Provided is methods for producing mixtures of antibodies from a single host cell clone, wherein, a nucleic acid sequence encoding a light chain and nucleic acid sequences encoding different heavy chains are expressed in a recombinant host cell. The recombinantly produced antibodies in the mixtures according to the invention suitably comprise identical light chains paired to different heavy chains capable of pairing to the light chain, thereby forming functional antigen-binding domains. mixtures of the recombinantly produced antibodies are also provided by the invention. Such mixtures can be used in a variety of fields.

Patent
   RE47770
Priority
Jul 18 2002
Filed
May 18 2016
Issued
Dec 17 2019
Expiry
Jul 15 2023

TERM.DISCL.
Assg.orig
Entity
Large
1
315
EXPIRED<2yrs
14. A composition comprising at least a first antibody and a second antibody,
wherein the first and second antibodies have a common light chain,
wherein the first antibody comprises two copies of a first heavy chain,
wherein the second antibody comprises one copy of the first heavy chain and one copy of a second heavy chain, and
wherein the first heavy chain and the second heavy chain are different from one another, and
wherein at least one of the two different heavy chains is modified as compared to wild-type.
1. A composition comprising a mixture of two or three non-identical antibodies and a suitable pharmaceutically acceptable carrier,
wherein two different heavy chains and a common immunoglobulin light chain able to pair with the two different heavy chains are present in the mixture of the two or three non-identical antibodies, and
wherein the mixture of two or three non-identical antibodies comprises:
a bispecific antibody and at least one monospecific antibody, and
wherein at least one of the two different heavy chains is modified as compared to wild-type.
13. A composition comprising a mixture of two or three non-identical antibodies and a suitable pharmaceutically acceptable carrier,
wherein two different heavy chains are present in the two or three antibodies, and
wherein only a common immunoglobulin light chain able to pair with the two different heavy chains is present in the mixture of the two or three antibodies, and
wherein the mixture of two or three non-identical antibodies comprises a monospecific antibody and a bispecific antibody, and
wherein at least one of the two different heavy chains is modified as compared to wild-type.
12. A composition comprising a mixture of two or three non-identical antibodies and a suitable pharmaceutically acceptable carrier,
wherein only two different heavy chains are present in the two or three antibodies, and
wherein a common immunoglobulin light chain able to pair with the two different heavy chains is present in the mixture of the two or three non-identical antibodies, and
wherein the mixture of two or three non-identical antibodies comprises a monospecific antibody and a bispecific antibody, and
wherein at least one of the two different heavy chains is modified as compared to wild-type.
2. The composition of claim 1, wherein the mixture comprises a bispecific antibody and two different monospecific antibodies.
3. The composition of claim 1, wherein the two or three non-identical antibodies have differing specificities for the same target antigen.
4. The composition of claim 1, wherein the two or three non-identical antibodies have differing affinity for the same target epitope.
5. The composition of claim 1, wherein the two or three non-identical antibodies bind to different epitopes on the same target antigen.
6. The composition of claim 1, wherein the two or three non-identical antibodies bind to different antigens.
7. The composition of claim 1, wherein the two or three non-identical antibodies are independently selected from the group consisting of IgG1, IgG2, IgG3, IgG4, IgA1, IgA2, IgD, IgE, and IgM.
8. The composition of claim 1, wherein the different immunoglobulin heavy chains are of IgG isotype.
0. 9. A composition comprising a mixture of two or three non-identical antibodies that have been produced in the same host cells,
wherein two different heavy chains and a common immunoglobulin light chain are present in the mixture of the two or three non-identical antibodies,
wherein the two different heavy chains are able to pair to said common light chain,
wherein the two different heavy chains have different binding specificities, and
wherein said heavy chains further differ in their constant regions sufficiently so that the amount of bispecific antibodies is decreased as compared to the theoretical percentage of bispecific antibodies, and
wherein the mixture of two or three non-identical antibodies comprises two different monospecific antibodies.
0. 10. A composition comprising a mixture of two or three non-identical antibodies and a suitable carrier,
wherein only two different heavy chains are present in the two or three antibodies,
wherein a common immunoglobulin light chain able to pair with the two different heavy chains is present in the mixture of the two or three non-identical antibodies, and
wherein the mixture of two or three non-identical antibodies comprises:
a bispecific antibody and at least one monospecific antibody.
0. 11. A composition comprising a mixture of two or three non-identical antibodies and a suitable carrier,
wherein two different heavy chains are present in the two or three antibodies,
wherein only a common immunoglobulin light chain able to pair with the two different heavy chains is present in the mixture of the two or three antibodies, and
wherein the mixture of two or three non-identical antibodies comprises:
a bispecific antibody and at least one monospecific antibody.
0. 15. The composition of claim 1, wherein the mixture comprises three non-identical antibodies.
0. 16. The composition of claim 1, wherein the common light chain is identical in each light chain/heavy chain pair of the two or three non-identical antibodies.
0. 17. The composition of claim 1, wherein the common light chain is the only light chain present in the composition.
0. 18. The composition of claim 1, wherein the two different heavy chains differ in their variable region.
0. 19. The composition of claim 1, wherein the two different heavy chains differ in both the variable region and constant region.
0. 20. The composition of claim 1, wherein the mixture of two or three non-identical antibodies has been produced in the same host cells.
0. 21. The composition of claim 1, wherein:
the mixture comprises three non-identical antibodies;
the common light chain is identical in each light chain/heavy chain pair of the two or three non-identical antibodies;
the common light chain is the only light chain present in the composition;
the three non-identical antibodies have been produced in the same host cells; and
the two different heavy chains differ in their variable region.
0. 22. The composition of claim 1, wherein the mixture of two or three non-identical antibodies has been isolated from the host cells which produce the antibodies.
0. 23. The composition of claim 1, wherein the light chain is able to pair with one of the two heavy chains present in the mixture.
0. 24. The composition of claim 1 wherein the two or three non-identical antibodies have a different affinity for the same target epitope.
0. 25. The composition of claim 24 wherein the different affinity is analyzed by surface plasmon resonance.
0. 26. The composition of claim 1, wherein the two or three non-identical antibodies have been isolated separately or as a mixture from a culture of the host cell.

This application is a continuation of co-pending U.S. patent application Ser. No. 12/221,021, filed Jul. 29, 2008 now U.S. Pat. No. 7,927,834, which is a divisional patent application of co-pending application Ser. No. 11/593,279, filed Nov. 6, 2006 now U.S. Pat. No. 7,429,486, which is a divisional patent application of patent application Ser. No. 11/039,767, filed Jan. 18, 2005, now U.S. Pat. No. 7,262,028, issued Aug. 28, 2007, which is a continuation of PCT International Patent Application No. PCT/EP2003/007690, filed on Jul. 15, 2003, designating the United States of America, published in English as International Publication No. WO 2004/009618 A2 on Jan. 29, 2004, which itself claims the benefit of PCT International Patent Application No. PCT/EP03/50201, filed May 27, 2003, European Patent Application No. 02077953.4, filed Jul. 18, 2002, and U.S. Provisional Patent Application Ser. No. 60/397,066, filed Jul. 18, 2002, the contents of the entirety of each of which are incorporated by reference.

Pursuant to 37 C.F.R. §1.52(e)(1)(iii), a compact disc containing an electronic version of the Sequence Listing has been submitted concomitant with this application, the contents of which are hereby incorporated by reference. A second compact disc is submitted and is an identical copy of the first compact disc. The discs are labeled “copy 1” and “copy 2,” respectively, and each disc contains one file entitled “0079WO000RD.ST25.txt” which is 27 KB and created on Mar. 4, 2011.

The invention relates generally to the field of biotechnology, and more particularly, to the field of medicine and the production of antibodies, and even more particularly, to the production of mixtures of antibodies.

The essential function of the immune system is the defense against infection. The humoral immune system combats molecules recognized as non-self, such as pathogens, using immunoglobulins. These immunoglobulins, also called antibodies, are raised specifically against the infectious agent, which acts as an antigen, upon first contact (Roitt, Essential Immunology, Blackwell Scientific Publications, fifth edition, 1984; all references cited herein are incorporated in their entirety by reference). Antibodies are multivalent molecules comprising heavy (H) chains and light (L) chains joined with interchain disulfide bonds. Several isotypes of antibodies are known, including IgG1, IgG2, IgG3, IgG4, IgA, IgD, IgE, and IgM. An IgG contains two heavy and two light chains. Each chain contains constant (C) and variable (V) regions, which can be broken down into domains designated CH1, CH2, CH3, VH, and CL, VL (FIG. 1). Antibody binds to antigen via the variable region domains contained in the Fab portion and, after binding, can interact with molecules and cells of the immune system through the constant domains, mostly through the Fc portion.

B-lymphocytes can produce antibodies in response to exposure to biological substances like bacteria, viruses and their toxic products. Antibodies are generally epitope-specific and bind strongly to substances carrying these epitopes. The hybridoma technique (Kohler and Milstein, 1975) makes use of the ability of B-cells to produce monoclonal antibodies to specific antigens and to subsequently produce these monoclonal antibodies by fusing B-cells from mice exposed to the antigen of interest to immortalized murine plasma cells. This technology resulted in the realization that monoclonal antibodies produced by hybridomas could be used in research, diagnostics and therapies to treat different kinds of diseases like cancer and auto-immune-related disorders.

Because antibodies that are produced in mouse hybridomas can induce strong immune responses in humans, it has been appreciated in the art that antibodies required for successful treatment of humans needed to be less immunogenic or, preferably, non-immunogenic. For this to be done, murine antibodies were first engineered by replacing the murine constant regions with human constant regions (referred to as chimeric antibodies). Subsequently, domains between the complementarity-determining regions (CDRs) in the variable domains, the so-called framework regions, were replaced by their human counterparts (referred to as humanized antibodies). The final stage in this humanization process has been the production of fully human antibodies.

In the art, bispecific antibodies, which have binding specificities for two different antigens, have also been described. These are generally used to target a therapeutic or diagnostic moiety, for instance, T-cell, a cytotoxic trigger molecule, or a chelator that binds a radionuclide, that is recognized by one variable region of the antibody to a cell that is recognized by the other variable region of the antibody, for instance, a tumor cell (for bispecific antibodies, see Segal et al., 2001).

One very useful method known in the art to obtain fully human monoclonal antibodies with desirable binding properties, employs phage display libraries. This is an in vitro, recombinant DNA-based, approach that mimics key features of the humoral immune response (for phage display methods, see, e.g., C. F. Barbas III et al., Phage Display, A laboratory manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 2001). For the construction of phage display libraries, collections of human monoclonal antibody heavy- and light-chain variable region genes are expressed on the surface of bacteriophage particles, usually in single-chain Fv (scFv) or in Fab format. Large libraries of antibody fragment-expressing phages typically contain more than 109 antibody specificities and may be assembled from the immunoglobulin V regions expressed in the B lymphocytes of immunized or non-immunized individuals.

Alternatively, phage display libraries may be constructed from immunoglobulin variable regions that have been partially assembled or rearranged in vitro to introduce additional antibody diversity in the library (semi-synthetic libraries) (De Kruif et al., 1995b). For example, in vitro-assembled variable regions contain stretches of synthetically produced, randomized or partially randomized DNA in those regions of the molecules that are important for antibody specificity.

The genetic information encoding the antibodies identified by phage display can be used for cloning the antibodies in a desired format, for instance, IgG, IgA or IgM, to produce the antibody with recombinant DNA methods (Boel et al., 2000).

An alternative method to provide fully human antibodies uses transgenic mice that comprise genetic material encoding a human immunoglobulin repertoire (Fishwild et al., 1996; Mendez et al., 1997). Such mice can be immunized with a target antigen and the resulting immune response will produce fully human antibodies. The sequences of these antibodies can be used in recombinant production methods.

Production of monoclonal antibodies is routinely performed by use of recombinant expression of the nucleic acid sequences encoding the H and L chains of antibodies in host cells (see, e.g., EP0120694; EP0314161; EP0481790; U.S. Pat. No. 4,816,567; WO 00/63403, the contents of the entirety of each which are incorporated herein by reference).

To date, many different diseases are being treated with either humanized or fully human monoclonal antibodies. Products based on monoclonal antibodies that are currently approved for use in humans include HERCEPTIN™ (trastuzumab, anti-Her2/Neu), REOPRO™ (abciximab, anti-Glycoprotein IIB/IIIA receptor), MYLOTARG™ (gemtuzumab, anti-CD33), RITUXAN™ (Rituximab, anti-CD20), SIMULECT™ (basiliximab, anti-CD25), REMICADE™ (infliximab, anti-TNF), SYNAGIS™ (palivizumab, anti-RSV), ZENAPAX™ (daclizumab, IL2-receptor), and CAMPATH™ (alemtuzumab, anti-CD52). Despite these successes, there is still room for new antibody products and for considerable improvement of existing antibody products.

The use of monoclonal antibodies in cancer treatment has shown that so-called “antigen-loss tumor variants” can arise, making the treatment with the monoclonal antibody less effective. Treatment with the very successful monoclonal antibody RITUXIMAB® (anti-CD20) has, for instance, shown that antigen-loss escape variants can occur, leading to relapse of the lymphoma (Massengale et al., 2002). In the art, the potency of monoclonal antibodies has been increased by fusing them to toxic compounds, such as radionuclides, toxins, cytokines, and the like. Each of these approaches, however, has its limitations, including technological and production problems and/or high toxicity.

Furthermore, it appears that the gain in specificity of monoclonal antibodies compared to traditional undefined polyclonal antibodies, comes at the cost of loss of efficacy. In vivo, antibody responses are polyclonal in nature, i.e., a mixture of antibodies is produced because various B-cells respond to the antigen, resulting in various specificities being present in the polyclonal antibody mixture. Polyclonal antibodies can also be used for therapeutic applications, for instance, for passive vaccination or for active immunotherapy, and currently are usually derived from pooled serum from immunized animals or from humans who recovered from the disease. The pooled serum is purified into the proteinaceous or gamma globulin fraction, so named because it contains predominantly IgG molecules.

Polyclonal antibodies that are currently used for treatment include anti-rhesus polyclonal antibodies, gamma globulin for passive immunization, anti-snake venom polyclonal (Cro-Fab), THYMOGLOBULIN™ for allograft rejection, anti-digoxin to neutralize the heart drug digoxin, and anti-rabies polyclonal antibodies. In currently marketed therapeutic antibodies, an example of the higher efficacy of polyclonal antibodies compared to monoclonal antibodies can be found in the treatment of acute transplant rejection with anti-T-cell antibodies. The monoclonal antibodies on the market (anti-CD25 BASILIXIMAB®) are less efficacious than a rabbit polyclonal antibody against thymocytes (THYMOGLOBULIN™) (press releases dated Mar. 12, Apr. 29, and Aug. 26, 2002, on sangstat.com). The use of pooled human sera, however, potentially bears the risk of infections with viruses such as HIV or hepatitis, with toxins such as lipopolysaccharide, with proteinaceous infectious agents such as prions, and with unknown infectious agents. Furthermore, the supply that is available is limited and insufficient for widespread human treatments. Problems associated with the current application of polyclonal antibodies derived from animal sera in the clinic include a strong immune response of the human immune system against such foreign antibodies. Therefore, such polyclonals are not suitable for repeated treatment or for treatment of individuals that were injected previously with other serum preparations from the same animal species.

The art describes the idea of the generation of animals with a human immunoglobulin repertoire, which can subsequently be used for immunization with an antigen to obtain polyclonal antibodies against this antigen from the transgenic animals (WO 01/19394, the entirety of which is incorporated herein by reference). However, many technological hurdles still will have to be overcome before such a system is a practical reality in larger animals than mice and it will take years of development before such systems can provide the polyclonal antibodies in a safe and consistent manner in sufficient quantities. Moreover, antibodies produced from pooled sera, whether being from human or animal origin, will always comprise a high amount of unrelated and undesired specificities, as only a small percentage of the antibodies present in a given serum will be directed against the antigen used for immunization. It is, for instance, known that in normal, i.e., non-transgenic, animals, about 1% to 10% of the circulating immunoglobulin fraction is directed against the antigen used for hyper-immunization; hence, the vast majority of circulating immunoglobulins is not specific.

One approach towards expression of polyclonal antibody libraries has been described (WO 95/20401; U.S. Pat. Nos. 5,789,208 and 6,335,163, the contents of the entirety of each of which are incorporated herein by reference). A polyconal library of Fab antibody fragments is expressed using a phage display vector and selected for reactivity towards an antigen. To obtain a sub-library of intact polyconal antibodies, the selected heavy and light chain-variable region gene combinations are transferred en mass as linked pairs to a eukaryotic-expression vector that provides constant region genes. Upon transfection of this sub-library into myeloma cells, stable clones produce monoclonal antibodies that can be mixed to obtain a polyclonal antibody mixture. While in theory it would be possible to obtain polyclonal antibodies directly from a single recombinant production process using this method by culturing a mixed population of transfected cells, potential problems would occur concerning the stability of the mixed cell population and, hence, the consistency of the produced polyclonal antibody mixture. The control of a whole population of different cells in a pharmaceutically acceptable large-scale process (i.e., industrial) is a daunting task. It would seem that characteristics, such as growth rates of the cells and production rates of the antibodies, should remain stable for all of the individual clones of the non-clonal population in order to keep the ratio of antibodies in the polyclonal antibody mixture more or less constant.

Disclosed are means and methods for producing a mixture of antibodies in recombinant hosts.

In one aspect, provided is a method of producing a mixture of antibodies in a recombinant host, the method comprising expressing in a recombinant host cell a nucleic acid sequence or nucleic acid sequences encoding at least one light chain and at least three different heavy chains that are capable of pairing with at least one light chain. A further aspect is the elimination of the production of potentially non-functional light-heavy chain pairing by using pre-selected combinations of heavy and light chains. It has been recognized that phage display libraries built from a single light chain and many different heavy chains can encode antibody fragments with very distinct binding properties. This feature can be used to find different antibodies having the same light chain but different heavy chains, against the same target or different targets, wherein a target can be a whole antigen or an epitope thereof. Such different targets may, for instance, be on the same surface (e.g., cell or tissue). Such antibody fragments obtained by phage display can be cloned into vectors for the desired format, e.g., IgG, IgA or IgM, and the nucleic acid sequences encoding these formats can be used to transfect host cells. In one approach, H and L chains can be encoded by different constructs that, upon transfection into a cell wherein they are expressed, give rise to intact Ig molecules. When different H chain constructs are transfected into a cell with a single L chain construct, H and L chains will be assembled to form all possible combinations. However, in contrast to approaches where different light chains are expressed, such as for the production of bispecific antibodies, this method will result only in functional binding regions. It would be particularly useful when the host, for example, a single cell line, is capable of expressing acceptable levels of recombinant antibodies without the necessity to first amplify in the cell the nucleic acid sequences encoding the antibodies. The advantage is that cell lines with only a limited copy number of the nucleic acids are expected to be genetically more stable, because there will be less recombination between the sequences encoding the heavy chains, than in cell lines where a multitude of these copies is present. A cell line suitable for use in these methods is the human cell line PER.C6® (human retina cells that express adenovirus E1A and E1B proteins). Using this method, a mixture of antibodies with defined specificities can be produced from a single cell clone in a safe, controlled, and consistent manner.

In certain embodiments, provided is a method for producing a mixture of antibodies in a recombinant host, the method comprising expressing a nucleic acid sequence or nucleic acid sequences encoding at least one light chain and at least three different heavy chains that are capable of pairing with at least one light chain in a recombinant host cell. In certain embodiments, the recombinant host cell comprises a nucleic acid sequence encoding a common light chain that is capable of pairing with at least three different heavy chains, such that the produced antibodies comprise a common light chain. Those of skill in the art will recognize that “common” also refers to functional equivalents of the light chain of which the amino acid sequence is not identical. Many variants of the light chain exist wherein mutations (deletions, substitutions, additions) are present that do not materially influence the formation of functional binding regions.

Further provided is a composition comprising a mixture of recombinantly produced antibodies, wherein at least three different heavy chain sequences are represented in the mixture. In certain embodiments, the light chains of such mixtures have a common sequence. The mixture of antibodies can be produced by the method according to the invention. Preferably, the mixture of antibodies is more efficacious than the individual antibodies it comprises. More preferably, the mixture acts synergistically in a functional assay.

Further provided is a recombinant host cell for producing mixtures of antibodies and methods for making such host cells.

Independent clones obtained from the transfection of nucleic acid sequences encoding a light chain and more than one heavy chain may express the different antibodies in the mixture at different levels. It is another aspect to select a clone using a functional assay for the most potent mixture of antibodies. Further provides a method for identifying at least one host cell clone that produces a mixture of antibodies, wherein the mixture of antibodies has a desired effect according to a functional assay, the method comprising: (i) providing a host cell with nucleic acid sequences encoding at least one light chain and nucleic acid sequences encoding at least two different heavy chains, wherein the heavy and light chains are capable of pairing with each other; (ii) culturing at least one clone of the host cell under conditions conducive to expression of the nucleic acid sequences; (iii) screening at least one clone of the host cell for production of a mixture of antibodies having the desired effect by a functional assay; and (iv) identifying at least one clone that produces a mixture of antibodies having the desired effect. This method, as used herein, can be performed using high-throughput procedures if desired. The clones identified by the method can be used to produce antibody mixtures.

In certain embodiments, further provided are transgenic non-human animals and transgenic plants or transgenic plant cells capable of expressing mixtures of antibodies and mixtures of antibodies produced by these.

In certain embodiments, further provided are pharmaceutical compositions comprising a mixture of recombinantly produced antibodies and a suitable carrier.

In certain embodiments, further provided are mixtures of antibodies for use in the treatment or diagnosis and for the preparation of a medicament for use in the treatment or diagnosis of a disease or disorder in a human or animal subject.

In certain embodiments, further provided is a method for producing a mixture of antibodies comprising different isotypes from a single host cell clone.

In certain embodiments, further provided is a method for identifying a mixture of antibodies having a desired effect in a functional assay.

In certain embodiments, further provided is a method for producing a mixture of antibodies that are capable of binding to a target, the method comprising: i) bringing a phage library comprising antibodies into contact with material comprising a target, ii) at least one step of selecting phages binding to the target, iii) identifying at least two phages that comprise antibodies binding to the target, wherein at least two antibodies comprise a common light chain, iv) introducing a nucleic acid sequence encoding the light chain and a nucleic acid sequence or sequences encoding the heavy chains of at least two antibodies into a host cell, v) culturing a clone of the host cell under conditions conducive to expression of the nucleic acid sequences.

FIG. 1 is a schematic representation of an antibody. The heavy and light chains are paired via interchain disulfide bonds (dotted lines). The heavy chain can be either of the α, γ, μ, δ or ϵ isotype. The light chain is either λ or κ. An antibody of IgG1 isotype is shown.

FIG. 2 is a schematic representation of a bispecific monoclonal antibody. A bispecific antibody contains two different functional F(Ab) domains, indicated by the different patterns of the VH-VL regions.

FIGS. 3A and 3B show a sequence alignment of VL (FIG. 3A) and VH (FIG. 3B) of K53, UBS-54 and 02-237. The DNA sequence of common VL of UBS54 and K53 is SEQ ID NO:1, while the amino acid sequence is given as SEQ ID NO:2. DNA sequences of VL of 02-237, VH of UBS54, K53 and 02-237 are SEQ ID NOS:3, 5, 7 and 9, respectively, while the amino acid sequences are given in SEQ ID NOS:4, 6, 8 and 10, respectively.

FIG. 4 is an overview of plasmids pUBS3000Neo and pCD46_3000 (Neo).

FIG. 5, Panel A, shows the isoelectric focusing (IEF) of transiently expressed pUBS3000Neo, pCD46_3000(Neo) and a combination of both. In Panel B, the upper part shows a schematic representation of the expected molecules when a single light chain and a single heavy chain are expressed in a cell, leading to monoclonal antibodies UBS-54 or K53. The lower part under the arrow shows a schematic representation of the combinations produced when both heavy chains and the common light chain are co-expressed in a host cell, with theoretical amounts when both heavy chains are expressed at equal levels and pair to each other with equal efficiency. The common light chain is indicated with the vertically striped bars.

FIG. 6 is a schematic representation of a possible embodiment of the method according to the invention (see, e.g., Example 9). At (1), introduction of nucleic acid sequences encoding one light chain and three different heavy chains capable of pairing to the common light chain to give functional antibodies into host cells is shown; at (2), selection of stable clones; (3) shows clones can be screened for, for instance, expression levels, binding; at (4), clones are expanded; and at (5), production of functional mixtures of antibodies is shown. Some or all of steps 2-5 could be performed simultaneously or in a different order.

FIGS. 7A and 7B show the sequence of VH and VL of phages directed against CD22 (clone B28), CD72 (clone II-2) (FIG. 7A), and HLA-DR (class II; clone I-2) (FIG. 7B). DNA sequences of μA μL human serum is added to the target cells. Subsequently, the assay is performed as described supra.

Alternatively, ADCC and CDC of the antibody mixtures is determined using a Europium release assay (Patel and Boyd, 1995) or using an LDH release assay (Shields et al., 2001).

The functionality of the antibody mixtures against Her-2 is also tested using in vivo animal models, such as, for instance, described in Spiridon et al., 2002.

The VH and VH VL sequences of phages against proteins present on human B-cells, i.e., CD22 (clone B28), CD72 (clone II-2) and HLA-DR (clone I-2) (FIG. 7) are cloned into expression plasmid pBC1 (as provided in the pBC1 Mouse Milk Expression System, Invitrogen Life Technologies) to obtain mammary gland- and lactation-specific expression of these human IgG molecules in transgenic animals, according to the manufacturer's instructions. These mammary gland-specific expression vectors encoding the antibody sequences for anti-CD22, anti-CD72 and anti-HLA-DR, are introduced into the murine germline according to the manufacturer's instructions. Obtained pups are screened for the presence of each of the three constructs by PCR on DNA isolated from the tail. Pups, either male or female, confirmed for being transgenic for each of the three antibodies, are weaned and matured. Female transgenic mice are fertilized at the age of 6-8 weeks and milk samples are obtained at several time points after gestation. Male transgenic mice are mated with non-transgenic females and female transgenic offspring (as determined with PCR as described above) is mated and milked as described above for the female transgenic founders. Whenever needed, female or male transgenic founders are mated for another generation to be able to obtain sufficient amounts of transgenic milk for each founder line. Transgenic milk is analyzed for the presence of human IgG with a human IgG-specific ELISA, which does not cross-react with mouse IgG or other mouse milk components. Human IgG is purified from transgenic mouse milk using Protein A-affinity chromatography according to standard procedures. Purified human IgG is analyzed on SDS-PAGE, Iso-electric focusing and binding on the targets CD22, CD72 and HLA-DR. Functionality of the antibody mixture is analyzed as described supra.

The VH-VL sequences of the phage UBS-54 directed against the homotypic adhesion molecule EP-CAM (Huls et al., 1999) was not only cloned into a vector encoding the constant domains of a human IgG1 with Kappa light chain (expression vector pUBS3000Neo), but also into an expression vector encoding the constant domains of a human IgA1 with Kappa light chain (expression vector pUBS54-IgA, FIG. 8). Hence, antibodies derived from pUBS3000Neo and pUBS54-IgA do bind to the same epitope on EPCAM. The only differences antibodies derived from pUBS3000Neo and pUBS54-IgA are in the sequences encoding the constant domains of the heavy chain, resulting in either an IgG1 or IgA1 isotype. The Kappa light chain sequences of these two vectors are identical.

Stable PER.C6® (human retina cells that express adenovirus E1A and E1B proteins)-derived cell lines expressing antibodies encoded by genetic information on pUBS3000Neo and pUBS54-IgA are generated by procedures well known to persons skilled in the art. Therefore, PER.C6® cells (human retina cells that express adenovirus E1A and E1B proteins) are seeded in DMEM plus 10% FBS in tissue culture dishes (10 cm diameter) or T80 flasks with approximately 2.5×106 cells per dish and kept overnight under their normal culture conditions (10% CO2 concentration and 37° C.). The next day, transfections are performed in separate dishes at 37° C. using Lipofectamine (Invitrogen Life Technologies) according to standard protocols provided by the manufacturer, with either 1-2 μg pUBS3000.Neo and pUBS54-IgA. As a control for transfection efficiency, a few dishes are transfected with a LacZ control vector, while a few dishes are not transfected and serve as negative controls.

After four to five hours, cells are washed twice with DMEM and given fresh medium without selection. The next day, medium is replaced with fresh medium containing 500 μg/ml G418. Cells are refreshed every two or three days with medium containing the same concentrations of G418. About 20 to 22 days after seeding, a large number of colonies are visible and from each transfection, at least 300 are picked and grown via 96-well plates and/or 24-well plates via 6-well plates to T25 flasks. At this stage, cells are frozen (at least one, but usually four vials per sub-cultured colony) and production levels of recombinant human IgG and human IgA antibody are determined in the supernatant using an ELISA specific for human IgG1 as well as an ELISA specific for human IgA. Also, at this stage, G418 is removed from the culture medium and never re-applied again. For a representative number of colonies, larger volumes are cultured to purify the recombinant human IgG1 and human IgA fraction from the conditioned supernatant using, for instance, a combination of Protein L- or LA-affinity chromatography, cation exchange chromatography, hydrophobic interaction chromatography and gel filtration. Purified human immunoglobulins from the various clones are analyzed on SDS-PAGE, Iso-electric focusing (IEF) and binding to the target EPCAM using cell lines having a high expression of this molecule. The clones will also be screened by PCR on genomic DNA for the presence or absence of pUBS3000Neo and pUBS54-IgA. The identity of the PCR products is further confirmed by DNA sequencing.

A limited number of clones, which are screened positive for the production of both EPCAM IgG1 and EPCAM IgA, are subjected to single cell sorting using a fluorescence-activated cell sorter (FACS) (Becton Dickinson FACS VANTAGE SE™). Alternatively, colonies are seeded at 0.3 cells/well to guarantee clonal outgrowth. Clonal cell populations, hereafter designated as sub-clones, are refreshed once a week with fresh medium. Sub-clones are grown and transferred from 96-well plates via 24- and 6-well plates to T25 flasks. At this stage, sub-clones are frozen (at least one, but usually four vials per sub-clone) and production levels of recombinant human IgG1 and IgA antibody are determined in the supernatant using a human IgG1-specific ELISA and a human IgA-specific ELISA. For a representative number of sub-clones, larger volumes are cultured to purify the recombinant human IgG1 and human IgA1 fraction from the conditioned supernatant using, for instance, a combination of Protein L- or LA-affinity chromatography, cation exchange chromatography, hydrophobic interaction chromatography and gel filtration. Purified human immunoglobulins from the various clones are analyzed on SDS-PAGE, Iso-electric focusing (IEF) and binding to the target EPCAM using cell lines having a high expression of this molecule.

Sub-clones will also be screened by PCR on genomic DNA for the presence or absence of pUBS3000Neo and pUBS54-IgA. The identity of the PCR products is further confirmed by DNA sequencing.

Other methods such as Southern blot and/or FISH may also be used to determine whether both constructs are present in the clonal cell line.

Phage clone UBS-54 and Clone K53 (FIG. 3) were obtained as described in Example 1. The VH and VL of clone UBS-54 was inserted into an expression vector containing the HAVT20 leader sequence and all the coding sequences for the constant domains of a human IgG1 with a Kappa light chain by a method essentially as described (Boel et al., 2000). The resulting plasmid was designated as pUBS3000Neo (FIG. 4). It will be clear that expression vectors containing heavy chain constant domains of any desired isotype can be constructed by routine methods of molecular biology, using the sequences of these regions that are all available in the art. The VH and VL sequences of Phage clone K53 are cloned into an expression vector containing the HAVT20 leader sequence and all the coding sequences for the constant domains of a heavy chain of a human IgG3 with a Kappa light chain by a method essentially as described (Boel et al., 2000). This expression vector is designated as pK53IgG3.

These plasmids are transiently expressed, either alone or in combination, in PER.C6® cells (human retina cells that express adenovirus E1A and E1B proteins). In brief, each 80 cm2 flask is transfected by incubation for four hours with 140 μl lipofectamine+10 μg DNA (either pUBS3000Neo, pK53IgG3 or 10 μg of both) in serum-free DMEM medium at 37° C. After four hours, this is replaced with DMEM+10% FBS and the cells are grown overnight at 37° C. Cells are then washed with PBS and the medium is replaced with Excell 525 medium (JRH Bioscience). The cells are allowed to grow at 37° C. for six days, after which the cell culture supernatant is harvested. Human IgG-specific ELISA analysis, i.e., measuring all IgG sub-types, is done to determine the IgG concentration in transfected and non-transfected PER.C6® cells (human retina cells that express adenovirus E1A and E1B proteins). Human IgG from each supernatant is subsequently purified using Protein A-affinity chromatography (Hightrap Protein A HP, cat. no. 1-040203) according to standard procedures, following recommendations of the manufacturer (Amersham Biosciences). After elution, samples are concentrated in a Microcon YM30 concentrator (Amicon) and buffer exchanged to 10 mM sodium phosphate, pH 6.7. Samples are analyzed for binding to the targets EPCAM and CD46 using cell lines having a high expression of these molecules such as LS174T cells. Twelve μg of purified IgG, either transiently expressed UBS-54 IgG1, K53 IgG3 or IgG from the cells in which both antibodies were co-transfected, is subsequently analyzed on iso-electric-focusing gels (Serva Pre-cast IEF gels, pH range 3-10, cat. no. 42866). Samples are loaded on the low pH side and, after focusing, stained with colloidal blue. The pI values of the major isoforms for each sample are determined to illustrate whether there has been expression of UBS-54 IgG1, K53 IgG3 or bispecific heterodimers, depending on how the cells were transfected. The identification of heterodimers would indicate that single cells have translated both the IgG3 heavy chain of K53 and the IgG1 heavy chain of UBS-54 and assembled these into a full-length IgG molecule together with the common light chain.

The absence of bispecific heterodimers indicates that it is possible to translate both the IgG3 heavy chain of K53 and the IgG1 heavy chain of UBS-54 in single cells, but that these do not assemble into a full-length IgG molecule together with the common light chain, i.e., there is preferential binding of IgG1 and IgG3 heavy chains. This could, however, also be explained by the lack of co-expression of UBS-54 IgG1 and K53 IgG3. Therefore, stable clonal cell lines expressing both pUBS3000Neo and pK53IgG3 are generated by procedures as such well known to persons skilled in the art. PER.C6® cells (human retina cells that express adenovirus E1A and E1B proteins) are seeded in DMEM plus 10% FBS in tissue culture dishes (10 cm diameter) or T80 flasks with approximately 2.5×106 cells per dish and kept overnight under their normal culture conditions (10% CO2 concentration and 37° C.). The next day, transfections are performed in separate dishes at 37° C. using Lipofectamine (Invitrogen Life Technologies) according to standard protocols provided by the manufacturer, with either 1-2 μg pUBS3000Neo, pK53IgG3 or both. As a control for transfection efficiency, a few dishes are transfected with a LacZ control vector, while a few dishes will be not transfected and serve as negative controls.

After four to five hours, cells are washed twice with DMEM and given fresh medium without selection. The next day, medium is replaced with fresh medium containing 500 μg/ml G418. Cells are refreshed every two or three days with medium containing the same concentrations of G418. About 20 to 22 days after seeding, a large number of colonies are visible and from each transfection, at least 300 are picked and grown via 96-well plates and/or 24-well plates via 6-well plates to T25 flasks. At this stage, cells are frozen (at least one, but usually four vials per sub-cultured colony) and production levels of recombinant human IgG antibody are determined in the supernatant using an ELISA specific for all sub-types of human IgG. Also, at this stage, G418 is removed from the culture medium and never re-applied again. For a representative number of colonies, larger volumes are cultured to purify the recombinant human IgG from the conditioned supernatant using Protein A-affinity chromatography (Hightrap Protein A HP, cat. no. 1-040203) according to standard procedures, following recommendations of the manufacturer (Amersham Biosciences). Purified human immunoglobulins from the various clones are analyzed on SDS-PAGE, Iso-electric focusing (IEF) and binding to the targets EPCAM and CD46 using cell lines having a high expression of these molecules such as LS174T cells. The clones are also screened by PCR on genomic DNA for the presence or absence of pUBS3000Neo and pK53IgG3. The identity of the PCR products is further confirmed by DNA sequencing.

A limited number of clones, which are screened positive for the production of both EPCAM IgG1 and K53 IgG3, are subjected to single cell sorting using a fluorescence-activated cell sorter (FACS) (Becton Dickinson FACS VANTAGE SE™). Alternatively, colonies are seeded at 0.3 cells/well to guarantee clonal outgrowth. Clonal cell populations, hereafter designated as sub-clones, are refreshed once a week with fresh medium. Sub-clones are grown and transferred from 96-well plates via 24- and 6-well plates to T25 flasks. At this stage, sub-clones are frozen (at least one, but usually four vials per sub-clone) and production levels of recombinant human IgG antibody are determined in the supernatant using a human IgG-specific ELISA. For a representative number of sub-clones, larger volumes are cultured to purify the'recombinant human IgG fraction from the conditioned supernatant usings Protein A-affinity chromatography (Hightrap Protein A HP, cat. no. 1-040203) according to standard procedures, following recommendations of the manufacturer (Amersham Biosciences). Purified human immunoglobulins from the various clones are analyzed on SDS-PAGE, Iso-electric focusing (IEF) and binding to the targets EPCAM and CD46 using cell lines having a high expression of this molecules, such as, for instance, LS174T cells, or transfectants expressing these molecules.

Sub-clones are also screened by PCR on genomic DNA for the presence or absence of pUBS3000Neo and pK53IgG3. The identity of the PCR products is further confirmed by DNA sequencing.

Other methods such as Southern blot and/or FISH may also be used to determine whether both constructs are present in the clonal cell line.

Once the clonal sub-clones are available and confirmed positive for the expression of both UBS-54 IgG1 and K53 IgG3, the presence of functional K53 and UBS-54 shows that it is possible to generate a mixture of functional IgGs with different isotypes with the common light chain in a single cell. Analysis of the expression of bispecific antibodies binding both EpCAM and CD46 will reveal to what extent the different heavy chains having a different sub-type will pair, which will influence the amount of bispecific antibodies produced. It is expected that no or very low levels of bispecific antibodies will be found in this case.

This example describes the production of mixtures of antibodies against the rabies virus as another potential target. As an antigen, the Rabies Virus Glycoprotein (RVGP) is chosen, but other rabies antigens may be chosen or included as well for this purpose. Several monoclonal antibodies recognizing RVGP have already been described in the art, and polyclonal antibodies have been recognized to be useful in treatment of rabies infections as well (e.g., EP0402029; EP0445625, the entirety of which are incorporated herein by reference).

Antibody fragments are selected using antibody phage display libraries and MAbstract™ technology, essentially as described in U.S. Pat. No. 6,265,150 and in WO 98/15833, the entirety of which is incorporated herein by reference. All procedures are performed at room temperature unless stated otherwise. The sequence of RVGP is available to one of ordinary skill in the art for cloning purposes (e.g., Yelverton et al., 1983, the entirety of which is incorporated herein by reference). An RVGP-Ig fusion protein consisting of whole RVGP fused genetically to the CH2 and CH3 domains of human IgG1 is produced using vector pcDNA3.1 Zeo-CH2-CH3 expressed in PER.C6® (human retina cells that express adenovirus E1A and E1B proteins) and coated for two hours at 37° C. onto the surface of MAXISORP™ (polystyrene based modified surface with a high affinity for polar groups) plastic tubes (Nunc) at a concentration of 1.25 μg/ml. The tubes are blocked for one hour in 2% fat-free milk powder dissolved in PBS (MPBS). Simultaneously, 500 μl (approximately 1013 cfu) of a phage display library expressing single chain Fv fragments (scFvs) essentially prepared as described by De Kruif et al. (1995a, b) and references therein, is added to two volumes of 4% MPBS. In this experiment, selections are performed using fractions of the original library constructed using only one single variable light chain gene species (e.g., a “Vκ1”-library). In addition, human serum is added to a final concentration of 15% and blocking is allowed to proceed for 30 to 60 minutes. The RVGP-Ig-coated tubes are emptied and the blocked phage library is added. The tube is sealed and rotated slowly for one hour, followed by two hours of incubation without rotation. The tubes are emptied and washed ten times in PBS containing 0.1% Tween-20, followed by washing five times in PBS. One ml glycine-HCL, 0.05 M, pH 2.2 is added, and the tube is rotated slowly for ten minutes. The eluted phages are added to 500 μl 1 M Tris-HCl pH 7.4. To this mixture, 3.5 ml of exponentially growing XL-1 blue bacterial culture is added. The tubes are incubated for 30 minutes at 37° C. without shaking. Then, the bacteria are plated on 2TY agar plates containing ampicillin, tetracycline and glucose. After overnight incubation of the plates at 37° C., the colonies are scraped from the plates and used to prepare an enriched phage library, essentially as described by De Kruif et al. (1995a, b). Briefly, scraped bacteria are used to inoculate 2TY medium containing ampicillin, tetracycline and glucose and grown at a temperature of 37° C. to an OD600nm of ˜0.3. Helper phages are added and allowed to infect the bacteria, after which the medium is changed to 2TY containing ampicillin, tetracycline and kanamycin. Incubation is continued overnight at 30° C. The next day, the bacteria are removed from the 2TY medium by centrifugation, after which the phages are precipitated using polyethylene glycol 6000/NaCl. Finally, the phages are dissolved in a small volume of PBS-1% BSA, filter-sterilized and used for a next round of selection. The selection/re-infection procedure is performed twice.

After the second round of selection, individual E. coli colonies are used to prepare monoclonal phage antibodies. Essentially, individual colonies are grown to log-phase and infected with helper phages, after which phage antibody production is allowed to proceed overnight. Phage antibody-containing supernatants are tested in ELISA for binding activity to human RVGP-Ig coated 96-well plates.

Selected phage antibodies that are obtained in the screen described above are validated in ELISA for specificity. For this purpose, human RVGP-Ig is coated to Maxisorp ELISA plates. After coating, the plates are blocked in 2% MPBS. The selected phage antibodies are incubated in an equal volume of 4% MPBS. The plates are emptied, washed once in PBS, after which the blocked phages are added. Incubation is allowed to proceed for one hour, the plates are washed in PBS 0.1% Tween-20 and bound phages are detected using an anti-M13 antibody conjugated to peroxidase. As a control, the procedure is performed simultaneously using a control phage antibody directed against thyroglobulin (De Kruif et al. 1995a, b), which serves as a negative control.

The phage antibodies that bind to human RVGP-Ig are subsequently tested for binding to human serum IgG to exclude the possibility that they recognized the Fc part of the fusion protein.

In another assay, the phage antibodies are analyzed for their ability to bind PER.C6® cells (human retina cells that express adenovirus E1A and E1B proteins) that express RVGP. To this purpose, PER.C6® cells (human retina cells that express adenovirus E1A and E1B proteins) are transfected with a plasmid carrying a cDNA sequence encoding RVGP or with the empty vector and stable transfectants are selected using standard techniques known to a person skilled in the art (e.g., J. E. Coligan et al. (2001), Current Protocols In Protein Science, volume I, John Wiley & Sons, Inc. New York, the entirety of which is incorporated herein by reference). For flow cytometry analysis, phage antibodies are first blocked in an equal volume of 4% MPBS for 15 minutes at 4° C. prior to the staining of the RVGP- and control-transfected PER.C6® cells (human retina cells that express adenovirus E1A and E1B proteins). The blocked phages are added to a mixture of unlabeled control-transfected PER.C6® cells (human retina cells that express adenovirus E1A and E1B proteins) and RGVP-transfected PER.C6® cells that have been labeled green using a lipophylic dye (PKH67, Sigma). The binding of the phage antibodies to the cells is visualized using a biotinylated anti-M13 antibody (Santa Cruz Biotechnology), followed by streptavidin-phycoerythrin (Caltag). Anti RVGP scFv selectively stains the PER.C6® RVGP transfectant while they do not bind the control transfectant.

An alternative way of screening for phages carrying single chain Fv fragments specifically recognizing human RVGP, is by use of RVGP-transfected PER.C6® cells (human retina cells that express adenovirus E1A and E1B proteins).

PER.C6® cells (human retina cells that express adenovirus E1A and E1B proteins) expressing membrane-bound RVGP are produced as described supra. Phage selection experiments are performed as described supra, using these cells as target. A fraction of the phage library comprised of scFv phage particles using only one single scFv species (500 μA μL, approximately 1013 cfu) is blocked with 2 ml RPMI/10% FCS/1% NHS for 15 minutes at RT. Untransfected PER.C6® cells (human retina cells that express adenovirus E1A and E1B proteins) (˜10×106 cells) are added to the PER.C6-RVGP cells (˜1.0×106 cells). This mixture is added to the blocked light chain restricted phage library and incubated for 2.5 hours while slowly rotating at 4° C. Subsequently, the cells are washed twice and were resuspended in 500 μl μL RPMI/10% FCS and incubated with a murine anti-RVGP antibody (Becton Dickinson) followed by a phycoerythrin (PE)-conjugated anti-mouse-IgG antibody (Caltag) for 15 minutes on ice. The cells are washed once and transferred to a 4 ml tube. Cell sorting is performed on a FACSvantage fluorescence-activated cell sorter (Becton Dickinson) and RVGP (PE positive) cells are sorted. The sorted cells are spun down, the supernatant is saved and the bound phages are eluted from the cells by resuspending the cells in 500 μl 50 mM Glycin pH2.2 followed by incubation for five minutes at room temperature. The mixture is neutralized with 250 μl 1 M Tris-HCl pH 7.4 and added to the rescued supernatant. Collectively, these phages are used to prepare an enriched phage library as described above. The selection/re-infection procedure is performed twice. After the second round of selection, monoclonal phage antibodies are prepared and tested for binding to RVGP-PER.C6® cells and untransfected PER.C6® cells (human retina cells that express adenovirus E1A and E1B proteins) as described supra. Phages that are positive on RVGP-transfected cells are subsequently tested for binding to the RVGP-IgG fusion protein in ELISA as described supra.

The selected scFv fragments are cloned in a human IgG1 format, according to methods known in the art (e.g., Boel et al., 2000). To this purpose, the VL fragment shared by the selected scFv is PCR amplified using oligos that add appropriate restriction sites. A similar procedure is used for the VH genes. Thus, modified genes are cloned in expression pCRU-K01 (ECACC deposit 03041601), which results in expression vectors encoding a complete huIgG1 heavy chain and a complete human light chain gene having the same specificity as the original phage clone. By this method, three different heavy chains are cloned into separate expression vectors, while only one of the vectors needs to comprise the common light chain sequence. These expression vectors are provisionally designated pCRU-RVGP-1, pCU-RVGP-2, and pCRU-RVGP-3. Alternatively, these three vectors may lack DNA encoding the VL region, which can then be encoded in a fourth, separate expression vector not encoding a heavy chain. It is also possible to have VL sequences present in all three or two of the three vectors comprising the different VH sequences.

Stable PER.C6® (human retina cells that express adenovirus E1A and E1B proteins)-derived cell lines are generated, according to methods known to one of ordinary skill in the art (see, e.g., WO 00/63403), the cell lines expressing antibodies encoded by genetic information on either pCRU-RVGP-1, pCRU-RVGP-2 or pCRU-RVGP-3 and a cell line expressing antibodies encoded by all three plasmids. Therefore, PER.C60 cells are seeded in DMEM plus 10% FBS in tissue culture dishes (10 cm diameter) or T80 flasks with approximately 2.5×106 cells per dish and kept overnight under their normal culture conditions (10% CO2 concentration and 37° C.). The next day, transfections are performed in separate dishes at 37° C. using Lipofectamine (Invitrogen Life Technologies) according to standard protocols provided by the manufacturer, with either 1-2 μg pCRU-RVGP-1, 1-2 μg pCRU-RVGP-2, 1-2 μg pCRU-RVGP-3 or 1 μg of a mixture of pCRU-RVGP-1, pCRU-RVGP-2 and pCRU-RVGP-3. As a control for transfection efficiency, a few dishes are transfected with a LacZ control vector, while a few dishes will not be transfected and serve as negative controls.

After four to five hours, cells are washed twice with DMEM and given fresh medium without selection. The next day, the medium is replaced with fresh medium containing 500 μg/ml G418. Cells are refreshed every two or three days with medium containing the same concentrations of G418. About 20 to 22 days after seeding, a large number of colonies are visible and from each transfection, at least 300 are picked and grown via 96-well plates and/or 24-well plates via 6-well plates to T25 flasks. At this stage, cells are frozen (at least one, but usually four vials per sub-cultured colony) and production levels of recombinant human IgG antibody are determined in the supernatant using an ELISA specific for human IgG1 (described in WO 00/63403). Also, at this stage, G418 is removed from the culture medium and never re-applied again. For a representative number of colonies, larger volumes will be cultured to purify the recombinant human IgG1 fraction from the conditioned supernatant using Protein A-affinity chromatography according to standard procedures. Purified human IgG1 from the various clones is analyzed on SDS-PAGE, Iso-electric focusing (IEF) and binding to the target RVGP using an RVGP PER.C6-transfectant described above.

Colonies obtained from the co-transfection with pCRU-RVGP-1, pCRU-RVGP-2 and pCRU-RVGP-3 are screened by PCR on genomic DNA for the presence or absence of each of the three constructs. The identity of the PCR products is further confirmed by DNA sequencing.

A limited number of colonies, which screened positive for the production of each of the three binding specificities (both by PCR at the DNA level as well as in the specified binding assays against RVGP), are subjected to single cell sorting using a fluorescence-activated cell sorter (FACS) (Becton & Dickinson FACS VANTAGE SE™).

Alternatively, colonies are seeded at 0.3 cells/well to guarantee clonal outgrowth. Clonal cell populations, hereafter designated as sub-clones, are refreshed once a week with fresh medium. Sub-clones are grown and transferred from 96-well plates via 24- and 6-well plates to T25 flasks. At this stage, sub-clones are frozen (at least one, but usually four vials per sub-clone) and production levels of recombinant human IgG1 antibody are determined in the supernatant using a human IgG1-specific ELISA. For a representative number of sub-clones, larger volumes are cultured to purify the recombinant human IgG1 fraction from the conditioned supernatant using Protein A-affinity chromatography according to standard procedures.

Purified human IgG1 from the various sub-clones is subsequently analyzed as described above for human IgG1 obtained from the parental clones, i.e., by SDS-PAGE, Iso-electric focusing (IEF) and binding to the target RVGP.

Sub-clones are also screened by PCR on genomic DNA for the presence or absence of each of the three constructs pCRU-RVGP-1, pCRU-RVGP-2 and pCRU-RVGP-3. The identity of the PCR products is further confirmed by DNA sequencing.

Other methods such as Southern blot and/or FISH can also be used to determine whether each of the three constructs are present in the clonal cell line.

Sub-clones that are proven to be transgenic for each of the three constructs are brought into culture for an extensive period to determine whether the presence of the transgenes is stable and whether expression of the antibody mixture remains the same, not only in terms of expression levels, but also for the ratio between the various antibody isoforms that are secreted from the cell. Therefore, the sub-clone culture is maintained for at least 25 population doubling times, either as an adherent culture or as a suspension culture. At every four to six population doublings, a specific production test is performed using the human IgG-specific ELISA and larger volumes are cultured to obtain the cell pellet and the supernatant. The cell pellet is used to assess the presence of the three constructs in the genomic DNA, either via PCR, Southern blot and/or FISH. The supernatant is used to purify the recombinant human IgG1 fraction as described supra. Purified human IgG1 obtained at the various population doublings is analyzed as described, i.e., by SDS-PAGE, Iso-electric focusing (IEF) and binding to the target RVGP.

The efficacy of the antibody mixtures against rabies is tested in in vitro cell culture assays where the decrease in spread of rabies virus is measured, as well as in in vivo animal models infected by rabies. Such models are known to one of ordinary skill in the art and are, e.g., described in EP0402029.

A method for producing a mixture of antibodies according to the invention using expression in a recombinant host cell of a single light chain and three different heavy chains capable of pairing to the single light chain to form functional antibodies, is exemplified herein and is schematically shown in FIG. 6.

Human IgGs UBS54 and K53 against the EP-CAM homo-typic adhesion molecule (Huls et al., 1999) and the membrane cofactor protein CD46 (WO 02/18948), respectively, are described in Example 1. Another clone that was identified to bind to cofactor protein CD46 was clone 02-237 (sequence of VH provided in FIG. 12, SEQ ID NO:10). DNA sequencing of this clone revealed that it contained the same light chain as UBS54 and K53 but a unique heavy chain-variable sequence (see alignment in FIG. 3). As a result, the CDR3 of the heavy chain of 02-237 differs at four positions from that of K53 (see alignment in FIG. 13). The heavy and light chain-variable sequences of phage 02-237 were cloned into the expression plasmid pCRU-K01 (pCRU-K01 is deposited at the European Collection of Cell Cultures (ECACC) under number 03041601), which contains the heavy and light chain constant domains for an IgG 1 antibody.

The resulting plasmid was designated pgG102-237. Due to the cloning strategy followed, the resulting N-terminus of the light chain of 02-237 as encoded by pgG102-237 differed slightly from the N-terminus of UBS54 and K53 as present by pUBS3000Neo, pCD46_3000(Neo), respectively (FIG. 3). Plasmid pgG102-237 was transiently produced in human 293 (T) cells or stably in PER.C6® cells (human retina cells that express adenovirus E1A and E1B proteins). It appeared that purified 02-237 IgG had a much higher affinity for purified CD46 (FIG. 14) than K53 IgG, i.e., the affinity had increased from 9.1×10−7 M to 2.2×10−8 M for K53 and 02-237, respectively. Also, 02-237 bound much better to CD46 on human colon carcinoma LS174T cells than K53 (FIG. 15).

Stable PER.C6® (human retina cells that express adenovirus E1A and E1B proteins)-derived cell lines expressing a combination of the plasmids pUBS3000Neo, pCD46_3000 (Neo) and pgG102-237 encoding human IgG 02-237 were generated according to methods known as such to one of ordinary skill in the art (see, e.g., WO 00/63403). Therefore, PER.C6® cells (human retina cells that express adenovirus E1A and E1B proteins) were seeded in DMEM plus 10% FBS in tissue culture dishes (10 cm diameter) with approximately 2.5×106 cells per dish and kept overnight under their normal culture conditions (10% CO2 concentration and 37° C.). The next day, transfections were performed in separate dishes at 37° C. using Lipofectamine (Invitrogen Life Technologies) according to standard protocols provided by the manufacturer, with 2 μg of an equimolar mixture of pUBS3000Neo, pCD46_3000(Neo) and pgG102-237. As negative control for selection, a few dishes were not transfected.

After four to five hours, cells were washed twice with DMEM and given fresh medium without selection. The next day, medium was replaced with fresh medium containing 500 μg/ml G418. Cells were refreshed every two or three days with medium containing the same concentrations of G418. About 20 to 22 days after seeding, a large number of colonies were visible and about 300 were picked and grown via 96-well plates and/or 24-well plates via 6-well plates to T25 flasks. During sub-culturing, production levels of recombinant human IgG antibody were determined in the supernatant using an ELISA specific for human IgG1 (described in WO 00/63403). About 25% of all colonies appeared to be positive in this highly specific assay. The production levels measured at this stage were comparable to the levels when a single IgG is expressed in PER.C6® cells (human retina cells that express adenovirus E1A and E1B proteins) (expression of a single IgG described in Jones et al., 2003). It is important to stress that these high expression levels were obtained without any methods for amplification of the transgene and that they occur at a low copy number of the transgene.

The 30 best producing colonies were frozen down in vials and the 19 highest producing clones were selected for purification of the IgG (Table 1). They were sub-cultured in T80 flasks and human IgG from each clone was subsequently purified using Protein A-affinity chromatography. Therefore, 15 to 25 ml of conditioned medium was loaded on a 5 ml Protein A FF Sepharose column (Amersham Biosciences). The column was washed with 4 mM phosphate buffered saline, pH 7.4 (PBS) before elution with 0.1 M citrate pH 3.0. The eluted fraction was subsequently desalted on a Sephadex G25 Fine HiPrep Desalting column (Amersham Biotech) to PBS. The concentration of the purified IgG fraction was determined by absorbance measurement at 280 nm using a coefficient of 1.4 for a 0.1% (w/v) solution (Table 1).

The purified IgG samples were analyzed on non-reduced and reduced SDS-PAGE and IEF. Non-reduced SDS-PAGE (FIG. 16A) showed that all IgG samples migrated comparable to the control K53 or 02-237 as an assembled, intact IgG molecule of approximately 150 kDa. On reduced SDS-PAGE (FIG. 16B), the IgG samples migrated as heavy and light chains of about 50 and 25 kDa, respectively, comparable to the heavy and light chain of the control K53 or 02-237.

On IEF, the purified IgG fractions were first compared to a mixture of equal amounts of K53, UBS54 and 02-237 (FIG. 17). Clearly, some of the samples contained isoforms with a unique pI profile when compared to the mixture containing purified K53, UBS54 and 02-237. Some major unique isoforms have a pI in between the pI of K53 and 02-237 on one hand and UBS54 on the other hand. This is also anticipated on the basis of the theoretic pI when calculated with the Prot-Param tool provided on the Expasy homepage (expasy.ch; Appel et al., 1994). K53, 02-237 and UBS54 have a theoretic pI of 8.24, 8.36 and 7.65, respectively, whereas an isoform representing a heterodimer of one UBS54 heavy chain and one K53 heavy chain, has a theoretical pI of 8.01. Assembly, of such a heterodimer can only occur when a single cell translates both the heavy chain of K53 and the heavy chain of UBS54 and assembles these into a full-length IgG molecule together with the common light chain. Hence, these results suggest that certain clones at least express two functional antibodies. To confirm the unique identity of some of the isoforms, samples of the most interesting clones were run in parallel with K53, UBS54 and 02-237, either alone or in a mixture (FIG. 18). This furthermore showed that some clones expressed at least two antibodies (241, 282, 361). Moreover, it provided evidence that some clones express all three functional antibodies (280 and 402).

To confirm that the clones expressed IgG mixtures comprising all three heavy chains, peptide mapping (Garnick, 1992; Gelpi, 1995, the entirety of which are incorporated herein by reference) was used to analyze the polyclonal IgG fraction. We previously employed peptide mapping to recover 99% of the protein sequence of K53.

Based on the protein sequence provided in FIG. 12, the mass of the theoretical tryptic peptides of K53, UBS54 and 02-237 was calculated (Table II and III). A few unique peptides for each IgG could be identified, for instance, the CDR3 peptides for K53, 02-237 and UBS54 with a Mw of 2116.05, 2057.99 and 2307.15 Da, respectively. Next, a tryptic digest of Poly1-280 was prepared and this was analyzed using LC-MS (FIG. 19).

Peptides with Mw of 2116, 2057 and 2308 Da, representing the unique CDR3 peptides of K53, 02-237 and UBS54, respectively, were detected. The precise amino acid sequence of these peptides (as listed in Table III) was confirmed by MS-MS analysis (Tables IV, V and VI). The presence of the two unique N-terminal light chain peptides with Mw of 2580 and 2554 Da, respectively, was also confirmed. The peptide mapping data unequivocally showed that a mixture of antibodies comprising a common light chain and three different heavy chains was expressed by PER.C6® (human retina cells that express adenovirus E1A and E1B proteins) clone Poly1-280. Also, clones 055, 241 and 402 were screened by peptide mapping. Clones 241 and 402 were confirmed positive for all three heavy chain sequences, whereas clone 055 only showed expression of the heavy chains of K53 and 02-237, and not of UBS54. This confirms the IEF screening (FIG. 18) where no UBS54-related band was seen in sample 055.

Poly1-280 was analyzed by BIACORE™ (surface plasmon resonance) for binding to CD46 (FIG. 20). The affinity of poly1-280 for CD46 was 2.1×10−8 M, which shows that the IgG mixture contains CD46-binding molecules having the same affinity as 02-237 IgG alone.

Taken together, this experiment shows that it is possible to express a mixture of functional IgG molecules comprising three unique heavy chains in a single cell and that next to the homodimers, heterodimers consisting of two binding specificities are also formed. Furthermore, the frequency of clones expressing three different heavy chains suggests that it will also be possible to obtain clones expressing at least 4, 5, or more, heavy chains, using the same procedure. In the case where it would be difficult to obtain clones expressing higher numbers of heavy chains, a clone expressing at least three heavy chains according to the invention can be used to introduce more heavy chains in a separate round of transfection, for instance by using a different selection marker.

Next, it was demonstrated that a single cell is able to produce a mixture of more than two functional human IgGs. Therefore, clones 241, 280 and 402, which were screened positive for the production of each of the three IgGs, both by IEF and MS, were subjected to limiting dilution, i.e., seeded at 0.3 cells/well in 96-well plates to guarantee clonal outgrowth.

Clonal cell populations, hereafter designated as sub-clones, were refreshed once a week with fresh medium. Sub-clones were grown and transferred from 96-well plates via 24- and 6-well plates, T25, T80 and T175 flasks. At the T80 stage, sub-clones were frozen. Production levels of recombinant human IgG1 antibody were determined in the supernatant using a human IgG1-specific ELISA. For each parental clone, three sub-clones were chosen and cultured in a few T175 flasks to obtain sufficient conditioned medium for purification using Protein A-affinity chromatography as described above.

Purified human IgG1 from the sub-clones was subsequently analyzed as described above for human IgG1 obtained from the parental clone by iso-electric focusing (IEF). The result is shown in FIG. 21. Sub-clones from clone poly 1-241 each have the same pattern, but differ from the parental clone in that they appear to miss certain bands.

Sub-clones from clone poly 1-280 all appear to differ from each other and from the parental clone. Patterns obtained by IEF for sub-clones from parental clone poly 1-402 are identical for all three sub-clones and the parent clone.

From these data, it can be concluded that clone 402 is stably producing a mixture of antibodies. This demonstrates that it is feasible to produce a mixture of antibodies according to the invention from a single cell clone. The clones have undergone about 25 population doublings (cell divisions) from the transfection procedure up to the first analysis (shown in FIG. 18) under selection pressure and, from that point on, have undergone about 30 population doublings during the sub-cloning procedure in the absence of selection pressure before the material analyzed in FIG. 21 was harvested. Therefore, the production of a mixture of antibodies from a clone from a single cell can be stable over at least 30 generations.

Purified IgG1 from the parental 241, 280 and 402 clones, and sub-clones, were also analyzed for binding reactivity towards the CD46 and EpCAM antigens. To this end, cDNA of EpCAM, CD46, and control antigen CD38 were cloned into expression vectors pcDNA (Invitrogen). These vectors were transfected into CHO (dhfr-) cells using Fugene (Roche) according to the protocol supplied by the manufacturer. Cells were cultured in Iscove's medium containing 10% FBS and HT supplement (Gibco). After culturing for two days, cells were harvested by trypsinization and suspended in PBS-1% BSA (PBSB) for use in FACS analysis.

Purified IgG1 of the clones producing the mixtures of antibodies and control IgG1 samples of anti-GBSIII, an anti-CD72 antibody (02-004), as well as antibodies from anti-EpCAM clone UBS54 and anti-CD46 clones K53 and 02-237, were diluted in PBSB to a concentration of 20 μg IgG1/ml. Twenty μl of each was added to 200,000 transfected cells and incubated on ice for one hour. Thereafter, cells were washed once in ice-cold PBSB. Bound IgG was then detected using incubation with goat-anti-human IgG-biotin followed by streptavidin-PE. After a final washing step, cells were suspended in PBSB containing 1 μg/ml propidium iodide. The samples were analyzed on a FACS (FACSvantage, Becton Dickinson). Live cells were gated and Mean Fluorescent Intensities (MFI) were calculated from the FACS plots. The results are represented in FIG. 22. As expected, UBS54 bound selectively to EpCAM-transfected cells and 02-237 and K53 bound selectively to CD46 transfectants, while unrelated antibodies did not bind to these transfectants.

The results demonstrate that binding activities towards both EpCAM and CD46 were present in the purified IgG1 preps of most clones expressing a mixture of antibodies according to the invention, demonstrating that a mixture of functional antibodies was produced by sub-clones that have undergone more than 30 cell divisions and that result from a single cell. In sub-clone 280-015, binding patterns towards CD46 and EpCAM were similar as in the parent clone poly 1-280, in contrast to the other clones.

It should be stated that the quantitative aspect of this assay is not completely clear. Routine screening, for example, by a functional test, can be used to find a clone with the desired expression profile. Quantitative aspects may also be included in such screens. Such screening allows for the identification of desired clones, which express the mixture of antibodies with a given functionality in a quantitatively stable manner.

All references, including publications, patents, and patent applications, cited herein are hereby incorporated by reference to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein.

TABLE I
Overview of the clones used for purification of IgG.
Purification
Screening Conc.
Clone ELISA in feed Purified
Polyl- (μg/ml) (μg/ml) (mg)
209 6.1 98 1.37
233 10.0 53 0.75
234 8.0 51 0.71
241 6.6 91 1.42
250 12.5 117 2.10
280 6.3 36 0.80
282 8.5 67 1.48
289 8.2 33 0.64
304 7.2 161 3.91
320 6.3 43 0.83
322 15.2 168 3.27
340 6.0 109 2.64
361 10.4 71 1.73
379 9.5 78 1.75
402 39.9 135 3.14
022 16.2 83 1.69
040 7.8 67 1.43
048 6.5 43 0.94
055 11 55 1.04

TABLE II
Tryptic peptides of the variable domains of the light chain of
K53/UBS54 and 02-237.
Monoisotopic Monoisotopic
First Last MW (Da) MW (Da)
Peptide AA1) AA K53/UBS54 02-237
L1 1 24 2580.31(2) 2554.28(2)
L2 25 59 4039.02 4039.02
L3 60 66 700.35 700.35
L4 67 79 1302.61 1302.61
L5 80 82 374.23 374.23
L6 83 107 2810.29(2) 2810.29(2)
L7 108 111 487.30 487.30
L8 112 112 174.11 174.11
1)AA, amino acid
(2)One Cysteine residue alkylated

TABLE III
Tryptic peptides of variable domains of heavy
chains of K53, 02-237 and UBS54.
K53 02-237 UBS54
A B  C D   A B  C D   A B C  D  
H1 1  12 1267.68 H1 1  12 1267.68 H1  1 12 1267.68
H2 13  19 685.41 H2 13  19 685.41 custom character custom character custom character custom character
H3 20  23 492.24 H3 20  23 492.24 H3 20 23 492.24
H4 24  38 1693.81 H4 24  38 1693.81 custom character custom character custom character custom character
H5 39  63 2783.28 H5 39  63 2783.28 custom character custom character custom character custom character
H6 64  67 472.28 H6 64  67 472.28 custom character custom character custom character custom character
H7 68  84 1906.87 H7 68  84 1906.87 custom character custom character custom character custom character
H8 85  87 374.23 H8 85  87 374.23 —  
H9 88  98 1319.55 H9 88  98 1319.55 custom character custom character custom character custom character
custom character custom character custom character custom character custom character custom character custom character custom character custom character custom character custom character custom character
custom character custom character custom character custom character custom character custom character custom character custom character —  —  
Key:
A: peptide
B: first amino acid
C: last amino acid
D: monoisotopic Mw (Da)
Remarks:
1) for H1, amino acid residue 1 is a pyroglutamic acid
2) peptides H3 and H9 from K53 and 02-237, and peptides H3 and H8 of UBS54 contain one alkylated cysteine residue
3) Unique peptides that can be used to confirm the presence of the respective IgGs are indicated in bold italics

TABLE IV
MS/MS-data of CDR3 peptide (H11) of K53, obtained by collision
induced dissociation of doubly charged m/z 1059.06.
Ion m/z Ion m/z
Y″1  147.12 B1 n.d.
Y″2  248.18 B2 157.10
Y″3 335.21(1) B3 304.18
Y″4  406.25 B4 419.22
Y″5  507.30 B5 582.31
Y″6 594.33 B6 768.38
Y″7  693.40 B7 825.39
Y″8 794.46 B8 953.43
Y″9 893.54 B9 n.d.
Y″10 1006.63 B10 n.d.
Y″11 1107.67 B11 1224.65 
Y″12 1164.68 B12 1323.68 
Y″13 1292.81 B13 1424.79 
Y″14 1349.77 B14 1523.86 
Y″15 1535.85 B15 n.d.
Y″16 1698.95 B16 n.d.
Y″17 1813.95 B17 1782.96 
Y″18 1960.97 B18 n.d.
Y″19 n.d.(2) B19 n.d.
(1)Underlined m/z-values are main peaks in the MS/MS-spectrum.
(2)n.d. is not detected.

TABLE V
MS/MS-data of CDR3 peptide (H11) of 02-237, obtained by collision
induced dissociation of doubly charged m/z 1030.02.
Ion m/z Ion m/z
Y″1 147.12 B1 n.d.
Y″2 248.18 B2 189.09
Y″3 335.20 B3 n.d.
Y″4 406.24 B4 451.22
Y″5 493.30 B5 n.d.
Y″6 580.32 B6 n.d.
Y″7 679.40 B7 n.d.
Y″8 780.44 B8 n.d.
Y″9 879.53 B9 n.d.
Y″10 992.60 B10 n.d.
Y″11 1093.65  B11 n.d.
Y″12 1150.67 B12 n.d.
Y″13 1278.80  B13 n.d.
Y″14 1335.80 B14 n.d.
Y″15 1521.83  B15 n.d.
Y″16 1608.90  B16 n.d.
Y″17 1724.00  B17 n.d.
Y″18 n.d. B18 n.d.
Y″19 n.d. B19 n.d.
1Underlined m/z-values are main peaks in the MS/MS-spectrum.
2n.d. is not detected.

TABLE VI
MS/MS-data of CDR3 peptide (H9) of UBS54, obtained by collision
induced dissociation of triply charged m/z 770.09.
Ion m/z Ion m/z
Y″1 n.d. B1 n.d.
Y″2 248.17 B2 213.17
Y″3 335.20 B3 360.16
Y″4 406.25 B4 473.27
Y″5 507.30 B5 610.32
Y″6 594.33 B6 773.41
Y″7 693.42 B7 959.48
Y″8 794.45 B8 1016.50
Y″9 893.53 B9 1144.57
Y″10 1006.64 B10 1201.59
Y″11 1107.67  B11 1302.68
Y″12 1164.68  B12 1415.72
Y″13 n.d. B13 1514.78 
Y″14 n.d. B14 n.d.
Y″15 n.d. B15 n.d.
Y″16 n.d. B16 n.d.
Y″17 n.d. B17 n.d.
Y″18 n.d. B18 n.d.
Y″19 n.d. B19 n.d.
Y″20 n.d. B20 n.d.
1Underlined m/z-values are main peaks in the MS/MS-spectrum.
2n.d is not detected.

  • Appel R. D., Bairoch A. and Hochstrasser D. F. (1994) A new generation of information retrieval tools for biologists: the example of the ExPASy WWW server. Trends Biochem. Sci. 19:258-260.
  • Bendig M. M. (1988) The production of foreign proteins in mammalian cells. Genet. Eng. 7:91-127.
  • Boel E., Verlaan S., Poppelier M. J., Westerdaal N. A., Van Strijp J. A. and Logtenberg T. (2000) Functional human monoclonal antibodies of all isotypes constructed from phage display library-derived single-chain Fv antibody fragments. J. Immunol. Methods 239:153-166.
  • Brink M. F., Bishop M. D. and Pieper F. R. (2000) Developing efficient strategies for the generation of transgenic cattle which produce biopharmaceuticals in milk. Theriogenology 53:139-148.
  • Campbell K. H., McWhir J., Ritchie W. A. and Wilmut I. (1996) Sheep cloned by nuclear transfer from a cultured cell line. Nature 380:64-66.
  • Casellas R., Shih T. A., Kleinewietfelt M., Rakoniac J., Nemazee D., Rajewski K. and Nussenzweig M. C. (2001) Contribution of receptor editing to the antibody repertoire. Science 291:1541-1544.
  • Cockett M. I., Bebbington C. R. and Yarranton G. T. (1990) High level expression of tissue inhibitor of metalloproteinases in Chinese hamster ovary cells using glutamate synthetase gene amplification. Bio/technology 8:662-667.
  • De Kruif J., Terstappen L., Boel E. and Logtenberg T. (1995a) Rapid selection of cell sub-population-specific human monoclonal antibodies from a synthetic phage antibody library. Proc. Natl. Acad. Sci. U.S.A. 92:3938
  • De Kruif J., Boel E. and Logtenberg T. (1995b) Selection and application of human single chain Fv antibody fragments from a semi-synthetic phage antibody display library with designed CDR3 regions. J. Mol. Biol. 248:97
  • Dinnyes A., De Sousa P., King T. and Wilmut I. (2002) Somatic cell nuclear transfer: recent progress and challenges. Cloning Stem Cells 4:81-90.
  • Flavell D. J., Noss A., Pulford K. A., Ling N. and Flavell S. U. (1997) Systemic therapy with 3BIT, a triple combination cocktail of anti-CD19, -CD22, and -CD38-saporin immunotoxins, is curative of human B-cell lymphoma in severe combined immunodeficient mice. Cancer Res. 57:4824-4829.
  • 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. and Lonberg N. (1996) High-avidity human IgG kappa monoclonal antibodies from a novel strain of minilocus transgenic mice. Nat. Biotechnol. 14:845-51.
  • Garnick R L. (1992) Peptide mapping for detecting variants in protein products. Develop. Biol. Standard 76:117-130.
  • Gelpi E. (1995) Biomedical and biochemical applications of liquid chromatography-mass spectrometry. J. Chromatography A 703:59-80.
  • Ghetie M.-A., Podar E. M., Ilgen A., Gordon B. E., Uhr J. W. and 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. U.S.A. 94:7509-7514.
  • Giddings G., Allison G., Brooks D. and Carter A. (2000) Transgenic plants as factories for biopharmaceuticals. Nat. Biotechnol. 18:1151-1155.
  • Gorman C. and Bullock C. (2000) Site-specific gene targeting for gene expression in eukaryotes. Curr. Opin. Biotechnol. 11:455-460.
  • Hiatt A., Cafferkey R. and Bowdish K. (1989) Production of antibodies in transgenic plants. Nature 342:76-78.
  • Huls G. A., Heijnen I. A., Cuomo M. E., Koningsberger J. C., Wiegman L., Boel E., van der Vuurst de Vries A. R., Loyson S. A., Helfrich W., van Berge Henegouwen G. P., van Meijer M., de Kruif J. and Logtenberg T. (1999) A recombinant, fully human monoclonal antibody with antitumor activity constructed from phage-displayed antibody fragments. Nat. Biotechnol. 17:276-281.
  • Jespers L. S., Roberts A., Mahler S. M., Winter G. and Hoogenboom H. R. (1994) Guiding the selection of human antibodies from phage display repertoires to a single epitope of an antigen. Biotechnology (N.Y.) 12:899-903.
  • 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. and Bout A. (2003) High-level expression of recombinant IgG in the human cell line PER.C6®. Biotechnol. Prog. 19, 163-168.
  • Kim S. J., Kim N. S., Ryu C. J., Hong H. J. and 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.
  • Kohler G. and Millstein C. (1975) Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 256:495-497.
  • Koopman G., Reutelingsperger C. P., Kuijten G. A., Keehnen R. M., Pals S. T. and van Oers M. H. (1994) Annexin V for flow cytometric detection of phosphatidylserine expression on B cells undergoing apoptosis. Blood 84:1415-1420.
  • Larrick J. W. and Thomas D. W. (2001) Producing proteins in transgenic plants and animals. Curr. Opin. Biotechnol. 12:411-418.
  • Massengale W. T., McBurney E. and Gurtler J. (2002) CD20-negative relapse of cutaneous B-cell lymphoma after anti-CD20 monoclonal antibody therapy. J. Am. Acad. Dermatol. 46:441-443.
  • 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. and Jakobovits A. (1997) Functional transplant of megabase human immunoglobulin loci recapitulates human antibody response in mice. Nat. Genet. 15:146-56.
  • Merchant A. M., Zhu Z., Yuan J. Q., Goddard A., Adams C. W., Presta L. G. and Carter P. (1998) An efficient route to human bispecific IgG. Nat. Biotech. 16:677-681.
  • Nemazee D. (2000) Receptor editing in B cells. Adv. Immunol. 74:89-126.
  • Nissim A., Hoogenboom H. R., Tomlinson I. M., Flynn G., Midgley C., Lane D. and Winter G. (1994) Antibody fragments from a “single pot” phage display library as immunological reagents. EMBO. J. 13:692-698.
  • Nowakowski A., Wang C., Powers D. B., Amersdorfer P., Smith T. J., Montgomery V. A., Sheridan R., Blake R., Smith L. A. and Marks J. D. (2002) Potent neutralization of botulinum neurotoxin by recombinant oligoclonal antibody. Proc. Natl. Acad. Sci. U.S.A. 99:11346-11350.
  • Patel A. K. and Boyd P. N. (1995) An improved assay for antibody-dependent cellular cytotoxicity based on time resolved fluorometry. Journal of Immunological Methods 184:29-38.
  • Peeters K., De Wilde C., De Jaeger G., Angenon G. and Depicker A. (2001) Production of antibodies and antibody fragments in plants. Vaccine 19:2756-2761.
  • Pollock D. P., Kutzko J. P., Birck-Wilson E., Williams J. L., Echelard Y. and Meade H. M. (1999) Transgenic milk as a method for the production of recombinant antibodies. J. Immunol. Methods 231:147-157.
  • Radic M. C., Mascelli M. A., Shan H. and Weigert M. (1991) Ig H and L chain contributions to auto-immune specificities. J. Immunol. 146:176-182.
  • Schnieke A. E., Kind A. J., Ritchie W. A., Mycock K., Scott A. R., Ritchie M., Wilmut I., Colman A. and Campbell K. H. (1997) Human factor IX transgenic sheep produced by transfer of nuclei from transfected fetal fibroblasts. Science 278:2130-2133.
  • Segal D. M., Weiner G. J. and Weiner L. M. (2001) Introduction: bispecific antibodies. J. Immunol. Methods 248:1-6.
  • Shields R. L., Namenuk A. K., Hong K., Gloria Meng Y., Rae J., Biggs J., Xie D., Lai J., Stadlen A., Li B., Fox J. A. and Presta L. G. (2001) High resolution mapping of the binding site on human IgG1 for FcgRI, FcgRII, FcgRIII and FcRn and design of IgG1 variants with improved binding to the FcgR. J. Biol. Chem. 276:6591-6604.
  • Spiridon C. I., Ghetie M. A., Uhr J., Marches R., Li J. L., Shen G. L. and Vitetta E. S. (2002) Targeting multiple her-2 epitopes with monoclonal antibodies results in improved antigrowth activity of a human breast cancer cell line in vitro and in vivo. Clin. Cancer Res. 8:1720-1730.
  • Van der Vuurst de Vries A. and Logtenberg T. (1999) Dissecting the human peripheral B-cell compartment with phage display-derived antibodies. Immunology 98:55-62.
  • Vaughan T. J., Williams A. J., Pritchard K., Osbourn J. K., Pope A. R., Earnshaw J. C., McCafferty J., Hodits R. A., Wilton J. and Johnson K. S. (1996) Human antibodies with sub-nanomolar affinities isolated from a large non-immunized phage display library. Nat. Biotech. 14:309-314.
  • Wilmut I. and Clark A. J. (1991) Basic techniques for transgenesis. J. Reprod. Fertil. Suppl. 43:265-275.
  • Wilmut I., Schnieke A. E., McWhir J., Kind A. J. and Campbell K. H. (1997) Viable offspring derived from fetal and adult mammalian cells. Nature 385:810-813.
  • Wilson T. J. and Kola I. (2001) The LoxP/CRE system and genome modification. Methods Mol. Biol. 158:83-94.
  • Yelverton E., Norton S., Obijeski J. F. and Goeddel D. V. (1983) Rabies virus glycoprotein analogs: biosynthesis in Escherichia coli. Science 219:614-620.
  • Yoo E. M., Coloma M. J., Trinh K. R., Nguyen T. Q., Vuong L. U., Morrison S. L. and Chintalacharuvu K. R. (1999) Structural requirements for polymeric immunoglobulin assembly and association with J chain. J. Biol. Chem. 274:33771-33777.

Logtenberg, Ton, Bout, Abraham, Van Berkel, Patricius H. C., Brus, Ronald Hendrik

Patent Priority Assignee Title
10934571, Jul 18 2002 MERUS N V Recombinant production of mixtures of antibodies
Patent Priority Assignee Title
4399216, Feb 25 1980 TRUSTEES OF COLUMBIA UNIVERSITY IN THE CITY OF NEW YORK, THE Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
4599311, Aug 13 1982 ZYMOGENETICS, INC , 2121 N 35TH STREET, SEATTLE, WASHINGTON 98103 A CORP OF WASHINGTON Glycolytic promotersfor regulated protein expression: protease inhibitor
4634665, Feb 25 1980 The Trustees of Columbia University in the City of New York Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
4801687, Oct 27 1986 Bioprobe International, Inc. Monoclonal antibody purification process using protein A
4816567, Apr 08 1983 CITY OF HOPE, A CORP OF CA Recombinant immunoglobin preparations
4868103, Feb 19 1986 ENZO DIAGNOSTICS, INC , 40 OAK DRIVE, SYOSSET, NY 11791, A CORP OF NY Analyte detection by means of energy transfer
4937190, Oct 15 1987 Wisconsin Alumni Research Foundation Translation enhancer
5030002, Aug 11 1989 Becton, Dickinson and Company Method and apparatus for sorting particles with a moving catcher tube
5137809, Nov 09 1987 Becton, Dickinson and Company Method to determine the composition of bone marrow samples
5151504, Nov 17 1989 E. R. Squibb & Sons, Inc. Method for purification of monoclonal antibodies
5179017, Feb 25 1980 The Trustees of Columbia University in the City of New York Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
5223409, Sep 02 1988 Dyax Corp Directed evolution of novel binding proteins
5385839, Jan 30 1985 University of Iowa Research Foundation Transfer vectors and microorganisms containing human cytomegalovirus immediate-early promoter regulatory DNA sequence
5627037, Aug 07 1990 Becton Dickinson and Company One step method for detection and enumeration of absolute counts of one more cell populations in a sample
5631169, Jan 27 1992 LAKOWICZ, JOSEPH R Fluorescent energy transfer immunoassay
5641640, Jun 29 1992 Biacore AB Method of assaying for an analyte using surface plasmon resonance
5667988, Jan 27 1992 SCRIPPS RESEARCH INSTITUTE, THE Methods for producing antibody libraries using universal or randomized immunoglobulin light chains
5667998, Jul 30 1993 University of Medicine and Dentistry of New Jersey Efficient gene transfer into primary lymphocytes obviating the need for drug selection
5733779, Nov 03 1993 BIOGEN IDEC INC Impaired dominant selectable marker sequence and intronic insertion strategies for enhancement of expression of gene product and expression vector systems comprising same
5770429, Aug 29 1990 GENPHARM INTERNATIONAL, INC Transgenic non-human animals capable of producing heterologous antibodies
5772997, Jan 12 1988 Genentech, Inc. Monoclonal antibodies directed to the HER2 receptor
5783186, Dec 05 1995 AMGEN, INC Antibody-induced apoptosis
5789208, Jan 31 1994 The Trustees of Boston University Polyclonal antibody libraries
5789215, Aug 20 1991 GenPharm International Gene targeting in animal cells using isogenic DNA constructs
5827690, Dec 20 1993 LFB USA, INC Transgenic production of antibodies in milk
5830698, Mar 14 1997 BIOGEN IDEC INC Method for integrating genes at specific sites in mammalian cells via homologous recombination and vectors for accomplishing the same
5834237, Mar 10 1994 Transgene S.A. Combined use of two expression cassettes for the production of a protein of interest
5849500, Jul 08 1991 Deutsches Krebsforschungszentrum Stiftung Des Offentlichen Rechts Phagemid for antibody screening
5885827, Jan 23 1996 Regents of the University of California, The Eukaryotic high rate mutagenesis system
5888789, Nov 16 1993 CALIFORNIA, THE UNIVERSITY OF, REGENTS OF, THE Process for protein production in plants
5939598, Jan 12 1990 Abgenix, Inc Method of making transgenic mice lacking endogenous heavy chains
5965371, Mar 31 1993 DANA-FARBER CANCER INSTITUTE, INC Method of intracellular binding of target molecules
6004940, Jul 17 1992 Dana-Farber Cancer Institute Intracellular targeting of endogenous proteins
6054297, Jun 14 1991 Genentech, Inc. Humanized antibodies and methods for making them
6080560, Jul 25 1994 MONSANTO TECHNOLOGY LLC Method for producing antibodies in plant cells
6114598, Mar 15 1993 Abgenix, Inc Generation of xenogeneic antibodies
6180357, Oct 08 1999 Hoffman-La Roche Inc; Hoffmann-La Roche Inc Individualized patient-specific anti-cancer antibodies
6207446, Nov 06 1997 ADNEXUS THERAPEUTICS, INC Selection of proteins using RNA-protein fusions
6265150, Jun 07 1995 CRUCELL HOLLAND B V Phage antibodies
6291740, Mar 20 1997 Wisconsin Alumni Research Foundation Transgenic animals
6303341, Dec 30 1994 Planet Biotechnology, Inc. Method for producing immunoglobulins containing protection proteins in plants and their use
6335163, Jan 31 1994 The Trustees of Boston University Polyclonal antibody libraries
6570061, Aug 25 1992 KOLNER VEREIN ZURFORDERUNG DER IMMUNOLOGIE Targeted replacement of an immunoglobulin gene without endogenous and selectable residual sequences in mice
6586251, Oct 31 2000 REGENERON PHARMACEUTICALS, INC Methods of modifying eukaryotic cells
6596541, Oct 31 2000 REGENERON PHARMACEUTICALS, INC Methods of modifying eukaryotic cells
7067284, Jan 27 1992 The Scripps Research Institute Methods for producing antibody libraries using universal or randomized immunoglobulin light chains
7105348, Oct 31 2000 REGENERON PHARMACEUTICALS, INC Methods of modifying eukaryotic cells
7183076, May 02 1997 Genentech, Inc. Method for making multispecific antibodies having heteromultimeric and common components
7262028, Jul 18 2002 MERUS N V Recombinant production of mixtures of antibodies
7329530, Jun 15 2001 CRUCELL HOLLAND B V Chimaeric phages
7429486, Jul 18 2002 MERUS N V Recombinant production of mixtures of antibodies
7491516, Jun 29 2000 AbbVie Inc Dual specificity antibodies and methods of making and using
7579446, May 27 2004 Crucell Holland B.V. Binding molecules capable of neutralizing rabies virus and uses thereof
7642228, Mar 01 1995 Genentech, Inc. Method for making heteromultimeric polypeptides
7696330, Jul 22 2003 Crucell Holland B.V. Binding molecules against SARS-coronavirus and uses thereof
7740852, May 27 2004 Crucell Holland B.V. Binding molecules capable of neutralizing rabies virus and uses thereof
7777010, Dec 27 1999 CRUCELL HOLLAND B V Use of a native epitope for selecting evolved binding members from a library of mutants of a protein capable of binding to said epitope
7858086, Oct 12 2004 Crucell Holland B.V.; CRUCELL HOLLAND B V Binding molecules for treatment and detection of cancer
7901919, Jun 15 2001 Crucell Holland B.V. Chimaeric phages
7919257, May 30 2003 MERUS N V Method for selecting a single cell expressing a heterogeneous combination of antibodies
7927834, Jul 18 2002 MERUS N V Recombinant production of mixtures of antibodies
7932360, Jul 18 2002 MERUS N V Recombinant production of mixtures of antibodies
7960518, Jun 06 2006 Crucell Holland B.V.; CRUCELL HOLLAND B V Human binding molecules having killing activity against enterococci and uses thereof
7968092, Dec 23 2003 Johns Hopkins University, The Human binding molecule against CD1a
8052974, May 12 2005 Crucell Holland B.V.; CRUCELL HOLLAND B V Host cell specific binding molecules capable of neutralizing viruses and uses thereof
8106170, Nov 11 2004 Crucell Holland B.V.; CRUCELL HOLLAND B V Compositions against SARS-coronavirus and uses thereof
8148497, May 27 2004 Crucell Holland B.V. Binding molecules capable of neutralizing rabies virus and uses thereof
8192927, Sep 07 2006 Crucell Holland B.V.; CRUCELL HOLLAND B V Human bind molecules capable of neutralizing influenza virus h5n1 and uses thereof
8211431, Jun 06 2006 Crucell Holland B.V.; CRUCELL HOLLAND B V Human binding molecules having killing activity against staphylococci and uses thereof
8241631, Jun 06 2006 Crucell Holland B.V. Human binding molecules having killing activity against enterococci and uses thereof
8268756, Jan 20 2004 MERUS N V Mixture of binding proteins
8470327, May 11 2009 JANSSEN VACCINES & PREVENTION B V Human binding molecules capable of neutralizing influenza virus H3N2 and uses thereof
8592562, Jan 07 2008 Amgen Inc Method for making antibody Fc-heterodimeric molecules using electrostatic steering effects
8911738, May 12 2005 Crucell Holland B.V. Host cell specific binding molecules capable of neutralizing viruses and uses thereof
9012371, Jan 20 2004 MERUS N V Mixtures of binding proteins
9248181, Apr 20 2012 MERUS N V Methods and means for the production of Ig-like molecules
9248182, Apr 20 2012 MERUS N V Methods and means for the production of Ig-like molecules
9303081, Jul 18 2002 MERUS N V Recombinant production of mixtures of antibodies
9358286, Apr 20 2012 MERUS N V Methods and means for the production of IG-like molecules
9738701, May 30 2003 MERUS N V Method for selecting a single cell expressing a heterogeneous combination of antibodies
20020088016,
20020138857,
20030039958,
20030077739,
20030091561,
20030093820,
20030096225,
20030194403,
20030207346,
20030215914,
20030219829,
20030224408,
20050014261,
20050037001,
20050037427,
20050170398,
20060015949,
20060015957,
20060088520,
20060117699,
20060160184,
20060177437,
20060205077,
20060257397,
20060292634,
20070054362,
20070059766,
20070178552,
20070280945,
20080070799,
20080241166,
20090017521,
20090054254,
20090130652,
20090181855,
20090182127,
20090263864,
20100015133,
20100069614,
20100146647,
20100172917,
20100286374,
20100297153,
20100310572,
20100310586,
20100331527,
20110177073,
20110195454,
20110268739,
20120021409,
20120039898,
20120058907,
20120076794,
20120093823,
20120141493,
20120177637,
20120192300,
20120276115,
20120315278,
20130115208,
20130145484,
20130336981,
20140072579,
20140120096,
20140140999,
20140314755,
20140317766,
20150139996,
20150196637,
20160238600,
20160319320,
20180094289,
20180112247,
AU2003250074,
CA1341364,
CA2114353,
CA2405961,
CA2445255,
EP120694,
EP314161,
EP402029,
EP445625,
EP469025,
EP469897,
EP481790,
EP523949,
EP724639,
EP814159,
EP1325932,
EP1349234,
EP1399575,
EP1439234,
EP171142,
EP1870459,
EP2147594,
EP469025,
EP666868,
FR2817875,
JP2001523971,
JP2004008214,
JP2004524841,
JP20048218,
JP2006109711,
JP2006515503,
JP2008538912,
JP2010505418,
JP2010512749,
JP2011508604,
JP2011525808,
JP2013004215,
JP568599,
JP5749161,
JP8116978,
RU2236127,
WO48306,
WO63403,
WO70023,
WO71694,
WO76310,
WO100245,
WO119394,
WO127279,
WO132901,
WO148485,
WO164929,
WO188132,
WO2066630,
WO2074969,
WO2096948,
WO218948,
WO243478,
WO246233,
WO3002609,
WO3004704,
WO3016501,
WO3046560,
WO3102157,
WO3106684,
WO188132,
WO1999050657,
WO2004003211,
WO2004009618,
WO2004061104,
WO2004106375,
WO2005068622,
WO2005118635,
WO2006028936,
WO2006106905,
WO2006117699,
WO2007110205,
WO2007117410,
WO2007147901,
WO2008054606,
WO2008076379,
WO2008119353,
WO2009051974,
WO2009080251,
WO2009080252,
WO2009080253,
WO2009089004,
WO2009098596,
WO2009157771,
WO2010084197,
WO2010129304,
WO2011028952,
WO2011028953,
WO2011097603,
WO2011143545,
WO2012020096,
WO2012023053,
WO2012058768,
WO2012131555,
WO2012141798,
WO2059297,
WO236789,
WO3033670,
WO3106674,
WO44777,
WO9002809,
WO9004036,
WO9012878,
WO9100906,
WO9108216,
WO9117271,
WO9201047,
WO9203918,
WO9209690,
WO9215679,
WO9218619,
WO9220791,
WO9301288,
WO9312227,
WO9402602,
WO9402610,
WO9404667,
WO9423046,
WO9425591,
WO9517085,
WO9517500,
WO9520401,
WO9627011,
WO9630498,
WO9742313,
WO9747739,
WO9815627,
WO9815833,
WO9824893,
WO9824923,
WO9839416,
WO9841645,
WO9850431,
WO9852976,
WO99156894,
WO9920749,
WO9923221,
WO9926569,
WO9936569,
WO9945962,
WO9964582,
WO71694,
WO76310,
WO2074969,
WO2096948,
WO243478,
WO246233,
WO30026909,
WO3004704,
WO30102157,
WO3016501,
WO3046560,
WO3048306,
WO3106684,
WO2005068622,
WO9002809,
WO9926569,
/////////
Executed onAssignorAssigneeConveyanceFrameReelDoc
Nov 29 2004BOUT, ABRAHAMCRUCELL HOLLAND B V ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0387070688 pdf
Nov 29 2004BRUS, RONALD H P CRUCELL HOLLAND B V ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0387070688 pdf
Nov 30 2004VAN BERKEL, PATRICIUS H C CRUCELL HOLLAND B V ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0387070688 pdf
Dec 06 2004LOGTENBERG, TONCRUCELL HOLLAND B V ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0387070688 pdf
Jan 23 2009CRUCELL HOLLAND B V MERUS B V ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0387080444 pdf
May 18 2016Merus N.V.(assignment on the face of the patent)
May 20 2016MERUS B V MERUS N V CHANGE OF NAME SEE DOCUMENT FOR DETAILS 0388530599 pdf
Feb 10 2017MERUS N V MERUS N V CHANGE OF ADDRESS0416750842 pdf
Feb 10 2017MERUS N V MERUS N V CORRECTIVE ASSIGNMENT TO CORRECT THE ASSIGNEE ADDRESS PREVIOUSLY RECORDED AT REEL: 041675 FRAME: 0842 ASSIGNOR S HEREBY CONFIRMS THE CHANGE ADDRESS 0423220304 pdf
Date Maintenance Fee Events
Oct 05 2018BIG: Entity status set to Undiscounted (note the period is included in the code).
Nov 27 2023REM: Maintenance Fee Reminder Mailed.
May 13 2024EXP: Patent Expired for Failure to Pay Maintenance Fees.


Date Maintenance Schedule
Dec 17 20224 years fee payment window open
Jun 17 20236 months grace period start (w surcharge)
Dec 17 2023patent expiry (for year 4)
Dec 17 20252 years to revive unintentionally abandoned end. (for year 4)
Dec 17 20268 years fee payment window open
Jun 17 20276 months grace period start (w surcharge)
Dec 17 2027patent expiry (for year 8)
Dec 17 20292 years to revive unintentionally abandoned end. (for year 8)
Dec 17 203012 years fee payment window open
Jun 17 20316 months grace period start (w surcharge)
Dec 17 2031patent expiry (for year 12)
Dec 17 20332 years to revive unintentionally abandoned end. (for year 12)