Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
CHARACTERIZATION OF CHO-MIF GENE AND PROTEIN, AND USE THEREOF
Document Type and Number:
WIPO Patent Application WO/2013/050457
Kind Code:
A1
Abstract:
The present invention is concerned with the specific and highly sensitive detection of specific CHO-MIF (macrophage migration inhibitory factor from Chinese Ovarian Hamster cell line) complexes in the production of anti-MIF antibodies. The present invention is further concerned with the provision of specific antibodies which can be used for a CHO-MIF detection method. The present invention is also concerned with a CHO MIF knockout cell line and use thereof. The present invention also provides preparations obtained from recombinant production in CHO cell lines which are essentially free of CHO-MIF.

Inventors:
KERSCHBAUMER RANDOLF J (AT)
VOELKEL DIRK (AT)
ANTOINE GERHARD (AT)
SCHEIFLINGER FRITZ (AT)
MUDDE C GEERT (AT)
Application Number:
PCT/EP2012/069602
Publication Date:
April 11, 2013
Filing Date:
October 04, 2012
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BAXTER HEALTHCARE SA (CH)
BAXTER INT (US)
International Classes:
G01N33/68; C07K16/24; C12N5/10
Domestic Patent References:
WO2009086920A12009-07-16
WO2001064749A22001-09-07
WO2009086920A12009-07-16
Foreign References:
US6030615A2000-02-29
US6645493B12003-11-11
US20030235584A12003-12-25
Other References:
BLOOM ET AL., SCIENCE, vol. 153, 1966, pages 80 - 2
DAVID ET AL., PNAS, vol. 56, 1966, pages 72 - 7
WEISER ET AL., PNAS, vol. 86, 1989, pages 7522 - 6
SUN ET AL., PNAS, vol. 93, 1996, pages 5191 - 5196
CALANDRA ET AL., NATURE, vol. 377, 1995, pages 68 - 71
BAUGH ET AL., CRIT CARE MED, vol. 30, 2002, pages 27 - 35
MITCHELL, R.A., CELLULAR SIGNALLING, vol. 16, no. 1, 2004, pages 13 - 19
LUE, H. ET AL., ONCOGENE, vol. 26, no. 35, 2007, pages 5046 - 59
NISHIHIRA ET AL., J INTERFERON CYTOKINE RES., vol. 20, 2000, pages 751 - 62
SHIMIZU ET AL., FEBS LETT., vol. 381, 1996, pages 199 - 202
KAWAGUCHI ET AL., LEUKOC. BIOL., vol. 39, 1986, pages 223 - 232
WEISER ET AL., CELL. IMMUNOL., vol. 90, 1985, pages 16778
CALANDRA ET AL., J. INFLAMM., vol. 47, 1995, pages 39 - 51
GALAT ET AL., EUR. J. BIOCHEM, vol. 224, 1994, pages 417 - 21
WATARAI ET AL., PNAS, vol. 97, 2000, pages 13251 - 6
HARLOW; LANE: "Antibodies, A Laboratory Manual", 1988, COL SPRING HARBOUR PUBLICATIONS
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY PRESS
AUSUBEL ET AL.: "Current Protocols in Molecular Biology", 1992, GREENE PUBLISHING ASSOCIATES
HARLOW; LANE: "Antibodies; A Laboratory Manual", 1990, COLD SPRING HARBOR LABORATORY PRESS
KABAT: "Sequences of Proteins of Immunological Interest", 1987, NATIONAL INSTITUTES OF HEALTH
CHOTHIA ET AL., J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
CHOTHIA ET AL., NATURE, vol. 342, 1989, pages 878 - 883
ANGAL ET AL., MOL IMMUNOL., vol. 30, 1993, pages 105 - 108
"Bioanalytik", 1998, SPEKTRUM AKADEMISCHER VERLAG
Attorney, Agent or Firm:
ENGELHARD, Elisabeth et al. (Arabellastrasse 4, Munich, DE)
Download PDF:
Claims:
CLAIMS

1. A method for the detection of CHO-MIF contaminations in a monoclonal anti-MIP antibody preparation, comprising the s ep of

contacting the anti-M!F antibody preparation with a polyclonal anti-CHO-MIF antibody, affinity purified against CHO-MIF.

2. The method according to claim 1 wherein the CHO-MIF

contaminates a final CHO cell produced monoclonal anti- MIF antibody - preparation or a preparation of antigen- binding fragments thereof .

3. The method according to claim 1 and/or 2 wherein the

CHO-MIF is endogenous CHO-MIF produced by CHO cells.

4. The method according to any one or more of claims 1 to 3 wherein the detection step is carried out by a semiquantitative Western Blot analysis,

5. Use of a rabbit anti-CHO-MIF antibody, affinity purified against CHO-MIF for the detection of CHO-MIF

contaminations during production of monoclonal anti-MIF antibodies or antigen-binding fragments thereof or in the final preparation of monoclonal anti-MIF antibody or antigen-binding fragments thereof .

6. The use according to claim 5 wherein the detection step is carried out as a semi -quantitative Western Blot

analysis .

7. A method for the production of anti-MIF antibodies or antigen-binding fragments thereof in CHO cells, wherein said antibodies or antigen-binding fragments thereof are essentially free of CHO-MIF, wherein a detection method as defined in any one of claims 1 to 6 is carried out .

8. A CHO-MIF knock-out cell line,

3, Ά CHO-MIF knock-out cell line, wherein said CHO MIF

knock-out cell line comprises any one or more of the following plasmids: DSM 25110, DSM 25112, DSM 25111, DSM 25113, DSM 25114, DSM 25115, DSM 25853, DSM 25860, DSM 25861, DSM 25862, DSM 258S3 and DSM 25864,

10. The CHO-MIF knock-out CHO cell line according to claim 8 or 9, wherein the ceil line comprises the plasmids DSM

25110 and DSM 25112 or DSM 25861 and DSM 25862.

11. The CHO-MIF knock-out CHO cell line according to claim 8 or 3, wherein the cell line comprises the plasmids DSM

25111 and DSM 25113 or DSM 25859 and DSM 25860.

12. The CHO-MIF knock-out CHO cell line according to claim 8 or 9, wherein the cell line comprises the plasmids DSM 25114 and DSM 25115 or DSM 25863 and DSM 25864.

13. The use of the CHO-MIF knock-out CHO ceil line of any of claims 8 to 12 for the production of a preparation of monoclonal anti-MIF antibodies or binding fragments thereof , preferably for the production of any one of antibodies RAB0, RAB4 , RAMB9, RAM0 , RA 4 or RAM9.

14. Essentially CHO MIF free anti-MIF antibody preparation as obtainable by the method of claim 7, or by use of the CHO-MIF knock-out CHO cell line of any of claims 8 to

12.

15. A method for the production of an essentially CHO MIF free anti-MIF antibody preparation which is

characterized by the use of the CHO-MIF knock-out CHO ceil line according to any one of claims 8 - 12.

16. Preparation of a recombinant MIF binding protein, preferably an (h) MIF binding protein, like an (h) MIF binding peptide, ligand, agonist, antagonist, inhibitor, or a MIF receptor fragment, or an anti- (h) MIF antibody preparation, produced in a CHO cell line, characterized in that said preparation is essentially free of CHO-MIF.

17. Preparation of a recombinant MIF binding protein,

preferably an (h) MIF binding protein, like an (h)MIF binding peptide , ligand, agonist, antagonist, inhibitor, or a MIF receptor fragment, or an anti- (h) IF antibody preparation, which is obtainable by the method according to claim 15 or by a method of production which

comprises, preferably as a quality control step, the method of detection of any one of the above claims 1 - 4 or 7.

18, Preparation of a recombinant MIF binding protein,

preferably an (h) MIF binding protein, like an (h)MIF binding peptide, ligand, agonist, antagonist, inhibitor, or a MIF receptor fragment, or an anti - (h) MIF antibody preparation, produced by the method according to claim

15 or by a method of production which comprises, preferably as a quality control step, the method of detection of any one of the above claims 1 - 4 or 7.

19. The anti-hMIF antibody preparation of any of claims

16 - 18 above, which is essentially free of CHO MIF, wherein the anti-hMIF antibody is selected from the group of RAB4 , RABO , RA39 , RAM , RAMO and/or RAM9.

Description:
CHARACTERIZATION OF CHO-MIF GENE AMD PROTEIN, AND USE THEREOF

The present invention is based on the identification and characterization of the CHO-MIF gene. This allowed the

provision of CHO-MIF knock-out cells and formed the basis of a highly sensitive detection method of specific CHO-MIF complexes, particularly in the production of anti-MIF

antibodies . The present invention is further concerned with the provision of an advantageous polyclonal rabbit antiserum which can be used for a CHO-MIF detection method.

Furthermore, a method is shown, to avoid any contaminations of anti-MIF antibodies with CHO-MIF by knocking out the endogenous gene in CHO cells.

BACKGROUND

Macrophage migration inhibitory factor (MIF) is a cytokine initially isolated based upon its ability to inhibit the in vitro random migration of peritoneal exudate cells from

tuberculin hypersensitive guinea pigs (containing

macrophages) (Bloom et al . Science 1366, 153, 80-2; David et al . PNAS 1966, 56, 72-7) . Today, MIF is known as a critical upstream regulator of the innate and acquired immune response that exerts a pleiotropic spectrum of activities .

The human MIF cDNA was cloned in 1989 (Weiser et al . , PNAS 1989, 86, 7522-6) , and its genomic localization was mapped to chromosome 22. The product of the human MIF gene is a protein with 114 amino acids (after cleavage of the N-terminal methionine) and an apparent molecular mass of about 12.5 kDa . MIF has no significant sequence homology to any other

protein. The protein crystallizes as a trimer of identical subunits . Each monomer contains two antiparallel alpha- helices that pack against a four-stranded beta-sheet. The monomer has additional two beta-strands that interact with the beta- sheets of adjacent subunits to form the interface between monomers . The three subunits are arranged to form a barrel containing a solvent-accessible channel that runs through the center of the protein along a molecular threefold axis {Sun et al . PNAS 1996, 93, 5191-5196),

It was reported that MIF secretion from macrophages was

induced at very low concentrations of glucocorticoids

(Caiandra et al. Nature 1995, 377 , 68-71} . However, MIF also counter-regulates the effects of glucocorticoids and

stimulates the secretion of other cytokines such as tumor necrosis factor T F- and interleukin IL-1 β (Baugh et al . , Crit Care Med 2002 , 30, S27-35). MIF was also shown e.g. to exhibit pro-angiogenic , pro-proiiferative and anti-apoptotic properties, thereby promoting tumor cell growth (Mitchell, R . A . , Cellular Signalling, 2004. 16{1) : p. 13-19; Lue, H. et al . , Oncogene 2007. 26 (35) : p. 5046-59) . It is also e.g. directly associated with the growth of lymphoma, melanoma, and colon cancer (Nishihira et al . J Interferon Cytokine Res. 2000, 20 : 751-62) .

MIF is a mediator of many pathologic conditions and thus associated with a variety of diseases including inter alia inflammatory bowel disease (IBD) , rheumatoid arthritis (RA) , acute respiratory distress syndrome (ARDS) , asthma,

glomerulonephritis, IgA nephropathy, myocardial infarction (MI), sepsis and cancer, though not limited thereto.

Polyclonal and monoclonal anti-MIP antibodies have been developed against recombinant human MIF (Shimizu et al . , FEES Lett. 1996; 381, 199-202; Kawaguchi et al, Leukoc . Biol.

1986, 39, 223-232, and Weiser et al . , Cell. Immunol. 1985, 90, 16778} .

Anti-MIF antibodies have been suggested for therapeutic use. Caiandra et al . , (J. Inflamm. 1995. 47, 39-51) reportedly used anti-MIF antibodies to protect animals from experimentally induced gram-negative and gram-positive septic shock, Anti-MIF antibodies were suggested as a means of therapy to modulate cytokine production in septic shock and other inflammatory disease states.

US 6,645,493 discloses monoclonal anti-MIF antibodies derived from hybridoma cells, which neutralize the biological activity of MIF. It could be shown in an animal model that these mouse-derived anti-MIF antibodies had a beneficial effect in the treatment of endotoxin induced shock.

US 200310235584 discloses methods of preparing high affinity antibodies to MIF in animals in which the MIF gene has been horaozygously knocked=out.

Glycosylation- inhibiting factor (GIF) is a protein described by Galat et al . {Eur. J, Biochem, 1994, 224, 417-21). MIF and GIF are now recognized to be identical. Watarai et al . ( P AS 2000 , 97, 13251-6} described polyclonal antibodies binding to different GIF epitopes to identify the biochemical nature of the posttranslational modification of GIF in Ts cells.

In vie of the clear biological significance of MIF/GIF . , it is therefore necessary and would be highly desirable to

provide purified anti-MIF antibodies as diagnostic and

therapeutic tools.

Clearly, therefore a need exists for the production of anti- MIF antibodies, whereby these are free from contaminations.

Various methods for the production of anti-MIF antibodies are currently available. One major approach is to use the recombinant production of anti-MIF antibodies whereby a host cell expresses the desired anti-MIF antibody product. Chinese hamster ovary (CHO) cells are a cell line derived from the ovary of the Chinese hamster (Cricetulus griseus) . They are frequently and broadly used in biological and medical research production of therapeutic proteins, e.g. antibodies .

Today, CHO ceils are the most commonly used mammalian hosts for industrial production of recombinant protein

therapeutics, including antibodies.

CHO cells have been a cell line of choice because of their rapid growth and high protein production . They have become the mammalian equivalent of E. coli i research and

biotechnology today, especially when long-term, stable gene expression and high yields of proteins are required.

However, the present inventors, upon investigation of a possible preferable production and purification process of anti-MIF antibodies with the use of CHO cells as host cells discovered that CHO cells themselves produce MIF. This is surprisingly different from the situation e.g. when preparing MIF from hybridoma cells or in the preparation of polyclonal antisera where no such or corresponding contaminations are found. The MIF as produced by CHO cells is a Chinese hamster MIF, due to the fact that CHO cells are derived from ovary ceils of a Chinese hamster. This "Chinese hamster-MIF" (in the following and above also designated as "CHO-MIF" ) , possibly because of the high homology between CHO-MIF and other, e.g. human, MIF also binds to the anti-MIF antibodies to be produced. Thus , endogenous CHO-MIF could possibly contaminate the final CHO-cell based preparations of

antibodies directed to non-CHO-MIF (e.g. complexed to the desired anti-MIF antibodies) , like e.g. human MIF, or mouse MIF.

Therefore, t ere exists a need fo the provison of a ceil line which does not produce possibly contaminating CHO-MIF ; a further need exists for a sensitive method to detect minor amounts of CHO-MIF contaminations in preparations of ant i -MIF antibodies produced in CHO ceils producing the CHO -MIF and a specific method for the production and purification of such anti -MIF antibody preparations which are not contaminated by CHO -MIF . As a prerequisite for both the provision of an essentially CHO-MIF free CHO cell line and for developing a sensitive detection method for potential CHO-MIF

contaminations, there exists a need to identify and

characterize the CHO-MIF gene as a starting point for solving the problems mentioned above.

There also exists a need for such a CHO-MIP cell line which provides similar growth and production characteristics as the wild type CHO cell line .

DESCRIPTION OF THE INVENTION '

The present inventors have succeeded in identifying and characterizing the CHO-MIF gene. On that basis they further succeeded in the provision of tools and methods allowing production and testing of ant i -MIF antibody preparations in CHO cells, which preparations are essentially free of

contaminating CHO-MIF. These tools and methods further allowed production and testing of all recombinant

preparations as produced in CHO cells, which comprise

recombinant CHO-MIF-binding protein, whereupon these

preparations are essentially free of contaminating CHO-MIF . A recombinant CHO MIF binding protein in that context is a protein which binds to CHO MIF; thus , the protein binds to CHO MIF under immunoassay conditions , whereby a varie y of immunoassay formats can be used to determine this binding, as is well known to a person skilled in the art. For example, solid phase ELISA immunoassays are routinely used to

determine such binding reactions; see Harlow and Lane (1988) , Antibodies, A Laboratory Manual, Col Spring harbour publications, New York, for a description of immunoassay formats and conditions that can be used .

Thus , the present invention is directed to the analysis of the gene locus coding for CHO-MIF, This allows the generation of MIF knockout CHO cells producing recombinant antibodies or other products, preferably antibodies directed towards human MIF, essentially without any CHO-MIF contaminants.

The present invention is further directed to a knock-out cell line wherein the CHO-MIF gene is successfully knocked out .

The present invention is also directed to a highly sensitive method for the detection of ppm levels of CHO-MIF which during production in CHO cells of products, in particular antibodies and even more preferred anti -MIF antibodies or antigen-binding fragments thereof, can remain attached to the desired product in some cases . In a preferred embodiment, this detection method is based on the generation and

purification of highly specific anti CHO-MIF antibodies which are affinity-purified polyclonal rabbit antibodies.

Only with a detection method, as described in the present invention; which is able to detect CHO-MIF in very minor amounts, it can be ensured that a final preparation is pure and in particular free of CHO-MIF. Thereby, the present inventors succeeded in providing a recombinant product preparation, produced in CHO cells .. comprising a product which would bind to CHO-MIF, in the event that CHO-MIF was present , wherein said preparation is essentially free o CHO- MIF. Preferably, the product as produced in the CHO cells is an antibody, more preferred an anti-MIF antibody, very preferred an anti human MIF antibody. Preferred embodiments of these anti human IF antibodies are described below and are designated as RAB4, RABO , RAB3 , RAM4, RA G , and RAM9 respectively.

The present invention thus provides a recombinant

preparation, as defined above, which satisfies quality control requirements, in particular with regard to the essential absence of CHO-MIF contaminations.

The present i ven ion is further directed to the isolation of mRNA coding for CHO-MIF as produced by CHO cells. According to the invention, the cDNA created by reverse transcription of this mRNA is cloned into a prokaryotic expression vector. The CHO-MIF protein expressed thereo in E . coli is purified to homogeneity. The recombinant CHO-MIF is used to immunize rabbits in order to generate the inventive polyclonal rabbit antibodies specific to CHO-MIF.

DETAILED DESCRIPTION OF THE INVENTION

Definitions and General Techniques

Unless otherwise defined herein, scientific and technical terms used in connection wi the present invention sh ll ha e the meanings tha are commonly understood by those of ordinary skill in the art . Generally, nomenclatures used in connection with, and techniques of , cell and tissue culture , molecular biology, immunology, microbiology, genetics and protein and nucleic acid chemistry described herein are those well known and commonly used in the art. The methods and techniques of the present invention are generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present

specification unless otherwise indicated. See, e.g. , Sarabrook e al . , Molecular Cloning: A Laboratory Manual, 2nd ed. , Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.

(1989) and Ausubei et al . , Current Protocols in Molecular Biology, Greene Publishing Associates (1992) , and Harlow and Lane Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y, (1990} , which are incorporated herein by reference.

"MIF" or "macrophage migx-ation inhibitory factor" refers to the protein, which is known as a critical mediator in the immune and inflammatory response, especially as a counter- regulator of glucocorticoids. MIF includes mammalian MIF, specifically human MIF ( Swiss -Prot primary accession number: P14174) , wherein the monomeric form is encoded as a 115 amino acid protein but is produced as a 114 amino acid protein due to cleavage of the initial methionine. "MIF" also includes what was formerly known as "GIF" (glycosylation- inhibiting factor) .

Also known are MIF derivatives/fragments , which exhibit

functional or immunological properties of MIF, such as e.g. fragments or fusion proteins of MIF.

An "antibody" in this application refers to an intact

antibody or an antigen-binding portion that competes with the intact antibody for specific binding. See generally,

Fundamental Immunology, Ch. 7 (Paul, W. , ed. , 2nd ed. Raven Press, N.Y. (1989)) (incorporated by reference) . The term antibody includes human antibodies, mammalian antibodies, isolated antibodies and genetically engineered forms such as, but not limited to, chimeric, camelized or humanized

antibodies .

The term "antigen-binding portion" of an antibody refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g. MIF) . Antigen- binding portions may be produced by recombinant DMA

techniques or by enzymatic or chemical cleavage of intact antibodies. Antigen-binding portions include Fab, Fab',

F ( ab 1 ) 2 , Fv, and complementarity determining regions (CDR) and fragments thereof, single-chain antibodies (scFv) ,

chimeric antibodies, antibodies and polypeptides, that contain at least a portion of an antibody that is sufficient to confer specific antigen binding to the polypeptide. From K- terminus to C~ terminus, both the mature light and heavy chain variable domains comprise the regions FR1, CDRl , FR2, CDR2, FR3 , CDR3 and FR4. The assignment of amino acids to each domain is in accordance with the definitions of Rabat, Sequences of Proteins of Immunological Interest (National Institutes of Health, Be hesda, Md . (1987 and 1991}} , Chothia et al. J- Mol. Biol. 196:901-917 (1987) , or Chothia et al . , Nature 342:878-833 (1989) . An antibody or antigen-binding portion thereof can be derivatized or linked to another functional molecule (e.g. another peptide or protein) . For example, an antibody or antigen- binding portion thereof can be functionally linked to one or more other molecular entities, such as another antibody (e.g. a bispecif ic antibody or a diabody) , a detectable agent, a cytotoxic agent, a pharmaceutical agent, and/or a linking molecule.

The term "human antibody" refers to any antibody in which the variable and constant domains are human sequences . The term encompasses antibodies with sequences derived from human genes, but which have been changed, e.g. to decrease possible immunogenicity , increase affinity, eliminate cysteines that might cause undesirable folding, etc . The term encompasses such antibodies produced recombinan ly in non-human cells, which might impart glycosylation not typical of human cells.

The term "humanized antibody" refers to antibodies comprising human sequences and containing additionally non-human

sequences .

The term " camel i zed antibody" refers to antibodies wherein the antibody structure or sequence has been changed to more closely resemble antibodies from camels, also designated camelid antibodies . Methods for the design and production of camelized antibodies are part of the general knowledge of a person skilled in the art .

The term "chimeric antibody" refers to an antibody that comprises regions from two or more different species.

The term "isolated antibody" or "isolated antigen-binding portion thereof" refers to an antibody or an antigen-binding portion thereof that has been identified and selected from an antibody source such as a phage display library or a B-ceil repertoire and has then been e.g. recombinantly prepared.

The term "polyclonal antibody" refers to a polyclonal

antibody preparation, which may be a purified or partially purified polyclonal antibody fraction or which may be used in form of a crude serum from an animal immunized with the respective antigen, e.g. purified CHG-MIF.

The term "!¾" refers to the equilibrium dissociation constant of a Fab portion of a particular antibody with the respective antigen.

The term "epitope" includes any protein determinant capable of specific binding to an immunoglobulin or an antibody fragment. Epitopic determinants usually consist of chemically active surface groupings of molecules such as exposed amino acids, amino sugars, or other carbohydrate side chains and usually have specific three-dimensional structural

characteristics, as well as specific charge characteristics.

The term "vector" refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. In some embodiments, the vector is a plasmid, i.e., a circular double stranded DNA loop into which additional DNA segments may be iigated. The term "host cell" refers to a cell line, which is capable to produce a recombinant protein after introducing an

expression vector. The term "recombinant cell line" refers to a cell line into which a recombinant expression vector has been introduced . It should be understood that "recombinant cell line" does not only mean the particular subject ceil line but also the progeny of such a cell line. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may no , in fact , be identical to the parent cell, but is still included within the scope of the term "recombinant cell lin " as used he ein . The host cell as used according to the present invention is a CHO cell line.

The term "Western Blot" refers to the well-known and

established technique of blotting proteins on a carrier membrane whereupon these proteins can subsequently be

detected . The transfer to the membrane is carried out by well-known methods, of which diffusion, application of

capillary forces or electrophoresis are examples, which however are by no means limiting the present method. In the case of an immunoblot, the detection is carried out by use of monoclonal or polyclonal antibodies, A " semi -quantitative" Western Blot in the context of the present invention means a Western Blot where the signal intensity from a sample (e.g. CHO-MIF which can in some cases appear in com lex with an anti-MIF antibody) is compared with the signal intensity from the corresponding standards (e.g. CHO-MIF) , The signal can be e.g. a chemiluminescent signal quantified e.g. electronically by digital imaging systems .

BRIEF DESCRIPTION OF THE FIGURES

The present invention is further described in the following figures : Figure 1; describes the nucleotide and amino acid sequence of the CHO-MIF coding region. Lane 1 shows the number of the amino acids starting with the ATG as position +1. Lane 2 depicts the names of the amino acids. Lane 3 shows the DNA sequence triplets coding for these amino acids. Lane 4 shows the numbering of the base pairs. The total length of CHO-MIF is 115 amino acids translated from 345 base pairs .

Figure 2: shows the organization of the CHO-MIF locus. The genomic DNA (lane 2: gDNA, SEQ ID NO: 14 ) is organized in 3 exons, separated by two introns as explained in lane 1. The cDNA (lane 3 , SEQ ID NOs : 15 ( exon 1) , 16 ( exon 2 ) and 17 (exon 3 ) ) is translated into the CHO-MIF protein sequence shown in lane 4 (SEQ ID NOs : 18 (exon 1) , 19 {exon 2) and 20 (exon 3)) , The 3' -untranslated region of the cDNA is shown after the trans lac lonal stop codon TGA to the polyA tail . The translated parts of the gene are framed.

Figure 3 : is a schematic drawing of the the E.coli expression plasmid pETchoMIF 762-25 based on the pET19b vector (Novagen) . The complete cDNA of CHO-MIF is inserted behind the T7 promoter and transcribed by the T7 polymerase, which is part of the E.coli BL21 host strain.

Figure 4 : is a Western Blot of CHO-MIF detection from ant i - MIF antibodies resultant from different downstream process steps. The Blot was detected by the specific affinity purified rabbit anti CHO-MIF antibody and a commercial available horse radish peroxidase conjugated donkey anti rabbit IgG. Lane 1: molecular weight protein marker; Lane 2: 50 μ of anti-MIF antibody fractions before removal of CHO-MIF impurity; Lane 3-5; each 50 ^g of anti-MIF antibody fractions from different downstream process steps; Lane 6: 50 jug of anti-MIF antibody fraction after removal of CHO-MIF impurities; Lane 7-10: recombinant CHO-MIF reference 4 ng, 2 ng, 1 ng and 0.5 ng/Iane.

Figure 4a: is a bar chart of the CHO-MIF protein signals

resultant from a Western Blot as shown in figure 4. To that end, the CHO-MIF signals were scanned by a LAS4000 (Fujifilm Life Science*} using the Image Reader LAS4000 scanner software '" and directly quantified by the Image Quant LAS4000* software. The 4 ng CHO-MIF signal from lane 7 was set to 100% and directly compared to the other CHO-MIF signals; 189% signal intensity was found for the CHO-MIF impurity in 50 p.g anti MIF antibodies resultant after the first step of the downstream process as shown in lane/bar 2; 12% signal intensity was found for the CHO-MIF impurity in 50 ^g anti M1P antibodies resultant after the second step of the downstream process as shown in lane/bar 3; 11% signal intensity was found for the CHO-MIF impurity in 50 anti MIF antibodies resultant after the third step of the downstream process as shown in lane/bar 4 ; 5% signal intensity was found for the CHO-MIF impurity in 50 ,ug anti MIF antibodies resultant after the fourth step of the downstream process as shown in lane/bar 5; 0% signal intensity was found for the CHO-MIF impurity in 50 μο, anti M ' lF antibodies resultant after the last step of the downstream process as shown in lane/bar 6; 40% signal intensity was calculated for 2 ng recombinant CHO-MIF as shown in lane/bar 8; 29% signal intensity was calculated for 1 ng recombinant CHO-MIF as shown in lane/bar 9; 11% signal intensity was calculated for 0.5 ng

recombinant CHO-MIF as shown in lane/bar 10 ; ,

Figure 5: is a Western Blot for quantification of CHO-MIF

impurities in finalized anti-MIF production lots. The Blot was detected by the specific affinity purified rabbit anti CHO-MIF antibody and a commercially available horse radish peroxidase conjugated donkey anti rabbit IgG. Lane 1: molecular weight protein marker; Lane 2: final purified anti-MIF antibodies after removal of CHO- MIF contaminants (500 μg anti-MIF antibodies/ lane ) ; Lane 3: 500 μ final purified anti-MIF antibodies but spiked with 1 ng CHO-MIF; Lane 4-7 : recombinant CHO-MIF reference, 4 ng lane (corresponding to 8 ppm in 500 μ<^ anti-MIF antibodies) , 2 ng/lane

(corresponding to 4 ppm in 500 p.q anti-MIF antibodies) , 1 ng/lane (corresponding to 2 ppm in 500 9 anti-MIF antibodies) and 0.5 ng/lane

(corresponding to 1 ppm in 500 g anti-MIF antibodies) . Arrow A denotes the heavy chain of the anti-MIF antibody sample, arrow B the light chain of the anti-MIF antibody sample and arrow C denotes the CHO-MIF bands.

Figure 5a: is is a bar chart of the CHO-MIF protein signals resultant from, a Western Blot as shown in figure 5. To that end, the CHO-MIF signals were scanned by a LAS4GQ0 ( Fuj ifilm Life Science "" ) using the Image Reader LAS4000 scanner software *' and directly quantified by the Image Quant LAS4000 1 " software. The 4 ng CHO-MIF signal from lane 4 was set to 100% and directly compared to the other CHO-MIF signals. No CHO-MIF signals were found for the marker protein added on lane 1 as well as for the final purified anti MIF antibody preparation shown in lane/bar 1 and 2, 46% of the 100% reference signal was found for the purified anti MIF antibody spiked with 1 ng recombinant CHO-M1F as shown in lane/bar 3; 81% of the 100% reference signal was found for the 2 ng CHO-MIF protein shown in lane/bar 5; 42% of the 100% reference signal was found for the 1 ng CHO-MIF protein shown in lane/bar 6; 16% of the 100% reference signal was found for the 0.5 ng CHO- MIF protein shown in lane/bar 7.

Figure 6: is a bar chart of CHO-MIF proteins resultant from

Western Blots detected by different rabbit anti MIF antibodies {data not shown) . The bars demonstrate the highest sensitivity of affinity purified rabbit anti CHO-MIF antibodies to CHO-MIF in contrast to rabbit anti huMIF and rabbit anti moMIF antibodies. To that end, to each Western Blot the same amount of CHO-MIF proteins were blotted (2 ng/lane; 1 ng/lane; 0.5 ng/lane) and the resultant signal intensities of the CHO-MIF electronically compared by a LAS4000 (Fuj ifilm Life Science") . Each Western Blot was detected with the same amount of the respective rabbit antibodies (3.5 μg/mL} in

combination with a horse radish peroxidase conjugated donkey anti-rabbit IgG (1:6000) . The Western Blots detected by rabbit anti huMIF and rabbit anti moMIF were additionally controlled by 10 ng rhuMIF or 10 ng rmoMIF to demonstrate the functionality of both antibody preparations (data no shown) , Afterwards, the resultant CHO-MIF signals we e scanned by a LAS40GQ Image Reader LAS4000 scanner software "' and directly quantified by the Image Quant LAS4000 * software.

For the directly comparison of all CHO-MIF signals, the 2 ng CHO-M.1F signal from the rabbit anti CHO- MIF antibody blot was set to 100% . Black bars: CHO- MIF signals resultant from the rabbit anti CHO-MIF antibodies; dark grey bars : CHO-MIF signals

resultant from the rabbit anti huMIF antibodies; light grey bars : CHO-MIF signals resultant from the rabbit anti moMIF antibodi s .

Figure 7; shows a Western Blot with different amounts of CHO- MIF detected by the affinity purified rabbit anti CHO-MIF antibodies (3.5 jug/mL, HRP conjugate

1:6000, same conditions as described in Example 5} . This Western Blot is an example for the sensi ivity of the rabbit anti CHO-MIF antibodies. The lowest amount of CHO-MIF detected by the rabbit anti CHO- MIF antibodies was 0.25· ng Iane (corresponding to 0.5 ppm in 500 μ< human anti-MIF antibody preparation) .

Lane 1: 2 ng CHO-MIF (corresponding to 4 ppm CHO- MIF impurity in 500 μg human anti-MIF antibodies) ; lane 2: 1 ng CHO-MIF (corresponding to 2 ppm CHO- MIF impurity in 500 xg human anti-MIF antibodies) ; lane 3: 0.5 ng CHO-MIF (corresponding to 1 ppm CHO- MIF impurity in 500 μ human anti-MIF antibodies) ; lane 4: 0,25 ng CHO-MIF (correspending to 0.5 ppm CHO-MIF impurity in 500 μ human anti-MIF antibodies) ; lane M: molecular weight protein marker ,

Figure 7a: is a bar chart of the CHO-MIF protein signals

resultant from a Western Blot as shown in figure 7. To that end, the CHO-MIF signals were scanned by a LAS4000 (Fujifilm Life Science*) using the Image Reader LAS4000 scanner software " and directly quantified by the Image Quant LAS40GQ l> software. The 2 ng CHO-MIF signal from lane 1 was set to 100% and directly compared to the other CHO-MIF signals. 90% of the 100% reference signal was found 1 ng recombinant CHO-MIF as shown in lane/bar 2; 76% of the 100% reference signal was found for the 0.5 ng CHO-MIF protein shown in lane/bar 3; 26% of the 100% reference signal was found for the 0.25 ng

CHO-MIF protein shown in lane/bar 4,

Figure 8; is a cartoon showing the position and recognition site of the Zinc finger nuclease (ZFN) at the boundary of exonl/intronl of the MIF locus (SEQ ID NO: 21) , The lower part illustrates the strategy for the genetic characterization of MIF knockout clones. Exon. 1 is shown in bold letters; Intron 1 in italics. The 5 basepair cleavage site GGCCC is i between the 15 bp recognition sites of the two Zinc finger nuclease subunits. The Naei restriction site GCCGGC is underlined.

Two PCR primers, 9983 (SEQ ID NO: 11) and 9879 (SEQ ID NO: 12) binding in the CHO-MIF locus outsite the translated region were designed. Using these 2 primers, a 1260 bp fragment can be amplified by PCR containing 2 Naei sites in the case of the wildtype fragment. Due to the ZFN treatment, the first Naei site is expected to be destroyed. In case of a wildtype gene locus the 1260 bp fragment a Naei digest results in 3 fragments, in case of a knockout only 2 fragments are generated. The expected pattern after separation on a DNA- garose gel is shown on the cartoon of a gel (lane 1 knockout, lane 2 wildtype) As CHO cells are expected to be diploid, a heterozygous constellation as shown in lane 3 is expected.

Figure 9 ; shows an agarose gel of the genetic analysis of

individual CHO cell clones producing antibody RAB0 isolated after treatment with the Zinc finger nuclease . A PCR fragment spanning the gene locus of MIF is digested with Naei and separated on an agarose gel as described in figure 8. Five cell lines are homozygous M1F knockout (lanes 2,3; 4,5; 8,9; 10,11; 12,13), One cell line ( lane 6,7} is heterozygous. The original wildtype cell line is shown in lane 14 , 15.

Figure 9a: shows a schematically drawing of figure 9. The grey highlighted circles demonstrate the main signals of the PGR fragments as shown in figure 9.

Figure 10: is an example of a Western blot for the

characterisation of CHO-MIF knockout cell lines producing antibody RAB0 on the protein level . Cell extracts of individual CHO-MIF heterozygous (lane 1) , knockout {lanes 2 =4) and wild-type (lane 5 & 6) , cell clones are separated on a denaturing protein gel and transferred to a membrane. As a control, CHO-MIF purified from E.coli is on the gel (lane 8) . The blot is stained using a MIF specific antibody. There is no MIF detectable in knockout cell clones (lanes 2, 3 , 4) .

Figure 10a: is a schematically drawing of figure 10. The dark circles demonstrate the positive CHO-MIF signals (lane 1, 5, 6, 8) of the Western blot; the highlighted open circles demonstrate the negative signals (lane 2, 3, 4) of CHO-MIF protein as shown in the Western blot in figure 10.

Figure 11; is an example of a western blot analysis of a

human anti -MIF antibody purified from a CHO-MIF knockout cell line.

The MIF- specific antibody RAB0 is produced in a MIF knockout CHO cell line (lane 2) or in a wild type CHO cell line (lane 3) . Even after loading the purified antibody up to 500 ug per lane on a gel there is no CHO-MIF detectable when the antibody is produced in the MIF knockout cell line . Different amounts of purified CHO-MIF produced in E.coli were applied as controls ( lanes 4-6) .

Figure 11a; is a bar chart of the CHO-MIF protein signals

resultant from a Western Blot as shown in figure 11. To that end, the CHO-MIF signals were scanned by a LAS4000 {Fujifilm Life Science 0 ) using the

Image Reader LAS4000 scanner software* and directly quantified by the Image Quant LAS4000 J software. The 4 ng CHO-MIF signal from lane 4 was set to 100% and directly compared to the other CHO-MIF signals. 0% signal intensities for CHO-MIF was found in the purified anti MIF antibodies produced in knockout CHO cell line as shown in lane/bar 2; a huge signal {>>300 %) of CHO-MIF was found in purified antibodies produced from wild- type CHO cell line shown in lane /bar 3; 57% of the 100% reference signal was found for- the 2 ng CHO-MIF protein shown in lane/bar 5 and 28% signal intensities was found for 1 ng CHO-MIF as shown in lane/bar 5.

The present inventors have succeeded i identifying and characterizing the CHO-MIF gene. On that basis they further succeeded in the provision of tools and methods allowing production and testing of anti-MIF antibody preparations in CHO cells, which preparations are essentially free of

contaminating CHO-MIF. . These tools and methods further allowed production and testing of all preparations as

produced in CHO cells, which comprise components which can be bound by CHO-MIF (i.e. CHO MIF binding proteins), whereupon these preparations are essentially free of contaminating CHO- MIF.

Thus, the present invention is directed to the analysis of the gene locus coding for CHO-MIF. This allows the generation of MIF knockout CHO cells producing recombinant antibodies, or other products, which can be bound by CHO-MIF, e.g.

(CHO)MIF ligands, (CHO) MIF agonists or antagonists, (CHO)MIF inhibitors, like peptides binding (CHO) MIF, (CHO) MIF

receptor fragmen s , preferably antibodies which are directed towards human MIF, essentially free of any CHO-MIF

contaminants .

"Essentially without any CHO-MIF contamination" or

^essentially free of CHO-KIF contamination", which are used interchangeably, in the context of this application shall mean that the amount of CHO-MIF is below 0.5 ppm. Preferably, the amount of CHO MIF is below 0.2 ppm.

The present invention is further directed to a knock-out cell line wherein the CHO-MIF gene is successfully knocked out, wherein this k.o. cell line shows essentially the same characteristics as the parenteral CHO wild type cell line.

The present invention is also directed to a highly sensitive method for the detection of ppm levels of CHO-MIF which during production in CHO cells of products which bind to CHO MIF, e.g. antibodies, preferably anti-MIF antibodies or antigen-binding fragments thereof can remain attached to the desired product in some cases. In a preferred embodiment, this detection method is based on the generation and

purification of highly specific anti CHO-MIF antibodies which are affinity-purified polyclonal antibodies obtained from rabbits immunized with the CHO-MIF of the present invention. These antibodies are in a preferred embodiment generated by immunization with CHO-MIF produced by recombinant E, coli techniques .

The present invention is thus further directed to the

isolation of mRNA coding fo CHO-MIF as produced by CHO cells . The cDNA created by reverse transcription of this mRNA is cloned into a prokaryotic expression vector. The CHO-M ' IF protein expressed thereof in E, coli is purified to homogeneity. The recombinant CHO-MIF is used to immunize rabbits in order to generate the inventive polyclonal rabbit antibodies specific to CHO-MIF.

Very surprisingly, the affinity-purified, polyclonal rabbit antibodies (see Example 4) as provided by the present

inventors are capable of detecting CHO-MIF contaminations bound to the desired anti-MIF antibodies very sensitively; this enables the detection of these CHO-MIF contaminations down to the ppm range .

In a preferred embodiment, the detection step is carried out by a Western Blot analysis. Other analytical detection methods are, however , well known to a person skilled in the art , and include (though by no means limited to) e.g. enzyme- linked immunoassays, radioimmunoassays, fluorescent

immunoassays, bioluminescent and chemiluminescent

immunoassays, competitive immunoassays, dot blot technology, and immune precipitation HPLC, mass spectrometry or LC/MS/MS .

Based on the knowledge of the CHO-MIF sequence, the present invention is further directed to the analysis of the gene locus coding for CHO-MIF , This allows the generation of MIF knockout CHO cells or the detection of CHO-MIF raRNA to verify the presence or absence of MIF in CHO cells . The inventors successfully provided a CHO MIF knock out cell line, wherein it is surprising that this cell line was stable and useful for the expression of recombinant proteins, particularly those which can bind CHO-MIF, preferably, antibodies, more preferred anti-MIF antibodies, in particular as MIF per se is involved in quite a few important cellular processes and its absence in a knock out cell should have disturbed the

cellular processes to an extent where stable cell survival was not possible anymore.

Quite unexpectedly, the productivity for anti-MIF antibodies was however comparable to that as observed in the same cells without the knock out of CHO MIF (wild-type cells) (data not shown) .

The antibodies as produced in the present inventive CHO MIF knock out cell lines are also comparable in their physico- chemical characteristics to those as produced in v/ild type cell lines (data not shown) .

This invention is characterized particularly by the following features :

1. A method for the detection of CHO-MIF contaminations in a monoclonal anti-MlF antibody preparation, comprising the step of contacting the anti -MIF antibody preparation with a polyclonal anti-CHO-MIF antibody, affinity purified against CHO-MIF,

2. The method according to item 1 wherein the CHO-MIF

contaminates a final CHO cell produced monoclonal anti- MIF antibody - preparation or a preparation of antigen- binding fragments thereof .

3. The method according to item 1 and/or 2 wherein the CHO- MIF is endogenous CHO-MIF produced by CHO cells . . The method according to any one or more of items 1 to 3 wherein the detection step is carried out by a semiquantitative Western Blot analysis

5. Use of a rabbit anti-CHO-MIF polyclonal antibody,

affinity purified against CHO-MIF, for the detection of CHO-MIF contaminations during production of monoclonal anti -MIF antibodies or antigen-binding fragments thereof or in the final preparation of monoclonal anti-MIF antibody or antigen-binding fragments thereof. The use according to item 5 wherein the detection step is carried out as a. semi-quantita ive Western Blot analysis . A method for the production of anti- IF antibodies or antigen-binding fragments thereof in CHO cells, wherein said antibodies or antigen-binding fragments thereof are essentially free of CHO-MIF, wherein a detection method as defined in any one of claims 1 to 6 is carried out. A CHO-MIF knock-out CHO cell line. The CHO-MIF knock-out CHO cell line, wherein the cell line comprises any one or more of the following {in E.coli deposited) plasmids: DSM 25110, DSM 25112, DSM 25111, DSM 25113, DSM 25114, DSM 25115, DSM 25853, DSM 25860, DSM 25861, DSM 25862, DSM 25863 and DSM 25864. The CHO-MIF knock-out CHO cell line according to item 8 or 9, wherein the cell line comprises the plasmids DSM

25110 and DSM 25112 or DSM 25861 and DSM 25862. The CHO-MIF knock-out CHO cell line according to item 8 or 9, wherein the cell line comprises the plasmids DSM

25111 and DSM 25113 or DSM 25859 and DSM 25860. The CHO-MIF knock-out CHO cell line according to item 8 or 9, wherein the cell line comprises the plasmids DSM 25114 and DSM 25115 or DSM 25863 and DSM 25864. The use of the CHO-MIF knock-out CHO cell line of any of items 8 to 12 for the production of a preparation of monoclonal anti-MIF antibodies or binding fragments thereof, preferably for the production of any one of antibodies RAB0, RAB9, RAB4 , RAM0, RA or RA 4. Essentially CHO MIF free anti-MIF antibody preparation as obtainable by the method of item 7, or by use of the CHO -MIF knock-out cell line of any of items 8 to 12. A method for the production of an essentially CHO MIF free anti-MIF antibody preparation which is

characterized by the use of the CHO-MIF knock-out CHO cell line according to any one of items 8 - 12. Preparation of a recombinant MIF binding protein, preferably an (h)MIF binding protein, like an (h) MIF binding peptide, ligand, agonist, antagonist, inhibitor, or a MIF receptor fragment, or an anti-(h)MIF antibody preparation, produced in a CHO cell line, characterised in that said preparation is essentially free of CHO- MIF. Prepara ion of a recombinant MIF binding protein, preferably an (h) MIF binding protein, like an (h) MIF binding peptide, ligand, agonist, antagonist, inhibitor, or a MIF receptor fragment, or an anti-(h)MIF antibody preparation, which is obtainable by the method according to item 15 or by a method of production which comprises, preferably as a quality control step, the method of detection of any one of the above items 1 - 4 or 7. Preparation of a recombinant MIF binding protein, preferably an (h) MIF binding protein, like an (h) IF binding peptide, ligand, agonist, antagonist, inhibitor, or a MIF receptor fragment, or an anti- (h) IF antibody preparation, produced by the method according to item 15 or by a method of production which comprises, preferably as a quality control step, the method of detection of any one of the above items 1 - 4 or 7. The anti-hM!F antibody preparation of any of items 16 - 18 above, which is essentially free of CHO-MIF, wherein the anti-hMIF antibody is selected from the group of RAB , A30 , RAB9 , RAM , RAMO and/or RAM9.

The plasmids as deposited for the above anti-hMIF antibodies are characterized by their DSM number which is the official number as obtained upon deposit under the Budapest Treaty with the German Collection of Microorganisms and Cell

Cultures (DSMZ) , Mascheroder Weg lb, Braunschweig, Germany.

The plasmid with the DSM 25110 number comprises the light chain sequence of the anti-MIF antibody A34.

The plasmid with the DSM 25112 number comprises the heavy chain ( IgG4 ) sequence of the an i-MIF antibody RA3 .

The co-expression of plasmids DSM 25110 and DSM 25112 in a suitable, host cell, namely a CHO cell, results in the production of the preferred anti-MIF antibody RAB .

The plasmid with the DSM 25111 number comprises the light chain sequence of the anti-MIF antibody RAB9.

The plasmid with the DSM 25113 number comprises the heavy chain (IgG4) sequence of the anti-MIF antibody RAB9.

The co-expression of plasmids DSM 25111 and DSM 25113 in a suitable host cell, namely a CHO cell, results in the production of the preferred anti-MIF antibody RAB9.

The plasmid. with the DSM 25114 number comprises the light chain sequence of the anti-MIF antibody RAB0.

The plasmid with the DSM 25115 number comprises the heavy- chain (IgG4) sequence of the anti-MIF antibody RAB0. The co-expression of plasmids DSM 25114 and DSM 25115 in a suitable host cell, namely a CHO cell , results in the

production of the preferred anti-MIF antibody RAB0.

Also deposited are antibodies RAMO, RAM9 and RAM4 ; all have been deposited with the DSZM, Braunschweig, Germany on April 12, 2012 according to the Budapest Treaty, with the following designations:

RAM9 - - heavy chain : E. coli GA 662- 01 , pRAM9hc .... DSM 25860.

RAM4 - - light chain: E. coii GA 906- 04 . RAM lc - - DSM 25861.

RAM9 - - light chain : E. coli GA 661- 01 . pRAM91c - - DSM 25859.

RAM4 - - heavy chain : E. coli GA 657- 02 . RAM4he - - DSM 25862.

RAMO - - light chain : E. coli GA 906- 01 pRAMOlc - - DSM 25863.

RAMO - - heavy chain: E. coli GA 784- 01 , pRAMOhc - - DSM 25864.

The production of anti- (ox) MIF antibodies may also include any method known in the art for the cultivation of said

transformed cells, e.g. in a continuous or batchwise manner, and the expression of the anti- (ox) MIF antibody, e.g.

constitutive or upon induction. It is referred in particular to WO 2009/086920 for further reference for the production of anti- (ox) MIF antibodies. In a preferred embodiment, the anti- (ox) MIF antibodies as produced according to the present invention bind to oxM!F or an epitope thereof. Particularly preferred antibodies in accordance with the present invention are antibodies RAB9, RAB4 and/or RABO as well as RAM9 , RAM4 and/or RAMO .

The sequences of these antibodies are partly also disclosed in WO 2009/086920; see in addition the sequence list of the present application and the following:

SEQ ID NO: 22 for the amino acid sequence of the light ch in of RAE : DIQMTQSPSS LSASVGDRVT ITCRSSQRIM TYLNWYQQKP

GKAPKLLIFV ASHSQSGVPS RFRGSGSETD FTLTISGLQP EDSATYYCQQ SFWTPLTFGG GTKVEI RTV AAPSVFIFPP SDEQLKSGTA SWCLLNNFY PREAKVQWKV DNALQSGNSQ ESVTEQDSKD STYSLSSTLT LSKADYEKHK

VYACEVTHQG LSSPVTKSPN RGEC,

SEQ ID NO: 23 for the amino acid sequence of the light chain of RAB4 : DIQMTQSPGT LSLSPGERAT LSCRASQGVS SSSLAWYQQK

PGQAPRLLIY GTSSRATGIP DRFSGSASGT DFTLTISRLQ PEDFAVYYCQ QYGRSLTFGG GTKVEIKRTV AAPSVFIFPP SDEQLKSGTA SWCLLHNFY PREAKVQWKV DNALQSGNSQ ESVTEQDSKD STYSLSSTLT LSKADYEKHK VYACEVTHQG LSSPVTKSFN RGEC ,

SEQ ID NO; 24 for the amino acid sequence of the light chain of RA30 : DIQMTQSPGT LSLSPGERAT LSCRASQGVS SSSLAWYQQK

PGQAPRLLIY GTSSRATGIP DRFSGSASGT DFTLTISRLQ PEDFAVYYCQ QYGRSLTFGG GTKVEIKRTV AAPSVFIFPP SDEQLKSGTA SWCLLNNFY PREAKVQWKV DNALQSGNSQ ESVTEQDSKD STYSLSSTLT LSKADYEKHK VYACEVTHQG LSSPVTKSFN RGEC ,

SEQ ID NO; 25 for the amino acid sequence of the light chain of RAB2 :

DIQMTQSPVT LSLSPGERAT LSCRASQSVR SSYLAWYQQK PGQTPRLLIY

GASNRATGIP DRFSGSGSGT DFTLTISRLE PEDFAVYYCQ QYGNSLTFGG GTKVEIK RTV AAPSVFIFPP SDEQLKSGTA SWCLLNNFY PREAKVQWKV

DNALQSGNSQ ESVTEQDSKD STYSLSSTLT LSKADYEKHK VYACEVTHQG

LSSPVTKSFN RGEC

SEQ ID NO; 25 for the amino acid sequence of the heavy chain of RAB9 : EVQLLESGGG LVQPGGSLRL SCAASGFTFS IY5MNWVRQA

PGKGLEWVSS

IGSSGGTTYY ADSVKGRFTI SRDNSKNTLY LQM SLRAED TAVYYCAGSQ

WLYGMDVWGQ GTTVTVSSAS TKGPSVFPLA PCSRSTSEST AALGCLVKDY

FPEPVTVS N SGALTSGVHT FPAVLQSSGL YSLSSWTVP SSSLGTKTYT

CNVDHKPSNT KVDKRVESKY GPPCPPCPAP EFLGGPSVFL FPPKPKDTLM

ISRTPEVTCV WDVSQEDPE VQFNWYVDGV EVHNAKTKPR EEQFNSTYRV

VSVLTVLHQD WLNGKEYKCK VSNKGLPSSI EKTISKAKGQ PREPQVYTLP

PSQ'EEMTKNQ VSLTCLVKGF YPSDIAVEWE SNGQPENNYK TTPPVLDSDG

SFFLYSRLTV DKSR QEGNV FSCSVMHEAL HNHYTQKSLS LSLGK

SEQ ID NO; 27 foi" the amino acid sequence of the heavy chain Of RAB ; EVQLLESGGG LVQPGGSLRL SCAASGFTFS IYAMDWVRQA

PGKGLEWVSG

IVPSGGFTKY ADSVKGRFTI SRDNSKNTLY LQMNSLRAED TAVYYCA N VIAVAGTGYY YYGMDVWGQG TTVTVSSAST KGPSVFPLAP CSRSTSESTA

ALGCLVKDYF PEPVTVSWNS GALTSGVHTF PAVLQSSGLY SLSSWTVPS SSLGTKTYTC NVDHKPSNTK VDKRVESKYG PPCPPCPAPE FLGGPSVPLF PPKPKDTLMI SRTPEVTCW VDVSQEDPEV QFN YVDGVE VHNAKTKPRE EQFNSTYRW SVLTVLHQDW LNGKEYKCKV SNKGLPSSIE KTISKAKGQP REPQVYTLPP SQEEMTKNQV SLTCLVKGFY PSDIAVEWES NGQPENNYKT TPPVLDSDGS FFLYSRLTVD KSRWQEGNVF SCSVMHEALH NHYTQKSLSL SLGK

SEQ ID NO: 28 for he amino acid sequence of the heavy of RABO : EVQLLESGGG LVQPGGSLRL SCAASGPTFS WYA DWVRQA

PGKGLEWVSG

IYPSGGRTKY ADSVKGRFTI SRDNSKNTLY LQMNSLRAED TAVYYCARVN VIAVAGTGYY YYGMDVWGQG TTVTVSSAST KGPSVFPLAP CSRSTSESTA ALGCLVKDYF PEPVTVSWNS GALTSGVHTF PAVLQSSGLY SLSSWTVPS SSLGTKTYTC NVDHKPSNTK VDKRVESKYG PPCPPCPAPE FLGGPSVFLF PPKPKDTLMI SRTPEVTCW VDVSQEDPEV QFNWYVDGVE VHNAKTKPRE EQFNSTYRW SVLTVLHQDW LNGKEYKCKV SNKGLPSSIE KTISKAKGQP REPQVYTLPP SQEEMTKNQV SLTCLVKGFY PSDIAVEWES NGQPENNYKT TPPVLDSDGS FFLYSRLTVD KSRWQEGNVF SCSVMHEALH NHYTQKSLSL SLGK

SEQ ID NO; 29 for the amino acid sequence of the heavy chain of RAB2 : EVQLLESGGG LVQPGGSLRL SCAASGFTFS IYAMDWVRQA

PGKGLEWVSG IVPSGGFTKY ADSVKGRFTI SRDNSKNTLY LQMNSLRAED TAVYYCARVN VIAVAGTGYY YYGMDVWGQG TTVTVSSAST KGPSVFPLAP CSRSTSESTA

ALGCLVKDYF PEPVTVSWNS GALTSGVHTF PAVLQSSGLY SLSSWTVPS SSLGTKTYTC NVDHKPSNTK VDKRVESKYG PPCPPCPAPE FLGGPSVFLF PPKPKDTLMI SRTPEVTCW VDVSQEDPEV QFNWYVDGVE VHNAKTKPRE EQFNSTYRW SVLTVLHQDW LNGKEYKCKV SNKGLPSSIE KTISKAKGQP REPQVYTLPP SQEEMTKNQV SLTCLVKGFY PSDIAVEWES NGQPENNYKT TPPVLDSDGS FFLYSRLTVD KSRWQEGNVF SCSVMHEALH NHYTQKSLSL SLGK.

SEQ ID NO: 30 for the amino acid sequence of RAMOhc :

EVQLLESGGG LVQPGGSLRL SCAASGFTFS WYAMDWVRQA PGKGLEWVSG IYPSGGRTKY ADSVKGRFTI SRDNSKNTLY LQMNSLRAED TAVYYCARVN VIAVAGTGYY YYGMDVWGQG TTVTVSS ST KGPSVFPLAP SSKSTSGGT ALGCLVKDYP PEPVTVSW S GALTSGVHTF PAVLQSSGLY SLSSWTVPS SSLGTQTYIC NVNHKPSNTK VDKRVEPKSC DKTHTCPPCP APELLGGPSV FLFPPKPKDT LMISRTPEVT CVWDVSHED PEVKF WYVD GVEVHNAKTK PREEQY STY RWSVLTVLH QDWLNGKEYK CKVS KALPA PIEKTISKAK GQPREPQVYT LPPSREEMTK NQVSLTCLVK GFYPSDIAVE ESNGQPE YKTTPPVLDS DGSFFLYSKL TVDKSRWQQG NVFSCSVMHE ALK KYTQKS LSLSPG .

SEQ ID NO; 31 for the amino acid sequence of RA Olc : DIQMTQSPGT LSLSPGERAT LSCRASQGVS SSSLAWYQQK PGQAPRLLIY GTSSRATGIP DRPSGSASGT DFTLTISRLQ PEDFAVYYCQ QYGRSLTFGG GTKVEI RTV AAPSVFIFPP SDEQLKSGTA SWCLLNNFY PREAKVQWKV DNALQSG SQ ESVTEQDSKD STYSLSSTLT LS ADYEKHK VYACEVTHQG LSSPVTKSFN RGEC .

SEQ ID NO: 32 for the amino acid sequence of RAM9he : EVQLLESGGG LVQPGGSLRL SCAASGFTFS YSMNWVRQA PGKGLE VSS IGSSGGTTYY ADSVKGRFTI SRDNSKNTLY LQMNSLRAED TAVYYCAGSQ WLYGMDV GQ GTTVTVSSAS TKGPSVFPLA PSSKSTSGGT AALGCLVKDY FPEPVTVSWN SGALTSGVHT FPAVLQSSGL YSLSSWTVP SSSLGTQTYI CNVNHKPSNT KVDKRVEPKS CDKTHTCPPC PAPELLGGPS VFLFPPKPKD TLMISRTPEV TCWVDVSHE DPEVKFNWYV DGVEVHNAKT KPREEQYNST YRWSVLTVL HQDWLNGKEY KCKVSNKALP APIEKTISKA KGQPREPQVY TLPPSREEMT KNQVSLTCLV KGFYPSDIAV EWESNGQPEN NY TTPPVLD SDGSFFLYSK LTVDKSRWQQ G VFSCSVMH BALHNHYTQK SLSLSPGK. SEQ ID NO: 33 for the amino acid sequence of RAM9lc: DIQMTQSPSS LSASVGDRVT I CRSSQRIM TYLNWYOQRP GKAPKLLIFV ASKSQSGVPS RFRGSGSETD FTLTISGLQP EDSATYYCQQ SFWTPLTFGG GTKVEIKRTV AAPSVFIFPP SDEQLKSGTA SWCLLNNFY PREAKVQWKV DNALQSGNSQ ESVTEQDSKD STYSLSSTLT LSKADYEKHK VYACEVTHQG LSSPVTKSFN RGEC .

SEQ ID NO: 34 for the amino acid sequence of RAM4hc : EVQLLESGGG LVQPGGSLRL SCAASGFTFS IYAMDWVRQA PGKGLEWVSG

I PSGGFTKY ADSVKGRFTI SRDNSKNTLY LQMNSLRAED TAVYYCARVN VIAVAGTGYY YYGMDVWGQG TTVTVSSAST KGPSVFPLAP SSKSTSGGTA ALGCLVKDYP PEPVTVSWNS GALTSGVHTF PAVLQSSGLY SLSSWTVPS SSLGTQTYIC NVNHKPSNTK VDKRVEPKSC DKTHTCPPCP APELLGGPSV FLFPPKPKDT LMISRTPEVT CVWDVSHED PEVKFN YVD GVEVHNAKTK PREEQYNSTY RWSVLTVLH QDWLNGKEYK CKVSNKALPA PIEKTISKAK

GQPREPQVYT LPPSREEMTK NQVSLTCLVK GFYPSDIAVE WESNGQPENN

YKTTPPVLDS DGSFFLYS L TVDKS WQQG NVFSCSVMHE ALH HYTQKS LSLSPGK .

SEQ ID NO: 35 for the amino acid sequence of RAM4lc:

DIQMTQSPGT LSLSPGERAT LSCRASQGVS SSSLA YQQK PGQAPRLLIY

GTSSRATGIP DRFSGSASGT DFTLTISRLQ PEDFAVYYCQ QYGRSLTFGG

GTKVEIKRTV AAPSVFIFPP SDEQLKSGTA SWCLLNNFY PREAKVQWKV

DNALQSGNSQ ESVTEQDSKD STYSLSSTLT LSKADYEKHK VYACEVTHQG LSSPVTKSP RGEC .

The anti -MIF antibody of the invention is preferably an isolated monoclonal antibody. The anti -MIF antibody can be an IgG, an IgM , an IgE , an IgA, or an IgD molecule. In other embodiments, the anti-MIF antibody is an IgGl , IgG2 , IgG3 or IgG4 subclass. In other embodiments, the antibody is either subclass IgGl or IgG4. In other embodiments, the antibody is subclass IgG4. In some embodiments, the IgG4 antibody has a single mutation changing the serine (serine228, according to the Rabat numbering scheme) to proline. Accordingly, the CPSC sub-sequence in the Fc region of IgG4 becomes CPPC, which is a sub-sequence in IgGl (Angal et al . Mol Immunol. 1993 , 30, 105-108} .

Additionally, the production of anti- (ox) MIF antibodies may include any method known in the art for the purification of an antibody, e.g. via anion exchange chromatography o

affinity chromatography. In one embodiment the anti- (ox) MIF antibody can be purified from cell culture supernatants by size exclusion chromatography.

The terms "center region" and "C-terminal region" of MIF refer to the region of human MIF comprising amino acids 35-68 and aa 86-115, respectively, preferably aa 50-68 and aa 86 to 102 of human MIF, respectively. Particularly preferred antibodies of the present invention bind to either region aa 50-68 or region aa 86-102 of human IF. This is also reflected by the binding of the preferred antibodies RAB0, RAB4 RAB2 and RAB9 as well as RAM , RAMS and RAM0 which bind as follows:

RAB4 and RAM4 : aa 86-102

RAB9 and RAM9 : aa 50-68

RAB0 and RAM0 : aa 86-102

RAB2 : aa 86 - 102

The term "epitope" includes any protein determinant capable of specific binding to an immunoglobulin or an antibody fragment. Epitopic determinants usually consist of chemically active surface groupings of molecules such as exposed amino acids, amino sugars, or other carbohydrate side chains and usually have specific three-dimensional structural

characteristics, as well as specific charge characteristics.

The following description of the production of anti-MIF antibodies shall illuminate exemplariiy the process for producing a recombinant anti-MIF antibody preparation which process includes a step for testing whether the purified antibody preparation is free from contaminating MIF, i.e. a detection step for CHO-MIF contaminations.

The production process according to the present invention of the anti-MIF antibodies includes any method for the

generation of recombinant DNA by genetic engineering, e.g. via reverse transcription of RNA and/or amplification of DNA and cloning into expression vectors. In some embodiments, the vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome. In some embodiments, the vector is capable of autonomous replication in a host cell into which it is introduced (e.g. bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). In other embodiments, the vector (e.g. non-episomal mammalian vectors) can be integrated, into the genome of a host cell upon introduction into the host cell, and thereby replicated along with the host genome. Moreover . , certain vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as "recombinant expression vectors " (or simply "expression vectors") ,

Anti-MIF antibodies can be produced by means of conventional expression vectors , such as bacterial vectors (e.g. pBR322 and its derivatives), or eukaryotic vectors. Those sequences that encode the antibody can be provided with regulatory sequences that regulate the replication, expression and/or secretion from the host cell. These regulatory sequences comprise, for instance, promoters (e.g. CMV or SV40) and signal sequences. The expression vectors can also comprise selection and ampfification markers, such as the

dihydrofoiate reductase gene (DHF ' R) , hygromycin-B- phosphotransferase , and thymidine-kinase. The components of the vectors used, such as selection markers, replicons, enhancers, can either be commercially obtained or prepared by means of conventional methods . The vectors are constructed for the expression in cell cultures , namely in CHO cells .

The anti-MIF antibody light chain gene and the anti-MIF antibody heavy chain gene can be inserted into separate vectors or both genes are inserted into the same expression vector. The antibody genes are inserted into the expression vector by standard methods , e.g. ligation of complementary restriction sites on the antibody gene fragment and vector, or blunt end ligation if no restriction sites are present.

The production of anti-MIF antibodies or antigen-binding fragments thereof may include any method known in the art for the introduction of recombinant D A into eukaryotic cells by transfection, e.g. via electroporation or microinjection. For example , the recombinant expi'ession of anti-MIF antibody can be achieved by introducing an expression plasmid containing the anti-MIF antibody encoding DNA sequence under the control of one or more regulating sequences such as a strong

promoter, into a CHO-cell line . , by an appropriate

transfection method resulting in cells having the introduced sequences stably integrated into the genome . The lipofection method is an example of a transfection method which may be used according to the present invention.

The production of an i-MIF antibodies may also include any- method known in the art for the cultivation of said

transformed cells, e.g. in a continuous or batchwise manner, and the expression of the anti-MIF antibody, e.g.

constitutive or upon induction. It is referred in particular to WO 2009/086920 for further reference for the production of anti-MIF antibodies. In a preferred embodiment, the

antibodies of the CHO-MIF free anti-MIF antibody preparation as produced according to the present invention bind to MIF or a MIF fragment. Particularly preferred antibodies to be produced in accordance with the present invention are RAB9 , RAB4 and RAB0 (deposited as E. coli containing plasmids DSM 25114 and DSM 25115 for RABO, DSM 25111 and DSM 25113 for RAB9 and DSM 25110 and DSM 25112 for RAB4 , respectively) .

The host cell type, which is used in the production method for the production of MIF, as described herein, is a CHO cell. In one embodiment , the anti-MIF antibody is expressed in a DHFR-deficient CHO cell line, e.g. DXB11, and with the addition of G418 as a selection marker. When recombinant expression vectors encoding antibody genes are introduced into CHO host ceils , the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or secretion of the antibody into the culture medium in which the host cells are grown . Anti-MIF antibodies can be recovered from the culture medium using standard protein purification methods .

Additionally . , the production of anti-MlF antibodies may include any method known in the art for the purification of an antibody, e.g. via anion exchange chromatography or affinity chromatography. In one embodiment the anti-MIF antibody can be purified from cell culture supernatants by size exclusion chromatography.

The present invention now provides an advantageous method which clearly allows to improve and optimize the prior art methods for the production of anti-MIF antibodies or antigen- binding fragments thereof .

In particular, the present inventors were the first to show that antibody preparations prepared with CHO cells could comprise CHO-MIF contaminations which would render the final preparation useless for pharmaceutical or research purposes .

The present inventors were then the first to identify and characterize the CHO-MIF gene . Based on this knowledge, the inventors here additionally provide a specific detection method which allows the detection of a CHO-MIF contamination bound to anti-MIF antibodies, down to the ppm range.

Very surprisingly, the present invention thus provides for the possibility of verifying that the production process for anti-MIF antibodies, in particular the purification process, is suitable for generating a preparation essentially free of CHO-MIF. This is the prerequisiste to establish a production method for the preparation of anti-MIF antibodies free of CHO-MIF. In particular, this improvement allows the

optimization and combination of methods known in the art for purification of the antibody preparations in a manner which depletes the CHO-MIF contaminants, thus allowing the

provision of a highly pure final Ab- reparation, which is free of CHO-MIF contaminations. In addition, the inventive method is a highly sensitive detection method for said contaminations is a safeguard in the industrial production process, ensuring that a highly pure final Ab-preparation, which is free of CHO-MIF contaminations, is produced.

Preferably, this detection is carried with a detection step that uses a polyclonal rabbit anti-MIF antibody that has been obtained by affinity purification against CHO-MIF, Affinity purification is carried out as well known to a person skilled in the art and described e.g. in Lottspeich F. and Zorbas H . (1998) Bioanalytik, Spektrum Akademischer Verlag Heidelberg- Berlin, ISBN 3-8274-0041-4.

The CHO-MIF contaminations can be detected down to ppm-level, in particular it is possible to detect CHO-MIF contaminations down to 0.5 ppm (corresponding to 0.25 ng CHO-MIF in 500 tg antibody preparation) using a highly sensitive rabbit anti CHO-MIF antibody in a common Western Blot technology

quantified by a chemiluminescence signal in a digital

quantitative imaging system (e.g. ImageQ ant LAS 4000 from GE Healthcare) .

" CHO-MIF contamination" in this context means CHO-MIF bound to a recombinantly produced product, e.g. to anti-MIF

antibodies in a preparation of a recombinantly produced product, e.g. anti-MIF antibodies.

The high sensitivity of the present detection method is possible particularly with the polyclonal rabbit anti-MIF antibodies obtained by affinity purification.

In a further alternative embodiment of producing MIF- ee anti-MIF antibodies in CHO cells, the present invention is directed to knock-out CHO cell lines which do not produce CHO-MIF. With these knock-out cell lines it is possible to carry out a production method which provides an extremely pure an i-MlF antibody preparation which is essentially free of CHO-MIF contaminations .

In a preferred embodiment, the above described detection method can be used fox- quality control of a protein produced recombinantly in a CHO MIF cell line, preferably an ant -MIF antibody production, in particular to ensure that the final preparation is essentially free of CHO MIF . The detection method can also be used for a quality control of CHO MIF knock out cell lines .

Knock-out cells according to the present invention can be produced, according to methods known in the art, whereby one possibility is described below in the examples in detail though the invention should not be construed to be limited to this embodiment .

EXAMPLES Example 1

Determination of the DNA sequence encoding CHO-MIF

Oligonucleotides annealing o the 5 1 end and the 3' end of the coding region of CHO-MIF were designed by comparing DNA sequences of related species. Highly conserved areas were selected to design oligonucleotides 8951 and 8954 containing wobble bases, to ensure binding to their corresponding region in the CHO-MIF DNA. Using these oligonucleotides together with a polyT oligo it was possible to amplify a cDNA copy from mRNA isolated from CHO cells with standard cDNA cloning procedures. The resulting PGR product was subjected to DNA sequencing .

After knowing the DNA sequence of the CHO-MIF cDNA it was possible to design specific primers for the amplification of fragments from the genomic DNA purified from CHO cells using standard procedures . Three genomic fI'agments in the area of the cDNA were amplified by PGR and the following PGR products were achieved:

- P27463 with primers 9063 and 9196 , sequenced with oligos 9063 , 9196 ;

- P27465 using primers 9199 and 9064 , sequenced with 9064;

- P28254 with primers 9216 and 9244 , sequenced with 9216 , 9242.

To verify the sequence around the ATG start codon and the 5' upstream region, the genomic DBA of CHO cells was digested with BstHI and circularized . BstHI was known from the cDNA sequence to cut 140 bp downstream of the ATG in the cDN . The circularized DNA was amplified by inverse PGR using two

specific oligonucleotides 9216 (reverse primer) and 9242 (direct primer) binding in the already known part of the cDNA. Using this PGR product (P27883) the sequence of the genomic DNA several hundred base pairs upstream the ATG could be determined.

The DNA and corresponding protein sequence of CHO-MIF are shown in Figure 1 and SEQ ID No : 1 and 2.

Sequences of Oligonucleotides (primers) used to amplify and sequence the CHO-MIF cDNA: pPCR.MIPspec (1) -8951 : ATGTTCRTSGTRAACACCAAYG ( SEQ ID NO: 3) pPCR.MlFspec (4) -8954 : GCGAAGGTGGARYYGTTCCAGiSEQ ID NO; 4) pPCR.choMIF (I) - 9063 TGACTTTTAGCGGCTCTAGCGAC ( SEQ ID NO: 5) pPCR.choMIF (2) - 9064 GATGTGCAGGCGATCAGCCA(SEQ ID NO: 6) pPCR.choMIF - 9196 ATTTCTCCCGATCGGAAGGTGG (SEQ ID NO: 7} pPCR.choMIF - 9216 GGTGAGCTCGGAGAGAAGC ( SEQ ID NO: 8) pPCR.choMIF - 9242 CGGCCCAGTACATCGCAGT (SEQ ID NO : 9 ) pPCR.choMIF - 9244 GCTGCACGCAGCGTTCTGTT {SEQ ID NO: 10) pPCR.choMIFg - 9983 : CGTTAATCTGCAGCGTCTACCTGA (SEQ ID NO:

11)

pPCR.choMIFg - 9879 : GTAAGGCCACTATAGGAAAGCCTG ( SEQ ID NO:

12)

pPCR.choMIF - 9199: GCTTCTCTCCGAGCTCACC (SEQ ID NO: 13)

Example 2

Identification and characce iiati on of the CHO-MIF gene locus

The experimental strategy leading to the DNA sequence of CHO- MIF cDNA and genomic DNA is described in example 1. The analysis of cDNA and genomic DNA was carried out

simultaneously .

The overall organization of the gene locus was determined by aligning genomic and cDNA sequences. The coding region of

CHO-MIF is fragmented on three exons interrupted by two short introns. The sequence of the CHO-MIF gene locus is shown in Figure 2. 33

Example 3

Production and purification of recombinant CHO-MIF

The cDNA of CHO-MIF was cloned into the E.coli expression vector pET19b (Novagen) under the control of the T7 promoter. The plasmid is shown in Figure 3 ,

The plasmid was transformed in E. coli strain. BL21 - CodonPius

(DE3 } -RP (Stratagene) .This strain contains a stably inserted copy of the RNA-polymerase of the bacteriophage T7 under the control of the IPTG inducible lac promoter. The CHO-MIF protein was expressed to high levels after induction with IPTG and highly purified using a 2 step purification

protocol: First the sample was applied to an anion exchange DHAE - Sepharose column (buffer A: 20 mM Tris/HCl, pH 7.8;

buffer B; 20 mM Tris/HCl, pH 7.8 including 1 M NaCl ;

recombinant CHO-MIF eluates by a linear gradient at 10% buffer B) . In the second step the protein was loaded onto a Source S column (buffer A: 20 mM BisTris/HCl, pH 5.5; buffer B: 20 mM BisTris/HCl, pH 5.5 including 1 M NaCl; recombinant CHO-MIF eluates by a linear gradient between 7-10% buffer B) . Finally the protein was concentrated and re-buffered in PBS using common desalting columns {e.g. PD-10 columns). The purity of CHO-MIF was confirmed after gel electrophoresis procedure by Coomassie staining.

Example 4

"Detect: or. of ppm_ leve1s of CHO-MIF contamina t i ns in an anti- MiF antibody preparation.

A highly sensitive semi -quantitative Western Blot analysis was established to monitor CHO-MIF contaminations in recombinant anti-MIF antibody preparations that allow the detection of ppm levels of CHO-MIF.

Purified recombinant CHO-MIF expressed in E. coli was used to immunize rabbits in order to generate specific antibodies against CHO-MIF. Specific rabbit anti CHO-MIF antibodies were purified by a two-step purification { see a) and b) below) , The resultant specific rabbit anti CHO-MIF antibodies enabled a highly sensitive semi -quantitative Western Blot method that allowed the detection of the CHO-MIF contaminant in a lower picogram range. This allows a CHO-MIF impurity monitoring during the downstream process of human anti-MIF antibodies produced in CHO-cells {Figure 4, 4a, 5 and 5a) ,

The detection limit of CHO-MIF impurities in human anti-MIF antibodies was determined with 0.25 ng/lane which is

corresponding to 0.5 ppm in 500 ,u.g human anti-MIF antibody preparation {shown in Fig. 7 and 7a) . a) Immunization of rabbits by recombinant CHO-MIF,

To generate CHO-MIF specific antibodies, 10 rabbits we e immunized according to the following protocol . For the initial immunization; 25 μ-g of recombinant CHO-MIF (in 100 μΐ PBS) were mixed with 100 μΐ CF¾ (Complete Freund' s Adjuvant). The animals received subcutaneously 200 μΐ (4 x 50 μΐ) of the mixture. Two boost immunizations were performed in 2 -3 weeks intervals with the same dose per animal as described above using IFA (Incomplete Freund ' s Adjuvant). Sera were tested by ELISA. Two weeks after the second boost, the rabbits were exsanguinated after narcotization by Pentobarbital. Sera were pooled for the isolation of the anti CHO-MIF antibodies. b) Purification of tocal IgG from CHO-MIF immunized

rabbits . The purification was achieved by affinity chromatography using protein A MabSelect Sure affinity material from GE

Healthcare. Typically, serum from CHO-MIP immunized rabbits was diluted 1:2 in buffer A (=20 mM Na 2 HP0 4; pH 7.0) and applied to a 100 ml MabSelect Sure column. Unbound or

unspecific serum material was washed out by a 10 column volume/ s (CV) washing procedure with buffer A and the elution of total rabbit IgG was done by a pH shift using a 100% gradient step to buffer B (100 mM glycine, pH 2.8) . The elution fractions were pooled and re-buffered in 20 mM

a2HPC'4 H 7.0 for the next affinity purification step. c) Purification of GHO-MIF specific antibodies .

Affinity purified rabbit anti CHO-MIF antibodies were finally purified by a self prepared 5 ml NHS-column (GE Healthcare) coupled with recombinant CHO-MIF. Typically, 100 ml fractions of the re-buffered total rabbit IgG was applied to a 5 ml CHO-MIF affinity column. After a washing step (20 CV wi h buffer A) the elution of the specific anti CHO-MIF antibodies was achieved by a pH shift using a 100% gradient step to buffer B (100 mM glycine, pH 2.8). Eluted material was pooled, re-buffered in PBS , concentrated if necessary and stored at -80°. Functionality of the purified rabbit anti- CHO-MIF antibodies was proved by Western Blot and CHO-MIF ELISA. d} Detection of CHO-MIF in a monoclonal an i-MIF antibody preparat: o .

Test principle

Antibody samples of interest were separated by SDS-PAGE electrophoresis (sodium dodecyl sulphate -polyacrylamide- gel electrophoresis) and transferred to a commonly used membrane e.g. polyvinylidene fluoride (PVDF) or

nitrocellulose. The target protein CHO-MIF was identified and quantified by the specific polyclonal rabbit anti CHO-MIF antibody and chemi luminescence

reaction using a corresponding secondary antibody conjugate ,

Preparation of samples and controls to monitor downstream

process :

To monitor the removal of contaminating CHO-MIF during the purification of anti-MIF antibodies, samples were diluted with SDS buffer to a defined concentration. All samples had the same concentration before they were loaded on the gel {recommended; 30 - 80 μ anti-MIF antibody/ lane) (Figure 4 and 4a) .

The controls {recombinant CHO-MIF) were also loaded on the SDS gel at a final concentration of 0.5, 1, 2 and 4 ng/ lane .

Preparation of samples and controls to analyze the final

purified antibodies:

For analysis of potential CHO-MIF contamination in final purified anti-MIF antibody preparations, 500 g/lane were loaded on the SDS Page. The controls {recombinant CHO-MIF) were added to the gel at a final amount of 0.5, 1, 2 and 4 ng/lane. {Figure 5 and 5a)

Test details

Samples were diluted 1:1 in SDS buffer (100 mM Tris, 4% SDS , 0.2% bromophenol blue, 20% glycerin, 200 mM DTI, pH 6.8) and incubated for 5 minutes at 99°C (protein reduction and denaturation step) . Afterwards, a defined concentration of each sample was loaded on a 4-12%

Bis/Tris Gel (Invitrogen) and separated by gel

electrophoresis with subsequent electrotransfer to a suitable membrane (e.g. PVDF) . For the reduction of unspecific binding effects the membrane was blocked by 2% dry milk diluted in TEST buffer (25 mM Tris, 150 mM aCi , 0.1% polysorbate 20, pH 7.5 ) for 2 hours at RT. Removal of unbound proteins was achieved by washing steps again with TBST . The detection of CHO-MIF was done by affinity purified rabbit anti CHO-MIF antibodies diluted in 0.05% dry milk dissolved in TBST. A secondary antibody conjugated with horseradish peroxides (e.g. donkey anti rabbit/HRP) was incubated with the membrane for 1 hour at RT and washed again with TBST. The

specific CHO-MIF signal was detected and quantified by addition of a chemiluminescence substrate (e.g. Super Signal West Femto, Pierce) using a Luminescent Image Analyzer from Fujifilm (LAS-4000) .

Example 5

Highly sensitive detection of CHO-MIF by affinity purified rabbit anti CHO-MIF antibodies

The sensitivity of the affinity purified rabbit anti CHO-MIF antibodies to CHO-MIF was compared to two other affinity purified polyclonal antibodies directed against human MIF and mouse MIF. These polyclonal antibodies were also produced by the same procedure as described for the rabbit anti CHO-MIF antibodies, with the following exception: rabbit anti huMlF was affinity purified against rhuMIF and rabbit anti moMIF was affinity purified against rmoMIF (same conditions as described for rabbit anti CHO-MIF antibodies) .

Different amounts of CHO-MIF (2, 1 and 0.5 ng/lane) were applied to an SDS gel, separated by a common electrophoresis procedure and blotted to a PVDF membrane. To compare the sensitivity of each polyclonal rabbit anti-MIF antibody to CHO-MIF, they were applied to the Western Blots at the same concentration (each 3.5 g mL) . The functionality of the rabbit anti human and mouse MIF antibodies were additionally proved by a positive sample (10 ng huMIF and 10 ng moMIF) . As shown in Figure 6, the highest sensitivity for CHQ-MIF was found for the affinity purified rabbit anti CHO-MIF

antibodies (see black arrow, fig. 6).

The lowest concentration of CHO-MIF detected by rabbit anti CHO-MIF antibody was determined with the 0.25 ng/lane which is corresponding to 0,5 ppm i 500 μ-g human an i -MIF antibody preparation (shown in Fig, 7) .

Example 6

Generation of MIF knock out CKO cell lines

The exact knowledge of the genomic structure including exon/intron junctions of a gene locus is a prerequisite for the design of a Zinc finger nuclease (ZFN) (Sangamo - Sigma Aldrich) . The genomic organization of the CHO-MIF gene locus was determined in example 2 , The nuclease was designed to create a double strand break at the exonl/intronl junction,

(Figure 8) . The advantage of the ZFN technology is that both alleles of a gene can be knocked out in a single step with a very high frequency. A MIF knockout cell line was generated and the absence of MIF was demonstrated by genetic

characterization (figure 9 and 9a) and Western Blot analysis

(figure 10 and 10a) using the CHO-MIF specific antibodies described in Example 4. This cell line was used for the expression of anti-MIF antibodies avoiding the problems associated with the binding to its cellular target. a) Generation of unique MIF knockout cell clones.

A CHO cell line stably expressing the anti-MIF antibody RAB0 (RAB0. CHO-S .33 ) was transfected with two plasmids expressing both subunits of the specific ZFH, Under these conditions a functional nuclease is expressed destroying the endogenous MIF locus in the cellular genome. Two weeks after transfection the cell pool was diluted in semisolid medium. After growth for 1 week unique colonies were transferred to 96 well plates using the ClonePix (Genetix Limited) and grown up to small cultures.

Using the same strategy the endogenous MIF was knocked out in different CHO host cell lines like CHO-S and CHO-DG44. b) Genetic characterisation of MIF knockout RA3Q producing CHO-S cell lines.

The chromosomal DNA of unique cell clones was purified using the QIAamp DNA Mini Kit (50) form Qiagen according to the manufacturer's protocol. Using the DNA as a template a specific fragment was amplified by PGR using the following primers: pPCR.choMIFg 9933 : CGTTAATCTGCAGCGTCTACCTGA (SEQ ID NO: 11) and pPCR.choMIFg 9879 GTAAGGCCACTATAGGAAAGCCTG (SEQ ID NO: 12) .

In case of a wild- type (wt ) cell clone the expected fragment is 1260 bp long (figure 8) . In case of knockout cell lines the length is varying depending on the individual structure of the mutated gene . Usually nucleotides are deleted or inserted at the cleavage site of the ZFH. At the same time a recognition site of the restriction enzyme Nael overlapping the ZF cleavage site is destroyed.

The PGR products were cleaved with Nael and the fragments were separated on a 1% agarose gel . Due to the loss of the restriction site in knockout cell lines an altered

restriction pattern is expected (figure 9 and 9a) . c) Western ? Lot εηε.ι ys rs of MIF knockout RAB0 producing CHO-S cell lines

To prove the depletion of MIF protein of MIF, knockout cell- protein extracts were analyzed by western blots . Cell extracts were prepared using a commercially available lysis buffer (#9803 "Cell Signaling") , The samples were separated on an Invitrogen NU Page 4-12% Bis/Tris-Gel 1,5mm x 15 well and transferred to a nitrocellulose membrane,

CHO-MIF was detected by indirect immunofluorescence using a polyclonal rabbit anti-MIF antibody as described in Example 5 as first antibody and anti -rabbit IgG, horseradish peroxidase from Invitrogen as second antibody. The protein pattern was visualised using a Luminescent Image Analyser CB-SG-39

(Figure 10 and 10a) .

Example 7

Production of an Anti-MIF antibody in CHO-MIF knockout cell line

Anti-MIF antibody RABO was produced in a CHO cell line after knock out of the endogenous CHO-MIF. In comparison, the same antibody was produced in a CHO wildtype MIF cell line . The antibody was purified on a Protein A column without any further treatment to remove the CHO-MIF bound to the

antibody. The purified antibody was characterized by Western blot analysis as described in Example 4d. There was no remaining CHO-MIF detectable in the knockout cell line CHO- RABO MIPko.cp75 as compared to the same antibody produced in the wildtype CHO - S cell line (Figure 11 and 11a) .

Example 8

The above described examples 6 and 7 are repeated in an identical fashion, using a CHO cell line either stably expressing anti-MIF antibody RAB or anti-MIF antibody RAB9 resulting in the production of anti-MIF antibody RAB4 or RAB 9, respectively, and using a CHO cell line expressing anti- MIF antibody RAM4 or RAM9 or RAM0 , resulting in the production of anti-MIF antibody RAM4 , AM9 or RAMO , respectively.

Example 9 - Comparison of antibody productivity of MIF wildtype and MIF knockout cell lines producing the anti-MIF antibody RAMO in shake flask fermentation.

RAMOMIFko . CHO-S .33cp75 and the parenteral production cell line RAMO , CHO-S .33 showed highest expression level and cell viability at 28 °C . In this experiment, both cell lines were grown in shake flasks at 37°C to a cell density of

approximately 3x10 s , incubated at 37°C for one day and then shifted to 28°C for another 19 days . Cell counts and

viability were monitored using a CEDEX. Production of RAMO was quantitated by a MIF specific binding ELISA.

The experiment showed the following results (data not shown) :

- Cells are highly viable over a long period of time

- Cells stop growing at 28 °C

- Cells continuously produce the antibody

- The MIFko cell line very surprisingly shows essentially the same characteristics as the parenteral MIFwt cell line .

Example 10 - Comparison of antibody productivity of MIF

wildtype and MIF knockout cell line producing the anti-MIF antibody RAMS in a 3 liter scale batch fermentation.

Anti-MIF antibody KAM9 was produced in a CHC-DG4 cell line RAM9 , CHO-DG44#20 , containing the wildtype MIF gene and

RAM9.CHO-DG44.MIFko#10 containing the knocked out MIF gene . It was surprisingly shown, that similar levels of cell growth and productivity can be reached in both cell lines (data not shown) .