHFR selective media [DMEM, 4.5 g/l glucose (JRH Biosciences), dialyzed FBS (Life Technologies, Bethesda, MD), 5 mM glutamine, NEAA, supplemented S 20 with hygromycin (Calbiochem, San Diego, CA) at concentrations ranging from 250-750 14g/ml]. Recombinant protein expression can be increased by DHFR-mediated S. amplification of the transfected gene. Methods for selecting cell lines bearing gene amplifications are known in the art, for example, as described in Ausubel et al.
(1989) Current Protocols in Molecular Biology, John Wiley Sons, New York; such methods generally involve extended culture in medium containing gradually increasing levels of methotrexate.
Heavy chain transfectant CHO cells are selected in the presence of MTX and neomycin following the above described procedures.
Generation of a Hybrid Cell Expressing an Antibody Prior to fusion, PEG/DMSO fusion solution (50% PEG, 10% DMSO in PBS)(Sigma) is placed in a 37oC incubator overnight, and 500-1000 ml incomplete Ham's F12 solution (without FCS) is filtered. At fusion, warm fusion medium and incomplete -19- DMEM/Hams' F12 are placed in a 370C water bath. A water-filled beaker and a 15 ml conical tube filled with incomplete DMEM/Ham's F12 are also placed in the water bath.
Harvested transfected CHO cells are washed once with incomplete DMEM/Ham's F12, pelleted at 1200 rpm, resuspended in incomplete DMEM/Ham's F12, and counted. The kappa light chain and the y 4 transfected cells are mixed in a 1:1 ratio, and centrifuged at 800 x g (2060 rpm). The following fusion steps are followed: add 1 ml PEG/DMSO fusion solution to cells over 1 min period; stir cells gently for 1 min; add 2 ml incomplete DMEM/Ham's F12 over a 2 min period with slow stirring; and add 8 ml of incomplete DMEM/Ham's F12 over a 3 min period with slow stirring. The cells are then centrifuged at room temperature at 400 x g for 5 min (1460 rpm). Selection medium [complete DMEM/Ham's F12 10% FCS 250-750 pg/ml hygromycin 0.6 mg/ml G418] is added to the cell pellet. 10 ml of selection medium are added to the cell pellet; cells are gently stirred to resuspend.
The cells are plated onto 10 cm dishes as dilutions of 1:10, 1:20, and 1:40 in selection medium. The plates are refed with fresh medium every 3 days until clones appear. Clones are picked and transferred to a 96-well plate in selection medium. As will be appreciated, growth of cells to reach confluence, which demonstrates survival of cells through selection with hygromycin and G418, is indicative that the cells contain both the heavy and light chain Ig genes, since hygromycin resistance was contributed by the light 20 chain gene containing parental cells and neomycin resistance was contributed by the heavy chain gene containing parental cells. As such, dual marker selection provides an expedient method to initially determine whether a fusion has been successfully accomplished.
Following such an initial screen, supernatant can be assayed for expression of the desired Santibody as described below. When the wells are confluent, the supernatant may then be 25 assayed for expression of the desired antibody as described below.
Selection for Desired Hybrid Cell Expression of the desired antibody may be assayed by immunological procedures, such as Western blot or immunoprecipitation analysis of hybrid cell extracts, or by immunofluorescence of intact cells (using, the methods described in Ausubel et al.
(1989) supra). The desired antibody can be detected using antibody specific for each component of the desired antibody, antibodies specific to the kappa light chain and y 4 heavy chain.
Confirmation of Desired Characteristics of Antibody Produced by Hybrid Cell After the hybrid cell is produced and antibody production in the cell is confirmed, the hybrid cell is grown under conditions to allow expression of the antibody and secretion of the antibody into the cell culture supernatant. For example, the cells can be grown in roller bottles in selective growth medium (DMEM/Ham's F12 (50:50 mixture), 10% FBS, 2 mM glutamine, non-essential amino acids plus glycine, hypoxanthine and thymidine, plus hygromycin and G418 to provide continued selection for the heavy and light chain constructs in the hybrid cell) for several hours prior to assay. Cell culture supernatant is collected and the antibodies are tested for various desired characteristics, antigen binding affinity preferably antigen binding affinity that is similar to that of the original antibody from which the recombinant antibody is derived) using immunological assays well known in the art ELISA, or competition binding assays).
Example 2. Generation of Hybrid Cells Containing Light and Heavy Ig Chains.
According to the known conventional genetic engineering methods, an IgK expression vector (pLS413) and an IgH expression vector (pLS421) were constructed as shown in Fig. 6. Each of the constructs was introduced into the host cells to prepare IgKproducing cells and IgH-producing cells. The light chain gene and the heavy chain gene were cloned from a hybridoma, D39.2, which produces and anti- human IL-8 antibody.
.i Hybridoma D39.2 was prepared from a mouse engineered to produce human antibody S 20 ((1994) Nature Genetics, 7:13-21).
In the IgK expression vector, the light chain gene encoding the human variable and S. human constant regions was inserted. For class-switching, the gene encoding the constant region of-the heavy chain gene was replaced with the gene encoding the human Cy 4 constant region. Then, in the IgH expression vector, the heavy chain gene encoding the human variable and human constant Cy 4 regions was inserted. The IgK and IgH expression vectors have gpt gene and neomycin resistance gene as selectable markers, respectively. The vectors were digested with Sal I to linearize before electroporation.
IgK producing cell 6-Thioguanine resistant V79 cells (1 x 107) were transfected with 100 Ag of the linearized IgK expression vector by electroporation with pulse strength of 300V/0.4 cm and 960 The cells were cultivated with Minimum Essential Medium (MEM) containing 10 Fetal Bovine Serum (FBS) and HAT (100 pM hypoxanthine, 0.4 tM -21aminopterin, 16 /M thymidine). Fifteen days after the transfection, more than 1000 clones grew. Twenty clones were selected. After 10 to 20 days cultivation, IgK concentration in the medium was measured by ELISA as shown in Table 1. All clones produced IgK except K-17, which did not grow.
Table 1: TK producing cells Clone Concentration (ng/ml) K-l 1,250 K-2 750 K-3 1,250 K-4 1,250 200 K-6 2,000 K-7 1,250 K-8 1,250 K-9 500 250 K-11 200 K-12 1,200 K-13 200 K-14 1,500 K-15 500 K-16 700 K-18 700 K-19 1,000 25 K-20 IgH producing cell 6-thioguanine resistant V79 cells (lxl0 7 were transfected with 100 /g of the linearized IgH expression vector by electroporation with pulse strength of 300V/0.4 cm and 960 The cells were cultivated with MEM containing 10% FBS and G418 (400 30 Fifteen days after the transfection, more than 400 clones grew. Eleven clones were selected. Seven days after the initial culture, a lysate of 1 x 105 cells of each clone was prepared. As the IgH alone is not secreted into the medium, IgH in the lysate was measured by ELISA. Seven out of eleven clones produced IgH as shown in Table 2.
-22- Table 2: TgH producing cells Clone Ig Hng/- xl 5 2 H-1 H-2 H-3 0 H-4 0 H-6 H-7 0 H-8 H-9 0 H-11 12 Fusion process The IgK producing cell, clone K-15 (2 x 10 s) and the IgH producing cell (2 x 10 s were mixed and cultured for one night in a dish (10 cm diameter). After removing the medium, the cells were treated with 4 ml 50% PEG 1500 solution for one minute. The cells were washed with fresh MEM five times to remove PEG completely. The cells were removed by trypsin treatment and were suspended. The cells were transferred onto four 20 plates (10 cm diameter) and were cultivated with MEM containing 10% FBS, HAT (100 AM hypoxanthine, 0.4 AM aminopterin, 16 tM thymidine), and G418 (100 /g/ml) for 15 days. Eighteen clones were selected. After ten to twenty days cultivation, IgG concentration in the medium was measured by ELISA. As shown in Table 3, each fused cell makes IgG except the F-5 clone, which did not grow.
*oo -23- Table 3: Antibody-producing cells (following fusion) Clone Concentraon (ne/ml) F-l 98 F-2 347 F-3 132 F-4 F-6 1,133 F-7 124 F-8 292 F-9 310 97 F-11 264 F-12 290 F-13 408 F-14 193 1,258 F-16 654 F-17 324 F-18 203 Antigen specificity of the IgGs were examined by ELISA. The plate used was coated with human IL-8, and anti-human IgK antibody conjugated with peroxidase was used for detection in the ELISA. As shown in Figure 7, all IgGs were human IL-8 S-specific with similar affinity of the original antibody, D39.2.
Example 3.
25 The following is an example of another fusion process that has been utilized in accordance with the invention: reparation ofcells Prior to fusion, parental cell lines for use in the fusion are grown up and maintained in medium containing DMEM high, 10% FBS, 1% non-essential amino acids, 30 1 pen-strep, and 1 L-glutamine.
On the day prior to fusion, each of the parental cell lines are prepared and split to provide a cell density of approximately 10' cells/ml. On the day of the fusion, cells are counted and the fusion is commenced when, and assuming, that cell count for each of the parental cell lines are within the range of about 1.5-2.5 x 105 cells/ml. Sufficient quantities of each of the parental cell lines to make up 5 x 106 cells each are withdrawn from the cultures and added to a 50ml centrifugation tube and the cells are pelleted at 1200 rpm for approximately 5 minutes. Concurrently with the preparation of the cells, incomplete DMEM, PEG, and double selection media are prewarmed in a 37"C incubator bath. Following pelleting, cells are resuspended in 20 ml incomplete DMEM and pelleted again. Thereafter, the cells are resuspended in 5 ml incomplete DMEM and the two parental cell lines are pooled in a single tube and pelleted again to form a co-pellet containing both of the parental cell lines. The co-pellet is resuspended in 10 ml incomplete DMEM and again pelleted. All of the supernatant is then removed from the co-pellet and the cells are ready for fusion.
Fusion Following removal of all of the supernatant, 1 ml PEG-1500 is added over the course of 1 minute to the co-pellet while stirring. After addition of the PEG is completed, either gentle stirring with a pipet is continued for 1 minute or the suspended co-pellet can be allowed to stand for 1 minute. Thereafter, 10 ml of incomplete DMEM is added to the co-pellet over the course of 5 minutes with slow stirring. The mixture is then centrifuged at about 1200 rpm for 5 minutes and following centrifugation, the supernatant is aspirated off, and 10 ml of complete double selection medium is added and gently stirred into the cells. The cells are then plated at 100 pl/well into 10 96-well microtiter plates and placed into an incubator (370 C with 10% CO) where they are not disturbed for 1 week. After 20 the passage of a week, plates are fed by adding 100 1l of complete double selection medium to each well.
**.Clones surviving selection are isolated and productivity assays conducted in accordance with Example 4. Clones may be further subjected to limited dilution cloning using standard techniques.
Double selection medium is prepared depending upon the marker gene utilized in connection with the parental cell lines. In the majority of our experiments, the selectable xx markers conferring puromycin, hygromycin, or hypoxanthine aminopterine (HAT) resistance are utilized. Concentrations required to obtain complete cell killing of NS/0 cells were determined through use of kill curves and resulted in our use of 6 micrograms/ml of puromycin and 350 micrograms/ml of hygromycin. In connection with HPRT resistance, we used HAT media supplement from Sigma using standard conditions.
ExampleAk In this Example, a productivity assay is provided for the analysis of Ig expression by daughter cells obtained through the fusion process. In the assay, cells are counted and 200,000 are selected and washed with complete DMEM media (containing 10% fetal calf serum, 1 glutamine, 1 nonessential amino acids, and 1 pen-strep). The cells are pelleted (at about 1200 rpm for about 5 minutes) and the supernatant is removed. The cells are resuspended in 2 ml medium and plated in a 6-well plate. The cells are then grown at 370 C with 10% CO 2 in an incubator for 4 days. Thereafter cells are resuspended and counted using a hemacytometer. Cells are pelleted and the supematant is retained for ELISA.
The ELISA is conducted using a human K capture, followed by detection with a polyclonal human anti-IgG. Standards for the ELISA are isotype specific. The ELISA provides a quantitative measurement of the amount of secreted antibody. Through starting with a known number of cells and obtaining the number of cells after four days of growth, we can also estimate the antibody production per cell.
Exampe In this example, the generation of parental cell lines containing either a human heavy chain construct or a human x light chain construct is described.
So* Construct Generation 20 In connection with generation of either parental cell line, the construct shown schematically in Figure 8 was used. For preparation of the human Ig cassette, a human tanti-IL-8 IgG 2 monoclonal antibody producing hybridoma (designated D1. 1) was utilized for source DNA. The D1. 1 hybridoma was derived from a mouse engineered to produce human antibodies as described in Mendez et al. Nature Genetics 15:146-156 (1997) and U.S. Patent Application Serial No. 08/759,620, filed December 3, 1996 (the disclosures of which are incorporated by reference in their entirety) that was immunized with human IL-8.
Each of the heavy chain variable region and the entire K light chain was cloned from the D1. 1 hybridoma using RT-PCR and cDNA obtained. In the heavy chain construct, separately, genomic DNA encoding a human gamma-4 constant region was isolated and cloned and ligated to the cDNA encoding the heavy chain variable region.
-26- Each of the heavy chain construct and the light chain construct were ligated into the vector shown schematically in Figure 8 to form constructs H/pEE12.1 (heavy chain) and K/pEE12.1 (light chain). Such constructs were then fitted with appropriate selectable marker cassettes. For example, puromycin resistance was provided to the heavy chain construct and hygromycin resistance was provided to the light chain construct to form the vectors H/Pur/pEE12.1 and K/Hyg/pEE12.1, respectively, and as shown in the restriction maps provided in Figures 9 and 10, respectively.
Transfection We prepared an NS/0 and NS/O-bcl-2 myeloma cell line containing the human heavy chain construct (H/Pur/pEE12.1) and NS/0 and NS/0-bcl-2 cell lines containing the human K light chain construct (H/Hyg/pEE12.1) through standard transfection techniques.
In connection with the heavy chain construct, transfection was accomplished through linearizing the H/Pur/pEE12.1 construct at the PacI restriction site and electroporating 20 urg of the same into 5 x 106 cells (either NS/0 or NS/0-bcl-2 cells).
Electroporation was accomplished at 300 V, 960 After 10 minutes at room temperature, cells were plated in 10 ml DMEM complete media in P100 plates. In connection with the light chain construct, the same techniques were used, however, the DNA was linearized using the SalI restriction ste.
After 24 hours recovery, viability was assayed with trypan blue. Cells were 20 harvested, pelleted, resuspended in selection medium, and plated at 5,000-10,000 viable cells per well in 96-well plates, either in medium containing puromycin (heavy chain) or hygromycin (light chain). Clones surviving selection were isolated and expanded. Light chain and heavy chain production from cells surviving selection were nfeasured using ELISA and normalized to total protein assayed by Bradford on cell lysates. Heavy chain the ratio of ELISA OD to total protein OD in NS/0 cell lines ranged from about 3.0 to total protein (NS/0-bcl-2 cell lines containing heavy chains were not assayed). Light chain the ratio of ELISA OD to total protein OD in NS/0 cell lines ranged from about 0.5 to 5.99 total protein and NS/O-bcl-2 cell lines ranged from about 0.3 to 6.28. Based upon the above assay, the highest producing single heavy chain containing NS/0 cell line the highest producing light chain containing NS/0 cell line and the highest producing NS/0-bcl-2 cell line (6.28) were selected for use in fusions.
-27- Flxamnle6 In this example, results obtained through use of the fusion process described in connection with Example 3 are provided (Table 4)experiment, a human heavy chain Ig containing NS/O parental cell line was fused with a human ic light chain containing NS/0 parental cell line. Thus, in the experiment, an NS/O cell line containing the heavy chain VDJ gamma-4 cassette and a puromycin resistance cassette was fused with an NS/O cell line containing the human kappa light chain cassette and the hygromycin resistance cassette.
Fusion and selection were accomplished using the procedure described in Example 3 and productivity determined in accordance with Example 4.
Table 4 (1 subcloning) gkI ML pgi Cen Clone g/ml pg/cell 0 0 0eo e:ooeo (2n" ubc .nint 54.2 17.5 14.8 54.2.1 25.3 20.7 54.2.2 28.4 29.6 54.2.3 43.6 43.6 54.2.4 16.6 24.8 54.2.5 5.0 11.4 54.2.6 11.1 15.0 88.5 8.3 8.1 88.5.1 9.7 9.9 88.5.2 10.0 17.8 88.5.3 10.1 15.5 88.5.4 3.5 3.6 88.5.5 16.3 21.4 88.5.6 3.7 3.2 91.2 7.3 5.1 91.2.1 4.1 4.8 91.2.3 3.5 4.4 91.2.4 22.5 28.1 91.2.5 12.1 16.8 91.2.6 6.9 11.9 96.5 7.5 8.3 96.5.1 7.9 10.6 96.5.2 15.7 18.9 96.5.4 6.1 6.1 196.5.5 7.6 7.6 9 fi _f!14 -2 IR -Q Examptc2 In this example, results obtained through use of the fusion process described in connection with Example 3 are provided (Table In the experiment, a human heavy chain Ig containing NS/0 parental cell line was fused with a human K light chain -28containing NS/0-bcl-2 parental cell line. Thus, in the experiment, an NS/0 cell line containing the heavy chain VDJ gamma-4 cassette and a puromycin resistance cassette was fused with an NS/0-bcl-2 cell line containing the human kappa light chain cassette and the hygromycin resistance cassette. Fusion and selection were accomplished using the procedure described in Example 3 and productivity determined in accordance with Example 4.
Table *o go .o Sample ID hIgG/hK Concentration (A/ml) 1 2.2.1 4.75 2 2.2.2 3.18 3 2.2.3 3.57 4 2.2.4 3.19 2.2.5 0.559 6 2.2.6 5.05 7 27.6.3 9.87 8 27.6.4 11.7 9 29.1.3 14.2 29.1.4 4.61 11 29.1.6 12.9 12 23B3 __2 Example R In this example, results obtained through use of the fusion process described in connection with Example 3 are provided (Table In the experiment, a human heavy chain Ig.containing NS/0 parental cell line was fused with either a human Kc light chain containing NS/0-bcl-2 parental cell line (Clone ID's 17, 19, and 20) or a human K, light chain containing NS/0 parental cell line (Clone ID's 29, 37, and 39). Thus, in the experiment, an NS/0 cell line containing the heavy chain VDJ gamma-4 cassette and a puromycin resistance cassette was fused with an NS/O or NS/0-bcl-2 cell line containing the human kappa light chain cassette and the HPRT (IAT resistance) cassette. Fusion and selection were accomplished using the procedure described in Example 3 and productivity determined in accordance with Example 4.
-29- Table 6 :lone ID mg/ml le total mg Ig #f cells Total ee-[Ft7.1 9.76 19.52 1.020.000 2040000 9.57 7.3 4.68 936 700.000 1400000 6.69 4 6.40 12.8 610.000 1220000 1049 9. 1 15.90 31.8 890.000 1780000 17.87 19.2 16.80 33.6 900.000 1800000 18.67 [9.3 12.20 24.4 790.000 1580000 15.44 19.4 16.20 32.4 840.000 1680000 19.29 20.1 13.00 26 910.000 1820000 14.29 A2 L6.12 12.24 840.000 1680000 7.29 0._7.35 14.7 1.120.000 2240000 6.56 20.4 17.00 34 1.000.0002000000 1700 j9.1 0.19 0.372 960.000 1920000 0.19 ^9.2 0.05 0.1 930.000 1860000 0.05 9.3 6.02 2.04 900.000 1800000 6.69 .4 0.33 0.652 1.400.000 2800000 0.23 ;7.1 0.05 0.1 910.000 1820000 0.05 7.2 2.30 4.6 1.430.000 2860000 1.61 7.3 0.01 0.024 1.000.000 2000000 0.01 37.4 5.12 10.24 1.400.000 2800000 3.66 9.1 0.29 0.572 1.400.000 2800000 0.20 39.2 0.66 1.31 1.160.000 2320000 0,56 39.3 0.29 0.572 1.000.000 200000 0,29 \hQA In q n i nan nan tonnnnn \f} The instant inventions shown and described herein are what is considered to be the most practical and the preferred embodiments. It is recognized, however, that departures may be made therefrom which are within the scope of the invention, and that obvious modifications will occur to one skilled in the art upon reading this disclosure.
C C

Claims (21)

1. A method for producing an antibody, said method comprising: introducing an expression vector comprising a nucleotide sequence encoding a desired heavy chain into a first cell; introducing an expression vector comprising a nucleotide sequence encoding a desired light chain into a second cell; and fusing the first and second cells to form a hybrid cell, wherein the hybrid cell expresses an antibody comprised of the heavy chain and the light chain, and wherein an antibody is produced by the hybrid cell.
2. The method of claim 1, further comprising: culturing the hybrid cells so as to express the antibody; and recovering the antibody from the hybrid cell culture.
3. The method of claim 1, wherein said nucleotide sequence is obtained from a B-cell or a hybridoma cell, wherein said B-cell or hybridoma cell produces an antibody.
4. The method of claim 1, wherein the first cell expresses an irrelevant light chain and expresses the desired heavy chain prior to fusion with the second cell.
The method of claim 1, wherein expression of the heavy chain by the first cell is determined by ELISA analysis oflysate from the first cell.
6. The method of claim 1, wherein the antibody is expressed only after fusion of said first and second cells.
7. The method of claim 1, wherein the first cell expressing the desired heavy chain is further selected for high production rate or level of the heavy chain.
8. The method of claim 1, wherein both the second cell expressing the desired light chain and the first cell expressing the desired heavy chain are each further selected for high product rate of level of the heavy chain and of the light chain respectively. V
9. The method of claim 8, wherein the hybrid cell is further selected for expression of a heavy chain and expression of a light chain that combine to provide an antibody having a desired antigen binding affinity.
A method for producing an antibody, said method comprising: 30 introducing an expression vector comprising a nucleotide sequence encoding a desired heavy chain into a first cell, wherein the first cell expresses an irrelevant light **.chain; introducing an expression vector comprising a nucleotide sequence encoding a desired light chain into a second cell; and m:\speci\1 00000\1 06-107\1 06634divsoarob.doc fusing the first and second cells to form a hybrid cell, wherein the hybrid cell expresses an antibody comprised of the heavy chain and the light chain, and wherein an antibody is produced by the hybrid cell.
11. The method of claim 10, further comprising: culturing the hybrid cells so as to express the antibody; and recovering the antibody from the hybrid cell culture.
12. The method of claim 10, wherein said irrelevant light chain is present in an episomal vector.
13. A method for producing a human antibody, said method comprising: introducing an expression vector comprising a nucleotide sequence encoding a human heavy chain polypeptide into a first mammalian host cell; introducing an expression vector comprising a nucleotide sequence encoding a human light chain polypeptide into a second mammalian host cell; and fusing the first and second cells to form a hybrid cell, wherein the hybrid cell expresses a human antibody comprised of the human heavy chain polypeptide and the human light chain polypeptide and wherein an antibody is produced by the hybrid cell.
14. The method of claim 13, wherein the nucleotide sequence encoding a human heavy chain polypeptide and the nucleotide sequence encoding a human light chain polypeptide are each obtained from a B-cell or a hybridoma cell, wherein said B-cell or hybridoma cell produces a human antibody.
The method of claim 13, wherein the first cell expresses both an irrelevant light chain and the human heavy chain polypeptide prior to fusion with the second cell.
16. The method of claim 13, wherein the human antibody is expressed only after fusion of said first and second mammalian cells. 25
17. The method of claim 13, wherein the first cell expressing the heavy chain is V further selected for based on its rate of antibody production.
18. The method of claim 13, wherein both the second cell expressing the light chain and the first cell expressing the heavy chain are each further selected for based on their rate of production.
19. The method of claim 13, wherein the first mammalian host cell is a non- lymphoid cell and the second mammalian host cell is a non-lymphoid cell.
The method of claim 13, wherein the first mammalian host cell is a myeloma cell and the second mammalian host cell is a myeloma cell. m:\speci\1 00000\1 06-107\1 06634divsoarob.doc
21. The method of claim 13, wherein the first mammalian host cell is a NSO cell and the second mammalian host cell is a NSO cell. Dated this Tenth day of October 2003 Japan Tobacco, Inc., Abgenix, Inc. Patent Attorneys for the Applicant: F B RICE CO S* m:\speci\1 00000\1 06-107\106634divsoarob.doc
AU72174/01A 1996-10-11 2001-09-18 Production of a multimeric protein by cell fusion method Expired AU768101B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU72174/01A AU768101B2 (en) 1996-10-11 2001-09-18 Production of a multimeric protein by cell fusion method
AU2004200848A AU2004200848A1 (en) 1996-10-11 2004-03-03 Production of a multimeric protein by cell fusion method (D1)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/730639 1996-10-11
AU49898/97A AU734800B2 (en) 1996-10-11 1997-10-10 Production of a multimeric protein by cell fusion method
AU72174/01A AU768101B2 (en) 1996-10-11 2001-09-18 Production of a multimeric protein by cell fusion method

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU49898/97A Division AU734800B2 (en) 1996-10-11 1997-10-10 Production of a multimeric protein by cell fusion method

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2004200848A Division AU2004200848A1 (en) 1996-10-11 2004-03-03 Production of a multimeric protein by cell fusion method (D1)

Publications (2)

Publication Number Publication Date
AU7217401A AU7217401A (en) 2001-12-20
AU768101B2 true AU768101B2 (en) 2003-12-04

Family

ID=29741451

Family Applications (1)

Application Number Title Priority Date Filing Date
AU72174/01A Expired AU768101B2 (en) 1996-10-11 2001-09-18 Production of a multimeric protein by cell fusion method

Country Status (1)

Country Link
AU (1) AU768101B2 (en)

Also Published As

Publication number Publication date
AU7217401A (en) 2001-12-20

Similar Documents

Publication Publication Date Title
AU734800B2 (en) Production of a multimeric protein by cell fusion method
US6420140B1 (en) Production of multimeric protein by cell fusion method
AU2009248834B2 (en) Method of generating single VL domain antibodies in transgenic animals
RU2518340C2 (en) Recombinant expression vector elements (reves) to enhance expression of recombinant proteins in host cells
EP0488470B1 (en) Method for the production of antibodies
JP6019540B2 (en) Identification of antigen-specific or ligand-specific binding proteins
JP2005525800A (en) Methods for the production of genetically modified vertebrate precursor lymphocytes and their use for the production of heterologous binding proteins.
CN114026224B (en) Mammalian cell lines with SIRT-1 gene knockouts
US8859278B2 (en) Fully human hybridoma fusion partner cell lines
JP7026634B2 (en) B cell culture method
AU768101B2 (en) Production of a multimeric protein by cell fusion method
US20040053363A1 (en) Method for production of a multimeric protein by cell fusion
JP2011182645A (en) Method for displaying animal cell and method for maturing whole human monoclonal antibody in vitro using the same
CN114080451B (en) Method for generating protein expressing cells by targeted integration using Cre mRNA
US20040053359A1 (en) Method for production of a multimeric protein by cell fusion

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)