Efficient production of IgM in recombinant mammalian cells

Information

  • Patent Application
  • 20060063234
  • Publication Number
    20060063234
  • Date Filed
    November 09, 2005
    18 years ago
  • Date Published
    March 23, 2006
    18 years ago
Abstract
Described is an immortalized human retina cell expressing E1A and E1B proteins of an adenovirus, wherein the cell includes recombinant nucleic acid encoding an IgM molecule in expressible format. Also described are methods for recombinant production of an IgM molecule, such methods including culturing a cell of the invention and expressing the recombinant nucleic acid encoding an IgM.
Description
FIELD OF THE INVENTION

The invention relates generally to the field of biotechnology and recombinant protein production. More in particular, the invention relates to the production of immunoglobulins of class M (IgM) in recombinant mammalian host cells.


Immunoglobulin molecules may be one of five classes based on amino acid sequence of the constant region of the molecule. These classes are IgG, IgM, IgA, IgD and IgE. Each class has different biological roles based on class-specific properties (Roitt, I., Brostoff, J., Make, D. (2001). Immunology. 6th edition, pub. Mosby).


IgM is the first immunoglobulin produced by B cells in response to stimulation by antigen, and is present at around 1.5 mg/ml in serum with a half-life of 5 days. One IgM monomer comprises two heavy chains and two light chains. The heavy chains consist of an N-terminal variable region, followed by four constant regions. At the C-terminus there is a tail-piece which has a function in multimerization of the molecule. Within the constant regions are cysteine residues that form disulphide bonds with a second heavy chain, and each heavy chain is covalently linked by a disulphide bond to a light chain. Light chains may be of the kappa or lambda class. IgM is unique among immunoglobulins in that the monomeric unit exists mainly in a pentameric or hexameric structure, the five or six monomeric units being all identical (FIG. 1). A J chain may also be present in the structure: this binds to the tail-piece at the C-terminus of the heavy chains and helps to mediate multimerization (Yoo et al., 1999). It is commonly observed that IgM containing a J chain exists as a pentamer, and IgM without a J chain exists as a hexamer. The quaternary structure may also impact on the biological activity of the complete molecule (Wiersma, et al., 1998; Johansen et al., 2000). A complete IgM has a mass of approximately 1000 to 1200 kDa.


Each IgM heavy chain has five or six potential N-glycosylation sites on each of the 10 to 12 heavy chains in one IgM molecule, and the glycans make up around 12% of the mass of the molecule. While little is known about the biological activity of IgM glycans, it is likely that they play a role in protein function as has been demonstrated for the glycans present on IgG. The J chain is also glycosylated. Studies on the glycosylation of human IgM have mostly relied on data from pathological IgM derived from patients with a macroglobulinaemia, but there are some studies from cell-lines producing IgM (Leibiger et al., 1998; Wang et al., 2003). A wide range of glycan structures have been observed on IgM molecules, including high mannose, bi- and tri-antennary structures. However IgMs produced in non-human cell-lines have been seen to contain Galα (1,3) Gal structures, and N-glycolylneuraminic acid: these are not found in humans and are potentially immunogenic (Leibiger et al., 1998). This is an important factor in the production of IgM if it is to be administered therapeutically.


Natural IgM molecules often have a low affinity for antigen; however this is compensated by the high valency, which gives the IgM molecule a high avidity for its target. However if an antigen binding region with a high affinity, e.g. of an IgG, is converted to an IgM format, then the avidity is likely to be extremely high. Multivalency also results in the ability to aggregate bacteria and other cells, making them easier to eliminate.


IgM binds complement with higher affinity than IgG (the hexameric form being more active in this respect than the pentameric form; Wiersma et al., 1998), providing a highly potent mechanism for complement dependent cytotoxicity. As a result, IgM is often the preferred class of immunoglobulin physiologically to combat bacterial infection.


Another mechanism by which antibodies are thought to eliminate target cells is by binding to, and often cross-linking, cell surface receptors (Ghetie et al., 1997; Tutt et al., 1998; Longo, 2002). This can lead to activation of signalling pathways resulting in arrest of cell growth or apoptosis. The multivalency of IgM makes this molecule potentially very potent at cross-linking surface receptors: IgM molecules have been seen to sit like a crab on the surface of a cell, the variable regions bent over to bind at the cell surface.


There is already evidence in the literature that IgMs may be of value as therapeutics. A natural IgM antibody has been implicated in regression of neuroblastoma cells in human patients, suggesting that it can function as a physiological tumor defense mechanism (Ollert et al, 1996). This has also been studied as a potential therapeutic against neuroblastoma (Engler et al., 2001). There are also data which show that an IgM specific for human gastrointestinal adenocarcinomas is more potent than the IgG format of the same antibody in lysing a colon carcinoma cell-line, possibly as a result of increased complement deposition on the cells (Fogler et al., 1989).


The monoclonal IgG antibody OKT3 was the first monoclonal to be used in the clinic, and is used to treat renal allograft rejection; one drawback to this therapy is the release of cytokines. To try to reduce this, the same antibody was tested in an IgM format in a mouse model where it was observed successfully to reduce inflammation (Choi et al., 2002).


There is also much interest in anti-microbial activities of immunoglobulins, a function for which IgM is ideally suited for reasons mentioned above. Studies have also been performed which show that IgM molecules against bacterial lipopolysaccharides can reduce mortality in septic patients with Gram-negative bacteremia (Bogard et al., 1993; Krieger et al., 1993; Seifert et al., 1996).


While these potential advantages of IgM are clear, there is little data in the literature regarding production in cell lines (see Yoo et al, 2002; Knight et al, 1992). Wood et al (1990) reported that IgM could be produced in CHO cells with an initial production of 1 to 1.5 pg mu chain per cell per day. This rate rose to approximately 30 pg per cell per day after gene amplification with methotrexate and 2′-deoxycoformycin. However, amplification is often associated with instability of expression (Kim et al, 1998; Barnes et al, 2003). Moreover, IgMs produced in non-human cell-lines have been seen to contain Galα (1,3)Gal structures, and N-glycolylneuraminic acid: these are not found in humans and are potentially immunogenic (Leibiger et al., 1998).


Therefore, a need remains for a good production platform for recombinant IgM production, without the drawbacks associated with the existing platforms.


DESCRIPTION OF THE INVENTION

The characteristics of a platform for production of IgM would preferably include high IgM productivity and human-type glycosylation of the molecule. It is demonstrated herein that PER.C6™ cells are capable of efficiently producing and secreting recombinant IgM molecules. High levels of functional IgM are expressed from recombinant cells without the need for amplification of the copy number of the recombinant nucleic acid encoding the IgM. The expressed IgM is in multimeric form and contains mainly biantennary N-linked glycans with a high galactose content. Glycan structures that are known to be immunogenic in man, such as Galα (1,3) Gal structures and N-glycolylneuraminic acid, have not been found on the IgM produced according to the invention.


In certain embodiments, the invention provides an immortalized human retina cell expressing E1A and E1B proteins of an adenovirus, wherein said cell comprises recombinant nucleic acid encoding an IgM molecule in expressible format.


In certain embodiments, the invention provides a method for recombinantly producing an IgM molecule, the method comprising: a) providing an immortalized human retina cell expressing E1A and E1B proteins of an adenovirus, wherein the cell further comprises recombinant nucleic acid encoding an IgM molecule in expressible format; and b) culturing the cell and expressing the recombinant nucleic acid encoding an IgM. In certain embodiments, the method further comprises the step of: c) isolating the recombinant IgM from the cells, from the culture medium or from both the cells and the culture medium.


In certain embodiments, the invention provides for the use of an immortalized human retina cell expressing E1A and E1B proteins of an adenovirus for recombinant expression of IgM molecules. In preferred embodiments, the cells of the invention are PER.C6™ cells or derived therefrom.




BRIEF DESCRIPTION OF THE FIGURES


FIG. 1. Stylized schematic showing hexameric IgM (with no J chain).



FIG. 2. Vector for expression of intact IgM. IgM-encoding regions (light chain, heavy chain), CMV promoters and the neomycin resistance marker are indicated.



FIG. 3. Expression levels of IgM-expressing PER.C6™ cells. Cells from 25 different clones were seeded at 1×106 cells per well of a 6-well dish and allowed to grow for 4 days. Supernatant was then harvested and assayed for IgM by ELISA.



FIG. 4. Reducing SDS-PAGE (stained with Coomassie Blue) of crude cell culture supernatant from clones producing IgM, material after Q-sepharose chromatography, and final purified IgM.



FIG. 5. Gel filtration HPLC analysis of purified sample of IgM. A: PER.C6™ anti-EpCAM IgM; B: IgG standard; C: human IgM standard; D: molecular weight standards (proteins of 670, 158, 44, 17 and 1 kDa). Elution times of the main peaks are shown for the immunoglobulin samples; elution time of the 670 kDa molecular weight standard is also shown.



FIG. 6. Binding of IgM to LS174T cells. FACS-derived mean fluorescent intensity (MFI) is shown.



FIG. 7. Complement dependent cytotoxicity test of anti-EpCAM IgM on LS174T cells. Also present as controls are anti-EpCAM IgG, GBSIII IgG (an antibody which binds a bacterial surface antigen and hence acts as a negative control), as well as no antibody.



FIG. 8. MALDI-MS spectrum of (de-sialylated) glycans released from anti-EpCAM IgM produced in PER.C6™. The proposed structures of the two main glycan species are indicated. Key: □ N-Acetylglucosamine, ● galactose, ∘ mannose, Δ fucose




DETAILED DESCRIPTION OF THE INVENTION

As disclosed herein, cells derived from human retina cells, which have been immortalized by introduction of E1 sequences from an adenovirus, are a good production platform for recombinant IgM molecules. A method for immortalization of embryonic retina cells has been described in U.S. Pat. No. 5,994,128, the contents of which are incorporated herein by this reference. Accordingly, an embryonic retina cell that has been immortalized with E1 sequences from an adenovirus can be obtained by that method. Such a cell expresses at least the E1A region of an adenovirus, and preferably also the E1B region. E1A protein has transforming activity, while E1B protein has anti-apoptotic activities. The cells of the invention therefore preferably express E1A and E1B proteins of an adenovirus. In preferred embodiments, such cells are derived from PER.C6™ cells. A PER.C6™ cell as used herein, is a cell having essentially the characteristics as the cells deposited at the ECACC on 29 Feb. 1996, under number 96022940. Cells derived from a PER.C6™ cell according to the invention can be obtained by introduction of foreign genetic material encoding an IgM molecule into such PER.C6™ cells. Preferably, the cells are from a stable clone that can be selected and propagated according to standard procedures known to the person skilled in the art. A culture of such a clone is capable of producing recombinant IgM molecules. Cells according to the invention preferably are able to grow in suspension culture in serum-free medium.


It has previously been shown that PER.C6™ cells can express intact human IgG (WO 00/63403, the contents of which are incorporated herein by this reference), that such IgGs have human-type glycans and the cells can be grown at large scale (Jones et al, 2003; Nichols et al, 2002). However, no specific data have been provided for other immunoglobulin classes. The present invention teaches that these cells can efficiently produce an entirely different class of immunoglobulins that have very different characteristics from IgG, i.e., IgM molecules. It was unexpectedly found that IgM can be produced in PER.C6™ cells at levels comparable to those for IgG. Moreover, the produced IgM was shown to be functional and to have a human-type glycosylation. These aspects could not be foreseen based upon data of the much smaller IgG molecules.


To obtain expression of nucleic acid sequences encoding IgM, it is well known to those skilled in the art that sequences capable of driving such expression can be functionally linked to the nucleic acid sequences encoding the IgM molecules, resulting in recombinant nucleic acid molecules encoding an IgM in expressible format. “Functionally linked” is meant to describe that the nucleic acid sequences encoding the IgM antibody fragments or precursors thereof are linked to the sequences capable of driving expression such that these sequences can drive expression of the antibodies or precursors thereof. Useful expression vectors are available in the art, for instance, the pcDNA vector series of Invitrogen. Where the sequence encoding the IgM polypeptide of interest is properly inserted with reference to sequences governing the transcription and translation of the encoded polypeptide, the resulting expression cassette is useful to produce the IgM of interest, referred to as expression.


“Sequences driving expression” may include promoters, enhancers and the like, and combinations thereof. These should be capable of functioning in the host cell, thereby driving expression of the nucleic acid sequences that are functionally linked to them. Promoters can be constitutive or regulated, and can be obtained from various sources, including viruses, prokaryotic, or eukaryotic sources, or artificially designed. Expression of nucleic acids of interest may be from the natural promoter or derivative thereof or from an entirely heterologous promoter. Some well-known and much used promoters for expression in eukaryotic cells comprise promoters derived from viruses, such as adenovirus, for example, the E1A promoter, promoters derived from cytomegalovirus (CMV), such as the CMV immediate early (1E) promoter, promoters derived from Simian Virus 40 (SV40), and the like. Suitable promoters can also be derived from eucaryotic cells, such as metallothionein (MT) promoters, elongation factor 1α (EF-1α) promoter, actin promoter, an immunoglobulin promoter, heat shock promoters, and the like. In one embodiment the sequence capable of driving expression comprises a region from a CMV promoter, preferably the region comprising nucleotides −735 to +95 of the CMV immediate early gene enhancer/promoter. This gives particularly high expression levels in cells expressing E1A of an adenovirus.


Culturing a cell is done to enable it to metabolize, and/or grow and/or divide and/or produce recombinant proteins of interest. This can be accomplished by methods well known to persons skilled in the art, and includes but is not limited to providing nutrients for the cell. The methods comprise growth adhering to surfaces, growth in suspension, or combinations thereof. Several culturing conditions can be optimized by methods well known in the art to optimize protein production yields. Culturing can be done for instance in dishes, roller bottles or in bioreactors, using batch, fed-batch, continuous systems, hollow fiber, and the like. In order to achieve large scale (continuous) production of recombinant proteins through cell culture it is preferred in the art to have cells capable of growing in suspension, and it is preferred to have cells capable of being cultured in the absence of animal- or human-derived serum or animal- or human-derived serum components. Thus, purification is easier and safety is enhanced due to the absence of additional animal or human proteins derived from the culture medium, while the system is also very reliable as synthetic media are the best in reproducibility.


The conditions for growing or multiplying cells (see, e.g., Tissue Culture, Academic Press, Kruse and Paterson, editors (1973)) and the conditions for expression of the recombinant product may differ somewhat, and optimization of the process is usually performed to increase the product yields and/or growth of the cells with respect to each other, according to methods generally known to the person skilled in the art. In general, principles, protocols, and practical techniques for maximizing the productivity of mammalian cell cultures can be found in Mammalian Cell Biotechnology: a Practical Approach (M. Butler, ed., IRL Press, 1991).


The IgM is expressed in the cells according to the invention, and may be recovered from the cells or preferably from the cell culture medium, by methods generally known to persons skilled in the art. Such methods may include precipitation, centrifugation, filtration, size-exclusion chromatography, affinity chromatography, cation- and/or anion-exchange chromatography, hydrophobic interaction chromatography, and the like. In certain aspects of the invention, the isolation of the IgM comprises an anion exchange chromatography step and/or a gel filtration step.


It is demonstrated herein that IgM can be expressed at high levels without the necessity for first amplifying the nucleic acid sequences encoding the IgM within the host cells. This has the advantage that no large copy numbers are required for efficient expression according to the invention, in contrast to previously described recombinant IgM production systems, where amplification was required to obtain levels of around 30 pg mu chain per cell per day. PER.C6™ cells expressing IgG at high levels have been shown to contain usually between 1 and 10 copies of the nucleic acid encoding the IgG per cell (Jones et al, 2003). Methods to determine copy numbers are known to the person skilled in the art of molecular biology, and include Southern blotting, quantitative PCR, fiber-FISH, and the like. Hence, the invention provides for a method according to the invention wherein the cells comprise 1-20, usually between 1 and 10 copies per cell of the recombinant nucleic acid encoding the IgM molecule. This has the advantage of establishing a production system fast, as no labor-intensive and time consuming amplification step is needed to obtain clones with sufficiently high expression levels for analysis. Moreover, such cells are expected to be more stable than cells containing high copy numbers of the transgene, that are reported to display instability upon propagation of the cells (Kim et al, 1998; Barnes et al, 2003). Therefore, also in view of regulatory requirements the cells and methods according to the invention are an improvement over those of the prior art. Preferably, cells in the method of the invention express at least 5 pg IgM per seeded cell per day, more preferably at least 20 pg per seeded cell per day.


The IgM production system of the invention is not dependent upon co-expression of the J-chain for production of functional IgM in the form of multimers. Optionally however, J-chain may be co-expressed.


An IgM molecule is an immunoglobulin wherein the heavy chains are mu chains. An IgM molecule can have a pentameric or hexameric structure. An IgM molecule according to the invention may be of any origin, including human, rodent, chimeric, humanized, and the like, however human IgMs are preferred in the invention. Using a human cell line for the production provides these molecules with a human-type glycosylation, resulting in production of IgM molecules that are not recognized as foreign by the human immune system, because both the polypeptide and the glycan portion are human. The person skilled in the art will be aware of the possibilities to obtain human IgM sequences. The sequences for the constant regions are known, and are also provided herein. Sequences encoding human variable regions may e.g. be obtained by known methods such as phage display (methods e.g. described in CF Barbas III et al, Phage Display. A laboratory manual. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 2001) or by immunizing transgenic mice that comprise genetic material encoding a human immunoglobulin repertoire (Fishwild et al, 1996; Mendez et al, 1997).


Antibodies may be used as naked molecules, or they may be in the form of immunoconjugates or labeled molecules, and so used as a magic bullet to deliver a cargo to a tumor or infection for therapy or imaging (Carter, 2001; Borrebaeck and Carlsson, 2001; Park and Smolen, 2001). Immunoconjugates comprise antigen binding domains and a non-antibody part such as a toxin, a radiolabel, an enzyme, and the like. IgM molecules may be labeled in the same way as IgG molecules, but the high avidity would likely mean that they are less likely to dissociate from the target antigen once bound. This advantageously could deliver a cargo to a target cell more permanently. Hence, the term “IgM molecule” as used herein includes naked IgM molecules, but may also refer to immunoconjugates comprising IgM molecules.


The IgM generated in this study is against the human tumor antigen EpCAM (epithelial cell adhesion molecule), a 40 kDa glycoprotein expressed on the surface of colon carcinoma cells. The high expression of EpCAM on colon carcinomas makes it an attractive target for immunotherapy. The antibody was isolated from a semi-synthetic phage library as a single chain Fv fragment named UBS54 (WO 01/48485; Huls et al, 1999). In one aspect the invention therefore provides a recombinant human IgM molecule that is capable of binding to EpCAM. In other embodiments the human IgM molecules are used for the preparation of a medicament and/or for direct treatment of a disease such as cancer.


EXAMPLES

The invention will now be described by some examples, not to be construed to limit the scope of the invention. The practice of this invention will employ, unless otherwise indicated, conventional techniques of immunology, molecular biology, microbiology, cell biology, and recombinant DNA, which are within the skill of the art. See e.g. Sambrook, J. and Russell, D. W. (2001). Molecular cloning: a laboratory manual. Pub. Cold Spring Harbor Laboratory Press; Current Protocols in Molecular Biology, Ausubel F M, et al, eds, 1987; the series Methods in Enzymology (Academic Press, Inc.); PCR2: A Practical Approach, MacPherson M J, Hams B D, Taylor G R, eds, 1995; Antibodies: A Laboratory Manual, Harlow and Lane, eds, 1988.


Example 1
Construction of pEpcamIgM Expression Vector

An expression plasmid was generated which encodes both the light and heavy chains of an IgM antibody that recognizes EpCAM. The DNA encoding the antigen-binding region of this antibody was first isolated from a scFv phage display library (Huls et al, 1999). A leader sequence and constant regions were added essentially as described in Boel et al, 2000. The genomic DNA encoding the light and heavy chains (genomic sequences of the antibody-encoding regions provided in SEQ ID NOs. 1 and 2, amino acid sequences of the encoded anti-EpCAM IgM provided in SEQ ID NOs. 3 and 4) were then amplified using PCR to append convenient restriction sites, and then cloned into the expression vector pcDNA3002(Neo). The following primers were used for PCR of the light chain:

(SEQ ID NO:5)E001: CCTGGCGCGCCACCATGGCATGCCCTGGCTTCCTGTGG(SEQ ID NO:6)E002: CCGGGTTAACTAACACTCTCCCCTGTTGAAGC


The following primers were used for PCR of the heavy chain:

(SEQ ID NO:7)E003: GGAGGATCCGCCACCATGGCATGCCCTGGCTTCCTGTGG(SEQ ID NO:8)S02:  GGAACGCTAGCTCAGTAGCAGGTGCCAGCT


The start codon (E001, E003) and stop codon (E002, SO2) are in bold. Restriction sites AscI (E001), HpaI (E002), BamHI (E003) and NheI (SO2) are underlined. Primers E001 and E003 also include a Kozak sequence (italics). The light chain fragment of approximately 0.9 kb was digested with AscI-HpaI and inserted into pcDNA3002(Neo) digested with the same enzymes. The heavy chain fragment of approximately 2.3 kb was then digested with BamHI-NheI and inserted into pcDNA3002(Neo) containing the light chain digested with the same enzymes. The resulting plasmid is pEpcamIgM (FIG. 2). The generated construct contains DNA encoding a kappa light chain and a mu heavy chain, both preceded by a CMV promoter. The expression vector pcDNA3002(Neo), which has been described in International Patent Application PCT/NL02/00841, was deposited on Dec. 13, 2001 at the European Collection of Cell Cultures (ECACC) under number 01121318.


Example 2
Transfection of PER.C6™ Cell Line and Production of IgM

Cells were transfected with pEpcamIgM by a lipofectamine based method. In brief, PER.C6™ cells were seeded at 3.5×106 cells per 96 mm tissue culture dish. For each dish, 2 μg plasmid DNA was mixed with 10 μl lipofectamine (Gibco); this was added to the cells in serum free DMEM medium (total volume 7 ml) and incubated for 4 hours. This was then replaced with DMEM medium (i.e. containing serum). The following day (and for the ensuing 3 weeks) cells were grown in DMEM medium in the presence of 0.5 mg/ml Geneticin (G418) to select for clones that were stably transfected with the plasmid. Stable clones were picked from the plate and twenty-five were selected for analysis of IgM productivity by ELISA analysis. In brief, cells were plated at 1×106 cells per well of a 6-well dish in DMEM serum. These were incubated for 4 days, after which time supernatant was harvested and IgM concentration measured. For ELISA analysis, wells of a 96-well plate were coated with antibody raised against Ig kappa light chain. After blocking with a BSA solution, samples were added to wells at varying dilutions and incubated for 1 hour. The standard used was human IgM (Accurate Chemical cat. YSRTPHP003). After washing, detection antibody (HRP-labeled anti-IgM) was applied for 30 minutes. After a further washing step, substrate O-phenylene diamine dihydrochloride was added. Antibody concentration was determined by comparing optical density at 492 nm with that of the known antibody standard. The results are shown in FIG. 3, and the highest-producing clones picked up so far produce around 27 pg IgM/seeded cell/day. This is comparable to results seen when IgG-producing cell lines are measured with the same analysis. It is possible that further screening will allow picking up clones with even higher expression levels.


Production of antibody was performed in serum-free medium. Thus, the adherent cells in tissue culture flasks were transferred to roller bottles in PER.C6™ suspension growth medium (under which conditions the cells grow in suspension). After one week, supernatant was harvested and electrophoresed on reducing SDS-PAGE (FIG. 4). The heavy and light chains that comprise the intact, secreted antibody are the predominant protein species. The material was then purified using standard techniques over HiTrap Q (Amersham Biosciences) anion exchange chromatography (which primarily concentrates material) and then gel filtration chromatography. Material was loaded on Q-sepharose FF (Amersham Biosciences) in 20 mM sodium phosphate pH8.0 and eluted with a gradient of NaCl. Peak fractions were pooled and loaded on to a HiLoad 26/60 Superdex 200 (Amersham Biosciences) and eluted in PBS. Purity of IgM after each step is shown in FIG. 4.


Example 3
HPLC Gel Filtration of IgM

In order to test whether the IgM produced was monomeric or multimeric, purified IgM was electrophoresed over an HPLC gel filtration column (Zorbax GF450 (Agilent) in 250 mM sodium phosphate buffer pH6.8). Other control samples included recombinant human IgG, human IgM (Accurate Chemical cat. YSRTPHP003) and molecular weight standards. This is shown in FIG. 5. The small peak before the main IgM elution peak is likely to be the void volume of the column. PER.C6™ IgM elutes at a similar position to human serum IgM, and is larger than the protein standard of 670 kDa (the first peak in this chromatogram). There is no evidence for monomeric or other low valency molecules.


Example 4
Binding to LS174T Cells

LS174T cells (ATCC number CL-188) express EpCAM antigens and were therefore used as targets for determination of anti-EpCAM binding and hence IgM integrity. Cells were harvested from DMEM medium and washed in PBS. Cells were transferred to Falcon FACS tubes (0.25×106 cells per FACS tube) and washed with 2 ml PBS/0.5% BSA (wash and incubation buffer; further indicated as WB). After centrifugation at 300×g for 5 min, supernatant was removed and cells were resuspended in 100 ill antibody dilutions.


Serial dilutions of 16, 4, 1, 0.25, 0.062, 0.016 and 0.004 μg/ml IgM were prepared in WB. Three negative controls were used; 1) cells incubated without primary and secondary antibody, further referred to as “no antibody”, 2) cells incubated with only secondary antibody (PE-control), and 3) GBSIII, a negative control which recognizes a bacterial antigen, at a concentration of 40 μg/ml.


After 30 min of incubation at 4° C., cells were washed with 2 ml WB. Samples were centrifuged for 5 min at 300×g and supernatant was removed. Cells were resuspended in 100 μl goat anti-human kappa PE (diluted WB 1:100 or 1:250) and incubated for 20 min at 4° C. Subsequently, cells were washed with 2 ml WB, and centrifuged for 5 min at 300×g. The cell pellet was resuspended in 250 μl WB and samples were analyzed on a FACS Calibur in FL2 channel.


The IgM binds to the LS 174T cells in a concentration dependent manner (FIG. 6). This shows that the protein is correctly folded.


Example 5
CDC Activity

CDC activity of anti-EpCAM IgM was tested with LS174T colon carcinoma cells as target cells and human complement serum (Quidel).


Briefly, LS174T cells were harvested at 70% confluency from DMEM medium. At a concentration of 1×106 cells/ml, cells were labeled for 15 min at 37° C. with 1:50000 calcein-AM (stock concentration 3.3 μg calcein/μl DMSO) in CDC medium. Cells were washed 2 times in CDC medium and diluted to 1×106 cells/ml in the same medium. Anti-EpCAM IgG, anti-GBSIII and anti-EpCAM IgM were diluted in CDC medium to different concentrations varying from 160 to 0.006 μg/ml. Samples included 50 μl antibody, 50 μl of labeled target cells, 50 μl serum and 50 μl medium. Two negative controls were used; 1) GBS III, an antibody directed against antigen III of Streptococcus group B, and 2) samples without antibody (“no antibody”). Fifty μl CDC medium replaced the antibody solution in the “no antibody” control sample. Anti-Epcam IgG was used as a positive control. Samples were incubated for 4 hours at 37° C. in 10% CO2 incubator and then analyzed by FACS (FACSCalibur, Becton Dickinson). The percentage of lysis of target cells by the complement was determined after gating the calcein positive target cells in the FL1 channel. Propidium iodide (PI; 0.4 μg/ml) was added to determine the percentage of dead cells in the gated population. PI was detected in the FL2 channel. The percentage of dead cells was calculated by [the number of both PI positive and calcein positive cells], divided by the total number of calcein positive cells, multiplied by 100%. The presence of IgM and IgG caused complement-dependent cell lysis in a concentration-dependent manner (FIG. 7). The IgM was approximately 10-fold more potent in this assay than the IgG: 1.5 μg/ml IgG gave the same percentage cell lysis as 0.151 μg/ml IgM. Negative controls (no antibody and GBSIII) gave background signals. In other CDC assays the IgM was at least 10 times more potent than IgG in this assay (data not shown). This shows that the PER.C6™ produced IgM is functionally active.


Example 6
MALDI-MS Analysis of Glycans Present on PER.C6™ IgM

Purified IgM samples in 20 mM sodium phosphate (pH 7.2) were digested with PNGase F which releases the N-linked glycans. The glycan pools were desialylated using neuramindase and were analyzed in the reflector mode on an Applied Biosystems Voyager DE Pro MALDI mass spectrometer. The matrix was 2,5-dihydroxybenzoic acid (10 mg/ml) in 50/50/0.1 acetonitrile/water/trifluoroacetic acid. Spectra were obtained in the positive ion mode and glycans were detected as sodium adducts, [M+Na]+.


Results are shown in FIG. 8. The main peak is a biantennary, fully galactosylated structure with a core fucose. Therefore, in one aspect of the invention at least 50% of the N-linked sugar structures of the produced IgM are biantennary, fully galactosylated (not having a terminal N-Acetylglucosamine) structures with a core fucose. Mass data (combined with knowledge of fully analyzed glycan structures found on recombinant erythropoietin produced on PER.C6 cells ( . . . ) indicate that other minor structures include tri- and tetra-antennary glycans, as well as some hybrid structures. Glycans containing a Lewis X structure (structures with an additional fucose attached to the N-acetylglucosamine in the antenna) are also observed at low levels.


The examples above demonstrate that PER.C6™ cells may be transfected with a plasmid expressing IgM to give cells with a high IgM productivity. Moreover the IgM is structurally sound and functionally active, and glycans which may prove immunogenic to humans have not been observed.


REFERENCES



  • Barnes, L. M., Bentley, C. M., Dickson, A. J. (2003). Stability of protein production from recombinant mammalian cells. Biotechnol. and Bioeng. 81, 631-639.

  • Boel, E., Verlaan, S., Poppelier, M. J., Westerdaal, N. A., Van Strijp, J. A., Logtenberg, T. (2000). Functional human monoclonal antibodies of all isotypes constructed from phage display library-derived single-chain Fv antibody fragments. J Immunol Methods. 239, 153-66.

  • Bogard, W. C. Jr, Siegel, S. A., Leone, A. O., Damiano, E., Shealy, D. J., Ely, T. M., Frederick, B., Mascelli, M. A., Siegel, R. C., Machielse, B., et al. (1993). Human monoclonal antibody HA-1A binds to endotoxin via an epitope in the lipid A domain of lipopolysaccharide. J. Immunol. 150, 4438-49.

  • Borrebaeck, C. A. K. and Carlsson, R. (2001). Human therapeutic antibodies. Curr. Op. Pharmacol. 1, 404-408.

  • Carter, P. (2001). Improving the efficacy of antibody-based cancer therapies. Nature Reviews: Cancer 1, 118-129.

  • Choi, I., Schmitt, W. E., Bahre, A., Little, M., Cochlovius, B. (2002). Recombinant chimeric OKT3/IgM antibodies for immune suppression: evaluation in a human CD3 transgenic mouse model. Immunol Lett. 80, 125-8.

  • Engler, S., Thiel, C., Forster, K., David, K., Bredehorst, R., Juhl, H. (2001). A novel metastatic animal model reflecting the clinical appearance of human neuroblastoma: growth arrest of orthotopic tumors by natural, cytotoxic human immunoglobulin M antibodies. Cancer Res. 61, 2968-73.

  • Fishwild D M, O'Donnell S L, Bengoechea T, Hudson D V, Harding F, Bernhard S L, Jones D, Kay R M, Higgins K M, Schramm S R, Lonberg N. (1996). High-avidity human IgG kappa monoclonal antibodies from a novel strain of minilocus transgenic mice. Nat Biotechnol. 14, 845-51.

  • Fogler, W. E., Sun, L. K., Klinger, M. R., Ghrayeb, J., Daddona, P. E. (1989). Biological characterization of a chimeric mouse-human IgM antibody directed against the 17-1A antigen. Cancer Immunol Immunother. 30, 43-50.

  • Ghetie, M. A., Podar, E. M., Ilgen, A., Gordon, B. E., Uhr, J. W., Vitetta, E. S. (1997). Homodimerization of tumor-reactive monoclonal antibodies markedly increases their ability to induce growth arrest or apoptosis of tumor cells. Proc Natl Acad Sci USA. 94, 7509-14.

  • Huls G A, Heijnen I A F M, Cuomo M E, Koningsberger J C, Wiegman L, Boel E, van der Vuurst-de Vries A-R, Loyson S A J, Helfrich W, van Berge Henegouwen G P, van Meijer M, de Kruif J, Logtenberg T. (1999). A recombinant, fully human monoclonal antibody with antitumor activity constructed from phage-displayed antibody fragments. Nat Biotechnol. 17, 276-281.

  • Johansen F. E., Braathen R., Brandtzaeg P. (2000). Role of J chain in secretory immunoglobulin formation. Scand. J. Immunol. 52, 240-248.

  • Jones, D., Kroos, N., Anema, R., Van Montfort, B., Vooys, A., Van Der Kraats, S., Van Der Helm, E., Smits, S., Schouten, J., Brouwer, K., Lagerwerf, F., Van Berkel, P., Opstelten, D-J., Logtenberg, T., Bout, A. (2003). High-level expression of recombinant IgG in the human cell line PER.C6. Biotechnol Prog. 19, 163-8.

  • Kim S J, Kim Ns, Ryu C J, Hong H J, Lee G M. (1998). Characterization of chimeric antibody producing CHO cells in the course of dihydrofolate reductase-mediated gene amplification and their stability in the absence of selective pressure. Biotechnol Bioeng 58, 73-84.

  • Knight, D. M., McDonough, M., Moore, M. A., Abercrombie, D., Siegel, R., Ghrayeb, J. (1992). Stable expression of cloned human antibody genes in murine myeloma cells. Hum. Antibodies Hybridomas. 3, 129-136.

  • Krieger, J. I., Fletcher, R. C., Siegel, S. A., Fearon, D. T., Neblock, D. S., Boutin, R. H., Taylor, R. P., Daddona, P. E. (1993). Human anti-endotoxin antibody HA-1A mediates complement-dependent binding of Escherichia coli J5 lipopolysaccharide to complement receptor type 1 of human erythrocytes and neutrophils. J Infect Dis. 167, 865-75.

  • Leibiger, H., Kersten, B., Albersheim, P., Darvill, A. (1998). Structural characterization of the oligosaccharides of a human monoclonal anti-lipopolysaccharide immunoglobulin M. Glycobiol. 8, 497-507.

  • Longo, D. L. (2002). DR's orders: human antibody kills tumors by direct signaling. Nat. Med. 8, 781-783.

  • Mendez M J, Green L L, Corvalan J R, Jia X C, Maynard-Currie C E, Yang X D, Gallo M L, Louie D M, Lee D V, Erickson K L, Luna J, Roy C M, Abderrahim H, Kirschenbaum F, Noguchi M, Smith D H, Fukushima A, Hales J F, Klapholz S, Finer M H, Davis C G, Zsebo K M, Jakobovits A. (1997). Functional transplant of megabase human immunoglobulin loci recapitulates human antibody response in mice. Nat Genet. 15, 146-56.

  • Nichols, W. W., Lardenoije, R., Ledwith, B. J., Brouwer, K., Manam, S., Vogels, R., Kaslow, D., Zuidgeest, D., Bett, A. J., Chen, L., van der Kaaden, M., Galloway, S. M., Hill, R. B., Machotka, S. V., Anderson, C. A., Lewis, J., Martinez, D., Lebron, J., Russo, C., Valerio, D. and Bout, A. Propagation of adenoviral vectors: use of PER.C6™ cells. In Adenoviral vectors for gene therapy, (Ed. Curiel D. T. and Douglas, J. T.). Pub. Academic Press, 2002.

  • Ollert, M. W., David, K., Schmitt, C., Hauenschild, A., Bredehorst, R., Erttmann, R., Vogel, C-W. (1996). Normal human serum contains a natural IgM antibody cytotoxic for human neuroblastoma cells. Proc. Natl. Acad. Sci. USA 93, 4498-4503.

  • Park, J. W. and Smolen, J. (2001). Monoclonal antibody therapy. Adv. Prot. Chem. 56, 369-421.

  • Seifert, M., Schoenherr, G., Roggenbuck, D., Marx, U., von Baehr, R. R. (1996). Generation and characterization of a human monoclonal IgM antibody that recognizes a conserved epitope shared by lipopolysaccharides of different gram-negative bacteria. Hybridoma. 15, 191-8.

  • Tutt, A. L., French, R. R., Illidge, T. M., Honeychurch, J., McBride, H. M., Penfold, C. A., Fearon, D. T., Parkhouse, R. M. E., Klaus, G. G. B. and Glennie, M. J. (1998). Monoclonal antibody therapy of B cell lymphoma: signalling activity on tumor cells appears more important than recruitment of effectors. J. Immunol. 161, 3176-3185.

  • Wang, F., Nakouzi, A., Hogue Angeletti, R., Casadevall, A. (2003). Site-specific characterization of the N-linked oligosaccharides of a murine immunoglobulin M by high-performance liquid chromatography/electrospray mass spectrometry. Anal Biochem. 314, 266-280.

  • Wiersma, E. J., Collins, C., Fazel, S., Shulman, M. J. (1998). Structural and functional analysis of J chain-deficient IgM. J. Immunol. 160, 5979-89.

  • Wood, C. R., Domer, A. J., Morris, G. E., Alderman, E. M., Wilson, D., O'Hara, R. M., Kaufman, R. J. (1990). High level synthesis of immunoglobulins in Chinese hamster ovary cells. J. Immunol. 145, 3011-3016.

  • Yoo, E. M., Coloma, M. J., Trinh, K. R., Nguyen, T. Q., Vuong, L. U., Morrison, S. L., Chintalacharuvu, K. R. (1999). Structural requirements for polymeric immunoglobulin assembly and association with J chain. J Biol. Chem. 274, 33771-7.

  • Yoo, E. M., Chinalacharuvu, K. R., Penichet, M. L., Morrison, S. L. (2002). Myeloma expression systems. J. Immunol. Meths. 261, 1-20.


Claims
  • 1. A method for producing an IgM molecule, said method comprising: a) providing cells, said cells being immortalized human retina cells expressing E1A and E1B proteins of an adenovirus, and wherein said cells further comprise recombinant nucleic acid encoding an IgM molecule in expressible format; b) culturing said cell and expressing said recombinant nucleic acid encoding an IgM, so as to produce the IgM molecule.
  • 2. The method according to claim 1, wherein said cells comprise 1-20 copies per cell of said recombinant nucleic acid encoding the IgM molecule.
  • 3. The method according to claim 1, wherein the cells in culture produce at least 5 pg IgM molecule/seeded cell/day.
  • 4. The method according to claim 3, wherein the cells in culture produce at least 20 pg IgM molecule/seeded cell/day.
  • 5. The method according to claim 1, wherein the IgM molecule thus produced is essentially devoid of Galα(1,3)Gal structures.
  • 6. The method according to claim 1, wherein the IgM thus produced is essentially devoid of N-glycolylneuraminic acid.
  • 7. The method according to claim 1, wherein at least 50% of the N-linked sugar structures on the IgM thus produced are biantennary, fully glycosylated structures with a core fucose.
  • 8. The method according to claim 1, further comprising: c) isolating the IgM molecule from said cells, from the culture medium, or from both said cells and the culture medium.
  • 9. The method according to claim 1, wherein said IgM molecule is a human IgM molecule.
  • 10. The method according to claim 1, wherein said cells are in suspension in said culture.
  • 11. The method according to claim 1, wherein said culturing is performed at least part of the time in a serum-free culture medium.
  • 12. The method according to claim 1, wherein said IgM molecule is able to specifically bind to the EpCAM antigen.
  • 13. The method according to claim 1, wherein said cell does not comprise recombinant nucleic acid encoding a J-chain.
  • 14. An immortalized human retina cell expressing E1A and E1B proteins of an adenovirus, wherein said immortalized human retina cell comprises recombinant nucleic acid encoding an IgM molecule in expressible format.
  • 15. The immortalized human retina cell of claim 14, wherein said IgM molecule is a human IgM molecule.
  • 16. An improvement in a method of the type wherein a cell produces IgM molecules, the improvement comprising: using as said cell in said method, an immortalized human retina cell expressing adenoviral E1A and E1B proteins for recombinantly expressing an IgM molecule.
  • 17. The method according to claim 1, wherein said immortalized human retina cell is derived from a PER.C6™ cell.
  • 18. The immortalized human retina cell of claim 14, wherein said immortalized human retina cell is derived from a PER.C6™ cell.
  • 19. The immortalized human retina cell of claim 15, wherein said immortalized human retina cell is derived from a PER.C6™ cell.
  • 20. The improvement of claim 16, wherein said immortalized human retina cell is derived from a PER.C6™ cell.
Priority Claims (1)
Number Date Country Kind
PCT/EP03/50194 May 2003 WO international
CROSS-REFERENCE TO RELATED APPLICATION

This application is a continuation of PCT International Patent Application No. PCT/EP2004/050844, filed on May 18, 2004, designating the United States of America, and published, in English, as PCT International Publication No. WO 2004/104046 A1 on Dec. 2, 2004, which itself claims priority from PCT/EP03/50194, filed May 23, 2003, the contents of the entirety of both of which are incorporated by this reference.

Continuations (1)
Number Date Country
Parent PCT/EP04/50844 May 2004 US
Child 11271090 Nov 2005 US