CA2950515A1 - Rsv specific binding molecules and means for producing them - Google Patents

Rsv specific binding molecules and means for producing them Download PDF

Info

Publication number
CA2950515A1
CA2950515A1 CA2950515A CA2950515A CA2950515A1 CA 2950515 A1 CA2950515 A1 CA 2950515A1 CA 2950515 A CA2950515 A CA 2950515A CA 2950515 A CA2950515 A CA 2950515A CA 2950515 A1 CA2950515 A1 CA 2950515A1
Authority
CA
Canada
Prior art keywords
sequence
antibody
cell
ser
rsv
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA2950515A
Other languages
French (fr)
Inventor
Hergen Spits
Tim Beaumont
Mark Jeroen KWAKKENBOS
Etsuko YASUDA
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
MedImmune Ltd
Original Assignee
MedImmune Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by MedImmune Ltd filed Critical MedImmune Ltd
Publication of CA2950515A1 publication Critical patent/CA2950515A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • C07K16/1027Paramyxoviridae, e.g. respiratory syncytial virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • C07K16/1036Retroviridae, e.g. leukemia viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Abstract

The invention provides antibodies and functional equivalents thereof which are capable of specifically binding RSV, and means and methods for producing them.

Description

Title: RSV specific binding molecules and means for producing them This is a divisional of Canadian National Phase Patent Application Serial No.

filed May 30, 2008.
The invention relates to the fields of biology and medicine.
Respiratory Syncytial Virus (RSV) is a common cold virus belonging to the family of paramyxovirus. RSV is virulent, easily transmissible and the most common cause of lower respiratory tract disease in children of less than 2 years .
of age. Up to 98% of children attending day care will be infected in a single RSV
season. Between 0.5% and 3.2% of children with RSV infection require hospitalization. Approximately 90,000 hospital admissions and 4500 deaths per year were reported in United States. Major risk factors for hospitalization due to RSV are premature birth, chronic lung disease, congenital heart disease, compromised immunity, and age younger than 6 weeks in otherwise healthy children. No effective treatment of RSV positive bronchiolitis beside supportive .
care in the form of adequate nutrition and oxygen therapy is available.
Antiviral therapies such as Ribavirin have not been proven to be effective in RSV
infection.
One monoclonal antibody, Palivizumab (also called Synagis), is registered for prophylaxis against RSV infection. Palivizumab is a genetically engineered (humanized) monoclonal antibody to the fusion protein of RSV. However, Palivizumab is not always effective. Therefore, there is a need in the art for =
alternative antibodies and therapies against RSV.
It is an object of the present invention to provide means and methods for counteracting and/or preventing an RSV-related disease. It is a further object of the invention to provide alternative and/or improved antibodies against RSV, or functional equivalents of such antibodies, and to provide stable cells capable of producing antibodies - or functional equivalents thereof - against RSV.
=
The present invention provides antibodies and functional equivalents thereof which are capable of specifically binding RSV. Such antibodies and/or functional equivalents, also called herein "anti-RSV antibodies" or "RSV-specific antibodies", are capable of specifically binding at least one component of RSV, such as for instance an epitope of an RSV protein. Non-specific sticking is not encompassed by the term "specifically binding". Anti-RSV antibodies and functional equivalents according to the present invention are particularly suitable for counteracting and/or at least in part preventing an RSV-infection and/or adverse effects of an RSV infection. One particularly preferred anti-RSV
antibody according to the present invention is the antibody designated "D25", which has a heavy chain region and a light chain region as depicted in Figures 11A-D. The CDR sequences of D25, which in particular contribute to the antigen-binding properties of D25, are depicted in Figure 11D. Antibody D25 appears to have superior characteristics as compared to the registered anti-RSV antibody Palivizumab (Figure 8). For instance, D25 has an 1050 value of about 0.4-1.5 ng/ml in an in vitro neutralization assay wherein HEp-2 cells are infected with RSV, whereas Palivizumab has an 1050 value of about 453 ng/ml.
A functional equivalent of an antibody is defined herein as a functional part, derivative or analogue of an antibody.
A functional part of an antibody is defined as a part which has at least one same property as said antibody in kind, not necessarily in amount. Said functional part is capable of binding the same antigen as said antibody, albeit not necessarily to the same extent. A functional part of an antibody preferably comprises a single domain antibody, a single chain antibody, a single chain variable fragment (scFv), a Fab fragment or a F(ab')2 fragment.
A functional derivative of an antibody is defined as an antibody which has been altered such that at least one property - preferably an antigen-binding property - of the resulting compound is essentially the same in kind, not necessarily in amount. A derivative is provided in many ways, for instance through conservative amino acid substitution, whereby an amino acid residue is substituted by another residue with generally similar properties (size, hydrophobicity, etc), such that the overall functioning is likely not to be seriously affected.
A person skilled in the art is well able to generate analogous compounds of an antibody. This is for instance done through screening of a peptide library or phage display library. Such an analogue has essentially at least one same property as said antibody in kind, not necessarily in amount.
2 As is well known by the skilled person, a heavy chain of an antibody is the larger of the two types of chains making up an immunoglobulin molecule. A
heavy chain comprises constant domains and a variable domain, which variable domain is involved in antigen binding. A light chain of an antibody is the smaller of the two types of chains making up an immunoglobulin molecule. A light chain comprises a constant domain and a variable domain. The variable domain is, together with the variable domain of the heavy chain, involved in antigen binding.
Complementary-determining regions (CDRs) are the hypervariable regions present in heavy chain variable domains and light chain variable domains. The CDRs of a heavy chain and the connected light chain of an antibody together form the antigen-binding site.
Now that the present invention provides the insight that the CDR
sequences depicted in Figure 11 provide desired RSV-binding characteristics, a skilled person is well capable of generating variants comprising at least one altered CDR sequence. For instance, conservative amino acid substitution is applied. Conservative amino acid substitution involves substitution of one amino acid with another with generally similar properties (size, hydrophobicity, etc), such that the overall functioning is likely not to be seriously affected.
It is also possible to change at least one CDR sequence depicted in Figure 11 in order to generate a variant antibody, or a functional equivalent thereof, with at least one altered property as compared to D25. Preferably, an antibody or functional equivalent is provided comprising a CDR sequence which is at least 70% identical to a CDR sequence as depicted in Figure 11, so that the favorable binding characteristics of D25 are at least in part maintained or even improved.
A CDR sequence as depicted in Figure 11 is preferably altered such that the resulting antibody or functional equivalent comprises at least one improved property, such as for instance an improved binding affinity, selectivity and/or stability, as compared to D25. Variant antibodies or functional equivalents thereof comprising an amino acid sequence which is at least 70% identical to a CDR sequence as depicted in Figure 11 are therefore within the scope of the present invention. Various methods are available in the art for altering an amino acid sequence. For instance, a heavy chain or light chain sequence with a desired CDR sequence is artificially synthesized. Preferably, a nucleic acid sequence
3 encoding a CDR sequence is mutated, for instance using random - or site-directed - mutagenesis.
In a first aspect the invention thus provides an isolated, synthetic or recombinant antibody or a functional equivalent thereof which is capable of specifically binding Respiratory Syncytial Virus and which comprises:
- a heavy chain CDR1 sequence comprising a sequence which is at least 70%
identical to the sequence NYIIN, and/or - a heavy chain CDR2 sequence comprising a sequence which is at least 75%
identical to the sequence GIIPVLGTVHYAPKFQG, and/or - a heavy chain CDR3 sequence comprising a sequence which is at least 70%
identical to the sequence ETALVVSTTYLPHYFDN, and/or - a light chain CDR1 sequence comprising a sequence which is at least 85%
identical to the sequence QASQDIVNYLN, and/or - a light chain CDR2 sequence comprising a sequence which is at least 70%
identical to the sequence VASNLET.
Preferably, said antibody also comprises a light chain CDR3 sequence comprising a sequence which is at least 70% identical to the sequence QQYDNLP.
Preferably, an antibody or a functional equivalent according to the invention comprises a CDR sequence which is at least 75%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%
identical to at least one of the CDR sequences depicted in Figure 11D. Most preferably, an antibody or a functional equivalent according to the invention comprises a CDR
sequence which is at least 95% identical to at least one of the CDR sequences depicted in Figure 11D. The particularly preferred antibody D25, described above, comprises CDR sequences which consist of the CDR sequences depicted in Figure 11D. A particularly preferred embodiment according to the invention thus provides an isolated, synthetic or recombinant antibody or a functional equivalent thereof which is capable of specifically binding Respiratory Syncytial Virus and which comprises:
- a heavy chain CDR1 sequence comprising the sequence NYIIN, and/or - a heavy chain CDR2 sequence comprising the sequence GIIPVLGTVHYAPKFQG, and/or - a heavy chain CDR3 sequence comprising the sequence
4 ETALVVSTTYLPHYFDN, and/or - a light chain CDR1 sequence comprising the sequence QASQDIVNYLN, and/or - a light chain CDR2 sequence comprising the sequence VASNLET.
Preferably, said antibody also comprises a light chain CDR3 sequence comprising the sequence QQYDNLP.
In one embodiment an antibody or functional equivalent is provided which comprises the three heavy chain CDR sequences and the three light chain CDR
sequences as depicted in Figure 11D, or sequences that are at least 70%, preferably at least 80%, more preferably at least 85% identical thereto.
Further provided is therefore an isolated, synthetic or recombinant antibody or a functional equivalent thereof which comprises a heavy chain CDR1 sequence comprising a sequence which is at least 70% identical to the sequence NYIIN
and a heavy chain CDR2 sequence comprising a sequence which is at least 70%
identical to the sequence GIIPVLGTVHYAPKFQG and a heavy chain CDR3 sequence comprising a sequence which is at least 70% identical to the sequence ETALVVSTTYLPHYFDN and a light chain CDR1 sequence comprising a sequence which is at least 70% identical to the sequence QASQDIVNYLN and a light chain CDR2 sequence comprising a sequence which is at least 70%
identical to the sequence VASNLET, and a light chain CDR3 sequence comprising a sequence which is at least 70% identical to the sequence QQYDNLP. Said antibody or functional equivalent preferably comprises CDR sequences which are at least 75%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, most preferably at least 95% identical to the heavy chain CDR sequences and the light chain CDR sequences as depicted in Figure 11D. An antibody or functional equivalent comprising the above mentioned heavy chain CDR1, CDR2 and CDR3 sequences as well as the above mentioned light chain CDR1, CDR2 and CDR3 sequences is also provided.
Antibodies or functional equivalents thereof comprising a variable heavy chain amino acid sequence which is at least 70% identical to the heavy chain sequence as depicted in Figure 11 is also provided. Such heavy chain sequences provide desired RSV-binding properties, as evidenced by antibody D25. Further provided is therefore an antibody or a functional equivalent thereof, having a heavy chain sequence comprising a sequence which is at least 70% identical to the sequence QVQLVQSGAEVKKPGSSVMVSCQASGGPLRNYIINWLRQAPGQGPEWMGGII
PVLGTVHYAPKFQGRVTITAD ESTDTAYIHLISLRSEDTAMYYCATETALVVST
TYLPHYFDNWGQGTLVTVSS. Moreover, variable light chain amino acid sequences which are at least 70% identical to the light chain sequence as depicted in Figure 11 also provide desired RSV-binding properties, as evidenced by antibody D25. An antibody, or a functional equivalent thereof having a light chain sequence which is at least 70% identical to the sequence DIQMTQSPSSLSAAVGDRVTITCQASQDIVNYLNWYQQKPGKAPKLLIYVASN
LETGVPSRFSGSGSGTDFSLTISSLQPEDVATYYCQQYDNLPLTFGGGTKVEIK
RTV is therefore also provided. An antibody or functional part according to the invention preferably comprises a variable heavy chain sequence and/or a variable light chain sequence which is at least 75%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, most preferably at least 95% identical to the heavy chain sequence and/or the light chain sequence as depicted in Figure 11. The higher the homology, the more closely said antibody or functional part resembles antibody D25. An antibody or functional part according to the invention preferably comprises a heavy chain as well as a light chain which resemble the heavy and light chain of D25. Further provided is therefore an antibody or functional part comprising a heavy chain sequence and a light chain sequence which are at least 70%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, most preferably at least 95% identical to the heavy chain sequence and the light chain sequence as depicted in Figure 11.
One embodiment provides an antibody or functional equivalent thereof comprising a heavy chain sequence consisting of the heavy chain sequence as depicted in Figure 11, and a light chain sequence consisting of the light chain sequence as depicted in Figure 11. Alternatively, as is well known by the skilled person, it is possible to generate a shortened heavy chain or light chain sequence while maintaining a binding property of interest. Preferably, such a shortened heavy chain or light chain is generated which has a shorter constant region, as compared to the original heavy or light chain. The variable domain is preferably maintained. For instance, a Fab fragment or F(ab')2 fragment based on a heavy chain sequence or light chain sequence depicted in Figure 11 is produced. A
functional equivalent of an antibody comprising at least a functional part of a sequence as depicted in Figure 11 is therefore also provided. Said functional part has a length of at least 20 amino acids and comprises a sequence which is at least 70% identical to the heavy chain CDR1 sequence depicted in Figure 11D, and/or a sequence which is at least 75% identical to the heavy chain CDR2 sequence depicted in Figure 11D, and/or a sequence which is at least 70% identical to the heavy chain CDR3 sequence depicted in Figure 11D, and/or a sequence which is at least 85% identical to the light chain CDR1 sequence depicted in Figure 11D, and/or a sequence which is at least 70% identical to the light chain CDR2 sequence depicted in Figure 11D. Preferably, said functional part also comprises a sequence which is at least 70% identical to the light chain CDR3 sequence depicted in Figure 11D
Another particularly preferred anti-RSV antibody according to the present invention is the antibody designated "AM14", which has a heavy chain region and a light chain region as depicted in Figure 14A. The CDR sequences of AM14, which in particular contribute to the antigen-binding properties of AM14, are also depicted in Figure 14A.
Now that the present invention provides the insight that the CDR
sequences depicted in Figure 14A provide desired RSV-binding characteristics, a skilled person is well capable of generating variants comprising at least one altered CDR sequence. For instance, conservative amino acid substitution is applied. Conservative amino acid substitution involves substitution of one amino acid with another with generally similar properties (size, hydrophobicity, etc), such that the overall functioning is likely not to be seriously affected.
It is also possible to change at least one CDR sequence depicted in Figure 14A in order to generate a variant antibody, or a functional equivalent thereof, with at least one altered property as compared to AM14. Preferably, an antibody or functional equivalent is provided comprising a CDR sequence which is at least 70% identical to a CDR sequence as depicted in Figure 14A, so that the favorable binding characteristics of A1\'114 are at least in part maintained or even improved.
A CDR sequence as depicted in Figure 14A is preferably altered such that the resulting antibody or functional equivalent comprises at least one improved property, such as for instance an improved binding affinity, selectivity and/or stability, as compared to AM14. Variant antibodies or functional equivalents thereof comprising an amino acid sequence which is at least 70% identical to a CDR sequence as depicted in Figure 14A are therefore within the scope of the present invention. Various methods are available in the art for altering an amino acid sequence. For instance, a heavy chain or light chain sequence with a desired CDR sequence is artificially synthesized. Preferably, a nucleic acid sequence encoding a CDR sequence is mutated, for instance using random - or site-directed - mutagenesis.
In one aspect the invention thus provides an isolated, synthetic or recombinant antibody or a functional part, derivative and/or analogue thereof which is capable of specifically binding Respiratory Syncytial Virus and which comprises:
- a heavy chain CDR1 sequence comprising a sequence which is at least 70%
identical to the sequence GFSFSHYA, and/or - a heavy chain CDR2 sequence comprising a sequence which is at least 70%
identical to the sequence ISYDGENT, and/or - a heavy chain CDR3 sequence comprising a sequence which is at least 70%
identical to the sequence ARDRIVDDYYYYGMDV, and/or - a light chain CDR1 sequence comprising a sequence which is at least 70%
identical to the sequence QDIKKY, and/or - a light chain CDR2 sequence comprising a sequence which is at least 70%
identical to the sequence DAS, and/or - a light chain CDR3 sequence comprising a sequence which is at least 70%
identical to the sequence QQYDNLPPLT.
Preferably, an antibody or a functional equivalent according to the invention comprises a CDR sequence which is at least 75%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%
identical to at least one of the CDR sequences depicted in Figure 14A. Most preferably, an antibody or a functional equivalent according to the invention comprises a CDR
sequence which is at least 95% identical to at least one of the CDR sequences depicted in Figure 14A. The particularly preferred antibody AM14, described above, comprises CDR sequences which consist of the CDR sequences depicted in Figure 14A. A particularly preferred embodiment according to the invention thus provides an isolated, synthetic or recombinant antibody or a functional equivalent thereof which is capable of specifically binding Respiratory Syncytial Virus and which comprises:
- a heavy chain CDR1 sequence comprising the sequence GFSFSHYA, and/or - a heavy chain CDR2 sequence comprising the sequence ISYDGENT, and/or - a heavy chain CDR3 sequence comprising the sequence ARDRIVDDYYYYGMDV, and/or - a light chain CDR1 sequence comprising the sequence QDIKKY, and/or - a light chain CDR2 sequence comprising the sequence DAS, and/or - a light chain CDR3 sequence comprising the sequence QQYDNLPPLT.
In one embodiment an antibody or functional equivalent is provided which comprises the three heavy chain CDR sequences and the three light chain CDR
sequences as depicted in Figure 14A, or sequences that are at least 70%
identical thereto. Further provided is therefore an isolated, synthetic or recombinant antibody or a functional equivalent thereof which comprises a heavy chain CDR1 sequence comprising a sequence which is at least 70% identical to the sequence GFSFSHYA and a heavy chain CDR2 sequence comprising a sequence which is at least 70% identical to the sequence ISYDGENT and a heavy chain CDR3 sequence comprising a sequence which is at least 70% identical to the sequence ARDRIVDDYYYYGMDV and a light chain CDR1 sequence comprising a sequence which is at least 70% identical to the sequence QDIKKY and a light chain CDR2 sequence comprising a sequence which is at least 70% identical to the sequence DAS, and a light chain CDR3 sequence comprising a sequence which is at least 70% identical to the sequence QQYDNLPPLT. Said antibody or functional equivalent preferably comprises CDR sequences which are at least 75%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, most preferably at least 95% identical to the heavy chain CDR
sequences and the light chain CDR sequences as depicted in Figure 14A. An antibody or functional equivalent comprising the above mentioned heavy chain CDR1, CDR2 and CDR3 sequences of Figure 14A as well as the above mentioned light chain CDR1, CDR2 and CDR3 sequences of Figure 14A is also provided.
Antibodies or functional equivalents thereof comprising a heavy chain amino acid sequence which is at least 70% identical to a heavy chain sequence as depicted in Figure 14A is also provided. Such heavy chain sequences provide desired RSV-binding properties, as evidenced by antibody AM14. Further provided is therefore an antibody or a functional equivalent thereof, having a heavy chain sequence comprising a sequence which is at least 70% identical to the sequence EVQLVESGGGVVQPGRSLRLSCAASGFSFSHYAMHWVRQAPGKGLEWVAVIS
YDGENTYYADSVKGRFSISRDNSKNTVSLQMNSLRPEDTALYYCARDRIVDD
YYYYGNIDVWGQGATVTVSS. Moreover, light chain amino acid sequences which are at least 70% identical to a light chain sequence as depicted in Figure 14A also provide desired RSV-binding properties, as evidenced by antibody AM14.
An antibody, or a functional equivalent thereof having a light chain sequence which is at least 70% identical to the sequence DIQMTQSPSSLSASVGDRVTITCQASQDIKKYLNWYHQKPGKVPELLMHDASNLETGVPSRF
SGRGSGTDFTLTISSLQPEDIGTYYCQQYDNLPPLTEGGGTKVEIKRTV is therefore also provided. An antibody or functional part according to the invention preferably comprises a variable heavy chain sequence and/or a variable light chain sequence which is at least 75%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, most preferably at least 95% identical to a heavy chain sequence and/or a light chain sequence as depicted in Figure 14A. The higher the homology, the more closely said antibody or functional part resembles antibody AM14. An antibody or functional part according to the invention preferably comprises a heavy chain as well as a light chain which resemble the heavy and light chain of AM14. Further provided is therefore an antibody or functional part comprising a heavy chain sequence and a light chain sequence which are at least 70%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, most preferably at least 95% identical to the heavy chain sequence and the light chain sequence as depicted in Figure 14A.
One embodiment provides an antibody or functional equivalent thereof comprising a heavy chain sequence consisting of the heavy chain sequence as depicted in Figure 14A, and a light chain sequence consisting of the light chain sequence as depicted in Figure 14A. Alternatively, as is well known by the skilled person, it is possible to generate a shortened heavy chain or light chain sequence while maintaining a binding property of interest. Preferably, such a shortened heavy chain or light chain is generated which has a shorter constant region, as compared to the original heavy or light chain. The variable domain is preferably maintained. For instance, a Fab fragment or F(ab')2 fragment based on a heavy chain sequence or light chain sequence depicted in Figure 14A is produced. A
functional equivalent of an antibody comprising at least a functional part of a sequence as depicted in Figure 14A is therefore also provided. Said functional part has a length of at least 20 amino acids and comprises a sequence which is at least 70% identical to at least one of the CDR sequences depicted in Figure 14A.
Another particularly preferred anti-RSV antibody according to the present invention is the antibody designated "AM16", which has a heavy chain region and a light chain region as depicted in Figure 14B. The CDR sequences of AM16, which in particular contribute to the antigen-binding properties of AM16, are also depicted in Figure 14B.
Now that the present invention provides the insight that the CDR
sequences depicted in Figure 14B provide desired RSV-binding characteristics, a skilled person is well capable of generating variants comprising at least one altered CDR sequence. For instance, conservative amino acid substitution is applied. Conservative amino acid substitution involves substitution of one amino acid with another with generally similar properties (size, hydrophobicity, etc), such that the overall functioning is likely not to be seriously affected.
It is also possible to change at least one CDR sequence depicted in Figure 14B in order to generate a variant antibody, or a functional equivalent thereof, with at least one altered property as compared to A1V116. Preferably, an antibody or functional equivalent is provided comprising a CDR sequence which is at least 70% identical to a CDR sequence as depicted in Figure 14B, so that the favorable binding characteristics of AM16 are at least in part maintained or even improved.
A CDR sequence as depicted in Figure 14B is preferably altered such that the resulting antibody or functional equivalent comprises at least one improved property, such as for instance an improved binding affinity, selectivity and/or stability, as compared to AM16. Variant antibodies or functional equivalents thereof comprising an amino acid sequence which is at least 70% identical to a CDR sequence as depicted in Figure 14B are therefore within the scope of the present invention. Various methods are available in the art for altering an amino acid sequence. For instance, a heavy chain or light chain sequence with a desired CDR sequence is artificially synthesized. Preferably, a nucleic acid sequence encoding a CDR sequence is mutated, for instance using random - or site-directed - mutagenesis.

In one aspect the invention thus provides an isolated, synthetic or recombinant antibody or a functional part, derivative and/or analogue thereof which is capable of specifically binding Respiratory Syncytial Virus and which comprises:
- a heavy chain CDR1 sequence comprising a sequence which is at least 70%
identical to the sequence GFTFSSYN, and/or - a heavy chain CDR2 sequence comprising a sequence which is at least 70%
identical to the sequence ISAGSSYI, and/or - a heavy chain CDR3 sequence comprising a sequence which is at least 70%
identical to the sequence AREDYGPGNYYSPNWFDP, and/or - a light chain CDR1 sequence comprising a sequence which is at least 70%
identical to the sequence SSNIGAGYD, and/or - a light chain CDR2 sequence comprising a sequence which is at least 70%
identical to the sequence GNT, and/or - a light chain CDR3 sequence comprising a sequence which is at least 70%
identical to the sequence HSYDRSLSG.
Preferably, an antibody or a functional equivalent according to the invention comprises a CDR sequence which is at least 75%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%
identical to at least one of the CDR sequences depicted in Figure 14B. Most preferably, an antibody or a functional equivalent according to the invention comprises a CDR

sequence which is at least 95% identical to at least one of the CDR sequences depicted in Figure 14B. The particularly preferred antibody AM16, described above, comprises CDR sequences which consist of the CDR sequences depicted in Figure 14B. A particularly preferred embodiment according to the invention thus provides an isolated, synthetic or recombinant antibody or a functional equivalent thereof which is capable of specifically binding Respiratory Syncytial Virus and which comprises:
- a heavy chain CDR1 sequence comprising the sequence GFTFSSYN, and/or - a heavy chain CDR2 sequence comprising the sequence ISAGSSYI, and/or - a heavy chain CDR3 sequence comprising the sequence AREDYGPGNYYSPNWFDP, and/or - a light chain CDR1 sequence comprising the sequence SSNIGAGYD, and/or - a light chain CDR2 sequence comprising the sequence GNI', and/or - a light chain CDR3 sequence comprising the sequence HSYDRSLSG.
In one embodiment an antibody or functional equivalent is provided which comprises the three heavy chain CDR sequences and the three light chain CDR
sequences as depicted in Figure 14B, or sequences that are at least 70%
identical thereto. Further provided is therefore an isolated, synthetic or recombinant antibody or a functional equivalent thereof which comprises a heavy chain CDR1 sequence comprising a sequence which is at least 70% identical to the sequence GFTFSSYN and a heavy chain CDR2 sequence comprising a sequence which is at least 70% identical to the sequence ISAGSSYI and a heavy chain CDR3 sequence comprising a sequence which is at least 70% identical to the sequence AREDYGPGNYYSPNWFDP and a light chain CDR1 sequence comprising a sequence which is at least 70% identical to the sequence SSNIGAGYD and a light chain CDR2 sequence comprising a sequence which is at least 70% identical to the sequence GNT, and a light chain CDR3 sequence comprising a sequence which is at least 70% identical to the sequence HSYDRSLSG. Said antibody or functional equivalent preferably comprises CDR sequences which are at least 75%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, most preferably at least 95% identical to the above mentioned heavy chain CDR sequences and the above mentioned light chain CDR sequences as depicted in Figure 14B. An antibody or functional equivalent comprising the above mentioned heavy chain CDR1, CDR2 and CDR3 sequences of Figure 14B
as well as the above mentioned light chain CDR1, CDR2 and CDR3 sequences of Figure 14B is also provided.
Antibodies or functional equivalents thereof comprising a heavy chain amino acid sequence which is at least 70% identical to a heavy chain sequence as depicted in Figure 14B is also provided. Such heavy chain sequences provide desired RSV-binding properties, as evidenced by antibody AM16. Further provided is therefore an antibody or a functional equivalent thereof, having a heavy chain sequence comprising a sequence which is at least 70% identical to the sequence EVQLVETGGGLAQPGGSLRLSCAASGFTFSSYNMNWVRQAPGKGLEWVS HI SAGS SY IYYS D
SVKGRFTVSRDNVRNSVYLQMNSLRAADTAVYYCARE DYGPGNYYSPNWFDPWGQGTLVTVS
S. Moreover, light chain amino acid sequences which are at least 70% identical to =
a light chain sequence as depicted in Figure 14B also provide desired RSV-binding properties, as evidenced by antibody AM16. An antibody, or a functional equivalent thereof having a light chain sequence which is at least 70%
identical to the sequence QSVVTQPPSVSGAPGQRVTISCTGSSSNIGAGYDVHWYQQLPGTAPKLLIYGNTNRPSGVSD
RFSGSKSGTSASLAITGLQAEDEADYYCHSYDRSLSGSVEGGGTKLTV is therefore also provided. An antibody or functional part according to the invention preferably comprises a variable heavy chain sequence and/or a variable light chain sequence which is at least 75%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, most preferably at least 95% identical to the heavy chain sequence and/or the light chain sequence as depicted in Figure 14B. The higher the homology, the more closely said antibody or functional part resembles antibody AM16. An antibody or functional part according to the invention preferably comprises a heavy chain as well as a light chain which resemble the heavy and light chain of AM16. Further provided is therefore an antibody or functional part comprising a heavy chain sequence and a light chain sequence which are at least 70%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, most preferably at least 95% identical to the heavy chain sequence and the light chain sequence as depicted in Figure 14B.
One embodiment provides an antibody or functional equivalent thereof comprising a heavy chain sequence consisting of the heavy chain sequence as depicted in Figure 14B, and a light chain sequence consisting of the light chain sequence as depicted in Figure 14B. Alternatively, as is well known by the skilled person, it is possible to generate a shortened heavy chain or light chain sequence while maintaining a binding property of interest. Preferably, such a shortened heavy chain or light chain is generated which has a shorter constant region, as compared to the original heavy or light chain. The variable domain is preferably maintained. For instance, a Fab fragment or F(ab')2 fragment based on a heavy chain sequence or light chain sequence depicted in Figure 14B is produced. A
functional equivalent of an antibody comprising at least a functional part of a sequence as depicted in Figure 14B is therefore also provided. Said functional part has a length of at least 20 amino acids and comprises a sequence which is at least 70% identical to at least one of the CDR sequences depicted in Figure 14B.

=

Another particularly preferred anti-RSV antibody according to the present invention is the antibody designated "AM23", which has a heavy chain region and a light chain region as depicted in Figure 14C. The CDR sequences of AM23, which in particular contribute to the antigen-binding properties of AM23, are also depicted in Figure 14C.
Now that the present invention provides the insight that the CDR
sequences depicted in Figure 14C provide desired RSV-binding characteristics, a skilled person is well capable of generating variants comprising at least one altered CDR sequence. For instance, conservative amino acid substitution is applied. Conservative amino acid substitution involves substitution of one amino acid with another with generally similar properties (size, hydrophobicity, etc), such that the overall functioning is likely not to be seriously affected.
It is also possible to change at least one CDR sequence depicted in Figure 14C in order to generate a variant antibody, or a functional equivalent thereof, with at least one altered property as compared to AM23. Preferably, an antibody or functional equivalent is provided comprising a CDR sequence which is at least 70% identical to a CDR sequence as depicted in Figure 14C, so that the favorable binding characteristics of AM23 are at least in part maintained or even improved.
A CDR sequence as depicted in Figure 14C is preferably altered such that the resulting antibody or functional equivalent comprises at least one improved property, such as for instance an improved binding affinity, selectivity and/or stability, as compared to AM23. Variant antibodies or functional equivalents thereof comprising an amino acid sequence which is at least 70% identical to a CDR sequence as depicted in Figure 14C are therefore within the scope of the present invention. Various methods are available in the art for altering an amino acid sequence. For instance, a heavy chain or light chain sequence with a desired CDR sequence is artificially synthesized. Preferably, a nucleic acid sequence encoding a CDR sequence is mutated, for instance using random - or site-directed - mutagenesis.
In one aspect the invention thus provides an isolated, synthetic or recombinant antibody or a functional part, derivative and/or analogue thereof which is capable of specifically binding Respiratory Syncytial Virus and which comprises:
- a heavy chain CDR1 sequence comprising a sequence which is at least 70%

identical to the sequence GFNFHNYG, and/or - a heavy chain CDR2 sequence comprising a sequence which is at least 70%
identical to the sequence VWYDGSKK, and/or - a heavy chain CDR3 sequence comprising a sequence which is at least 70%
identical to the sequence VRDKVGPTPYFDS, and/or - a light chain CDR1 sequence comprising a sequence which is at least 70%
identical to the sequence NIGSET, and/or - a light chain CDR2 sequence comprising a sequence which is at least 70%
identical to the sequence DDD, and/or - a light chain CDR3 sequence comprising a sequence which is at least 70%
identical to the sequence QVWDRSNYHQV.
Preferably, an antibody or a functional equivalent according to the invention comprises a CDR sequence which is at least 75%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%
identical to at least one of the CDR sequences depicted in Figure 14C. Most preferably, an antibody or a functional equivalent according to the invention comprises a CDR

sequence which is at least 95% identical to at least one of the CDR sequences depicted in Figure 14C. The particularly preferred antibody AM23, described above, comprises CDR sequences which consist of the CDR sequences depicted in Figure 14C. A particularly preferred embodiment according to the invention thus provides an isolated, synthetic or recombinant antibody or a functional equivalent thereof which is capable of specifically binding Respiratory Syncytial Virus and which comprises:
- a heavy chain CDR1 sequence comprising the sequence GFNFHNYG, and/or - a heavy chain CDR2 sequence comprising the sequence VVVYDGSKK, and/or - a heavy chain CDR3 sequence comprising the sequence VRDKVGPTPYFDS, and/or - a light chain CDR1 sequence comprising the sequence NIGSET, and/or - a light chain CDR2 sequence comprising the sequence DDD, and/or - a light chain CDR3 sequence comprising the sequence QVWDRSNYHQV.
In one embodiment an antibody or functional equivalent is provided which comprises the three heavy chain CDR sequences and the three light chain CDR
sequences as depicted in Figure 14C, or sequences that are at least 70%
identical thereto. Further provided is therefore an isolated, synthetic or recombinant antibody or a functional equivalent thereof which comprises a heavy chain CDR1 sequence comprising a sequence which is at least 70% identical to the sequence GFNFHNYG and a heavy chain CDR2 sequence comprising a sequence which is at least 70% identical to the sequence VWYDGSKK and a heavy chain CDR3 sequence comprising a sequence which is at least 70% identical to the sequence VRDKVGPTPYFDS and a light chain CDR1 sequence comprising a sequence which is at least 70% identical to the sequence NIGSET and a light chain CDR2 sequence comprising a sequence which is at least 70% identical to the sequence DDD, and a light chain CDR3 sequence comprising a sequence which is at least 70% identical to the sequence QVWDRSNYHQV. Said antibody or functional equivalent preferably comprises CDR sequences which are at least 75%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, most preferably at least 95% identical to the above mentioned heavy chain CDR sequences and the above mentioned light chain CDR sequences as depicted in Figure 14C. An antibody or functional equivalent comprising the above mentioned heavy chain CDR1, CDR2 and CDR3 sequences of Figure 14C as well as the above mentioned light chain CDR1, CDR2 and CDR3 sequences of Figure 14C is also provided.
Antibodies or functional equivalents thereof comprising a heavy chain amino acid sequence which is at least 70% identical to a heavy chain sequence as depicted in Figure 14C is also provided. Such heavy chain sequences provide desired RSV-binding properties, as evidenced by antibody AM23. Further provided is therefore an antibody or a functional equivalent thereof, having a heavy chain sequence comprising a sequence which is at least 70% identical to the sequence EVQLVESGGNVVKPGTSLRLSCAATGFNFHNYGMNWVRQAPGKGLEWVAVVWYDGSKKYYAD
SVT GREAT SRDNS KNTLYLQMNS LRVE DTAVYYCVRDKVGPTPYFDSWGQGTLVTVS S.
Moreover, light chain amino acid sequences which are at least 70% identical to a light chain sequence as depicted in Figure 14C also provide desired RSV-binding properties, as evidenced by antibody AM23. An antibody, or a functional equivalent thereof having a light chain sequence which is at least 70%
identical to the sequence SYVLTQPPSVSLAPGGTAAI TCGRNNIGSETVHWYQQKPGQAPVLVVYDDDDRPS GI PERFS
GSNSGNTATLT I SRVEAGDEADYYCQVWDRSNYHQVFGGGTKLTV is therefore also provided. An antibody or functional part according to the invention preferably comprises a variable heavy chain sequence and/or a variable light chain sequence which is at least 75%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, most preferably at least 95% identical to the heavy chain sequence and/or the light chain sequence as depicted in Figure 14C. The higher the homology, the more closely said antibody or functional part resembles antibody AM23. An antibody or functional part according to the invention preferably comprises a heavy chain as well as a light chain which resemble the heavy and light chain of AM23. Further provided is therefore an antibody or functional part comprising a heavy chain sequence and a light chain sequence which are at least 70%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, most preferably at least 95% identical to the heavy chain sequence and the light chain sequence as depicted in Figure 14C.
One embodiment provides an antibody or functional equivalent thereof comprising a heavy chain sequence consisting of the heavy chain sequence as depicted in Figure 14C, and a light chain sequence consisting of the light chain sequence as depicted in Figure 14C. Alternatively, as is well known by the skilled person, it is possible to generate a shortened heavy chain or light chain sequence while maintaining a binding property of interest. Preferably, such a shortened heavy chain or light chain is generated which has a shorter constant region, as compared to the original heavy or light chain. The variable domain is preferably maintained. For instance, a Fab fragment or F(ab1)2 fragment based on a heavy chain sequence or light chain sequence depicted in Figure 14C is produced. A
functional equivalent of an antibody comprising at least a functional part of a sequence as depicted in Figure 14C is therefore also provided. Said functional part has a length of at least 20 amino acids and comprises a sequence which is at least 70% identical to at least one of the CDR sequences depicted in Figure 14C.
The present invention provides RSV-specific antibodies or functional equivalents thereof having improved properties as compared to prior art antibodies. The inventors have succeeded in generating RSV-specific antibodies with low 1050 values. Such antibodies have a particular high or strong affinity for RSV and are therefore particularly suitable for counteracting and/or at least in part preventing an RSV-infection and/or adverse effects of an RSV
infection.
One embodiment provides an antibody which has an IC50 value of less than 10 ng/ml in an in vitro neutralization assay wherein HEp-2 cells are infected with RSV, and a functional equivalent of said antibody. Said antibody or functional equivalent preferably has an IC50 value of less than 5 ng/ml, more preferably less than 2 ng/ml. The preferred antibody D25 has an IC50 value of about 0.5-1.5 ng/ml in the in vitro neutralization assay described in the examples (see Figure 8).
An antibody according to the invention is preferably a human antibody.
The use of human antibodies for human therapy diminishes the chance of side-effects due to an immunological reaction in a human individual against non-human sequences. In another preferred embodiment an antibody or functional part, derivative or analogue according to the invention is a chimeric antibody.
This way, sequences of interest, such as for instance a binding site of interest, can be included into an antibody or functional equivalent according to the invention.
The invention further provides an isolated, synthetic or recombinant nucleic acid sequence, or a functional part, derivative or analogue thereof, encoding an antibody or functional equivalent according to the invention. Such nucleic acid is for instance isolated from a B-cell which is capable of producing an antibody according to the invention, as outlined in more detail below. A
preferred embodiment provides a nucleic acid sequence comprising a sequence which is at least 70% homologous to at least a functional part of a nucleic acid sequence as depicted in Figure 11, Figure 12, Figure 14A, Figure 14B and/or Figure 14B.
Said nucleic acid sequence preferably comprises a sequence which is at least 75%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, most preferably at least 95% homologous to at least a functional part of a nucleic acid sequence as depicted in Figure 11, Figure 12, Figure 14A, Figure 14B and/or Figure 14B. Said functional part has a length of at least 30 nucleotides, preferably at least 50 nucleotides, more preferably at least 75 nucleotides. Preferably, said functional part encodes at least one nucleic acid sequence as depicted in Figure 11D, Figure 12, Figure 14A, Figure 14B and/or Figure 14B. Said sequence is preferably a CDR sequence.

An antibody or functional equivalent according to the invention is particularly suitable for use as a medicine or prophylactic agent. An antibody according to the invention, or a functional part, derivative or analogue thereof, for use as a medicament and/or prophylactic agent is therefore also herewith provided. In a particularly preferred embodiment said antibody comprises antibody D25, AM14, AM16 and/or AM23, or a functional part, derivative or analogue thereof. Said medicament or prophylactic agent is preferably used for counteracting or at least in part preventing an RSV-infection or for counteracting or at least in part preventing adverse effects of an RSV-infection. A use of an antibody, functional part, derivative or analogue according to the invention for the preparation of a medicament and/or prophylactic agent for at least in part treating and/or preventing a RSV-related disorder is therefore also provided, as well as a method for at least in part treating or preventing an RSV-related disorder, the method comprising administering to an individual in need thereof a therapeutically effective amount of an antibody or functional equivalent according to the invention. Said antibody preferably comprises antibody D25, AM14, AM16 and/or A1V123, or a functional part, derivative or analogue thereof.
In order to counteract RSV, an antibody or functional equivalent according to the invention is preferably administered to an individual before an RSV-infection has taken place. Alternatively, an antibody or functional equivalent according to the invention is administered when an individual is already infected by RSV. Said antibody or functional equivalent is preferably administered to individuals with an increased risk of RSV-related disorders, such as for instance children with premature birth, individuals with chronic lung disease, congenital heart disease and/or compromised immunity, and children with an age younger than 6 weeks. Also elderly people have an increased risk of RSV-related disorders. Antibodies or functional equivalents according to the invention are preferably administered orally or via one or more injections.
Dose ranges of antibodies and/or functional equivalents according to the invention to be used in the therapeutic applications as described herein before are designed on the basis of rising dose studies in the clinic in clinical trials for which rigorous protocol requirements exist. Typical doses are between 0.1 and 10 mg per kg body weight. For therapeutic application, antibodies or functional equivalents according to the invention are typically combined with a pharmaceutically acceptable carrier, adjuvant, diluent and/or excipient. Examples of suitable carriers for instance comprise keyhole limpet haemocyanin (KLH), serum albumin (e.g. BSA or RSA) and ovalbumin. Many suitable adjuvants, oil-based and water-based, are known to a person skilled in the art. In one embodiment said adjuvant comprises Specol. In another embodiment, said suitable carrier comprises a solution like for example saline.
In yet another embodiment a nucleic acid encoding an antibody or functional part according to the invention is used. Upon administration of such nucleic acid, antibodies or functional equivalents are produced by the host's machinery. Produced antibodies or functional equivalents are capable of preventing and/or counteracting RSV-infection and/or the adverse effects of an RSV-infection. A nucleic acid sequence, functional part, derivative and/or analogue according to the invention for use as a medicament and/or prophylactic agent is therefore also herewith provided. Said nucleic acid is preferably used for counteracting RSV. Further provided is therefore a use of a nucleic acid sequence, functional part, derivative and/or analogue according to the invention for the preparation of a medicament and/or prophylactic agent for at least in part treating and/or preventing a RSV-related disorder.
By at least a functional part of a nucleic acid of the invention is meant a part of said nucleic acid, at least 30 base pairs long, preferably at least 50 base pairs long, more preferably at least 100 base pairs long, comprising at least one expression characteristic (in kind not necessarily in amount) as a nucleic acid of the invention. Said functional part at least encodes an amino acid sequence comprising a sequence which is at least 70% identical to a CDR sequence as depicted in Figure 11D, Figure 14A, Figure 14B and/or Figure 14C.
The invention furthermore provides an isolated antibody producing cell capable of producing an antibody, functional part, derivative or analogue according to the invention. Possible (but not limiting) ways of obtaining such antibody producing cells are outlined in detail in the examples. The inventors have developed and used a new method in order to improve the stability of RSV-specific antibody producing cells. Using this method, RSV-specific antibody producing cells are generated which are stable for at least six months. An RSV-specific antibody producing cell according to the invention, which is stable for at least nine weeks, preferably for at least three months, more preferably for at least six months is therefore also herewith provided.
The present inventors have used their insight that the stability of an RSV-specific antibody producing cell is influenced by influencing the amount of BCL6 and/or Blimp-1 expression product within said antibody producing cell.
The amount of BCL6 and/or Blimp-1 expression product is either directly or indirectly influenced. Preferably the amounts of both BCL6 and Blimp-1 expression products within said antibody producing cell are regulated, since both expression products are involved in the stability of an antibody producing cell. The stability of an antibody producing cell is defined as the capability of said antibody producing cell to remain in a certain developmental stage (preferably after said cell has been brought into said stage). Different developmental stages of a cell involve at least one different characteristic of said cell. For instance, a memory B cell is known to differentiate upon stimulation into an antibody-secreting plasma cell via a stage which some researchers call a plasmablast. A memory B cell, a plasmablast and a plasma cell are different developmental stages of a B cell, wherein the B cell has different characteristics. A memory B cell exhibits low proliferation and antibody secretion. A plasmablast exhibits both higher proliferation and higher antibody secretion levels as compared to a memory B
cell, whereas a plasma cell secretes high antibody levels but is not capable of proliferating. With a method of the present inventors it has become possible to regulate the replicative life span of an antibody producing cell. A
replicative life span of an antibody producing cell is defined herein as the time span wherein a B cell and its progeny cells are capable of replicating while maintaining their capability of producing antibody and/or developing into a cell that produces antibody. Preferably the replicative life span of an antibody producing cell is prolonged, meaning that said antibody producing cell will not terminally differentiate - or only after a longer period as compared to the same kind of antibody producing cells that are currently used - and continue to proliferate in vitro. According to the inventors it is possible to regulate the amount of and/or Blimp-1 expression product in an antibody producing cell to such extent that the antibody producing cell is brought into, and/or kept in, a predetermined developmental state in which the cells continue to proliferate. With a method of the inventors it has therefore become possible to increase the replicative life span =

of an antibody producing cell since it is possible to maintain a B cell in a certain developmental stage wherein replication occurs. Reference is made to PCT/NL2006/000625, filed by the same applicant. The present invention provides means and methods for producing stable RSV-specific antibody producing cells.
An antibody producing cell is defined as a cell which cell is capable of producing and/or secreting antibody or a functional equivalent thereof, and/or which cell is capable of developing into a cell which is capable of producing and/or secreting antibody or a functional equivalent thereof. An RSV-specific antibody producing cell is defined herein as a cell capable of producing and/or secreting antibodies or functional equivalents thereof which are capable of specifically binding RSV and/or a component of RSV, such as for instance an epitope of the RSV F (fusion) protein, the RSV G (attachment) protein or RSV SH (small hydrophobic) protein. Preferably, said RSV-specific antibody producing cell comprises a B cell ancUor a B cell-derived plasma cell. A B cell is called herein an antibody producing cell, even when the B cell is in a stage wherein antibody production is low or not present at all, such as a naïve B cell or a memory B
cell, being activated or not, because such cells are capable of developing into cells that produce antibody, such as a plasmablast and/or plasma cell.
An RSV-specific antibody producing cell according to the invention preferably comprises a mammalian cell. Non-limiting examples include antibody producing cells derived from a human individual, rodent, rabbit, llama, pig, cow, goat, horse, ape, gorilla. Preferably, said antibody producing cell comprises a human cell, a murine cell, a rabbit cell and/or a llama cell.
BCL6 encodes a transcriptional repressor which is required for normal B cell and T cell development and maturation and which is required for the formation of germinal centers. (Ye, 1997). BCL6 is highly expressed in germinal center B cells whereas it is hardly expressed in plasma cells. BCL6 inhibits differentiation of activated B cells into plasma cells. The transcriptional repressor B lymphocyte induced maturation protein-1 (Blimp-1) is required for development of a B cell into a plasma cell. The human variant of Blimp-1 is named Prdml. As used herein, any reference to Blimp-1 includes a reference to Prdm1. Blimp-1 drives plasma cell differentiation. BCL6 and Blimp-1 repress =

expression of the other; thus in a natural situation when one reaches an higher expression level than the other, the stage of differentiation is enforced. In the human body, differentiation of plasma cells from activated naïve or memory B
cells involves downregulation of BCL6 and upregulation of Blimp-1. In germinal center cells BCL6 expression is high and Blimp-1 expression is low. In resting memory cells expression of BCL6 and Blimp-1 are low. Signals that trigger differentiation cause an upregulation of Blimp-1, and this Blimp-1 counteracts the expression of BCL6. The stage where both BCL6 and Blimp-1 are expressed is short-lived and is called a plasmablast. With progressively increasing Blimp-1 levels, BCL6 expression is extinguished, resulting in a plasma cell.
In one embodiment of the present invention, an RSV-specific antibody producing cell is provided wherein BCL6 and Blimp-1 are co-expressed (meaning that both BCL6 and Blimp-1 are expressed in said antibody producing cell for at least 1 day, preferably at least one week, more preferably at least six weeks, most preferably at least three months. Said RSV-specific antibody producing cell is capable of proliferating when an appropriate signal is provided. It has been found that co-expression of BCL6 and Blimp-1 results in an antibody producing cell which is capable of both proliferating and producing antibody. BCL6 and Blimp-are preferably co-expressed in a B cell, preferably a human B cell. Co-expression of BCL6 and Blimp-1 in a B cell results in stabilization of said B cell in a plasmablast-like stage. Plasmablasts, like plasma cells, are capable of secreting antibody. However, plasmablasts are still capable of proliferating, whereas plasma cells have lost their capability of proliferating. Plasma cells are therefore unsuitable for culturing antibody-producing cell lines.
One preferred embodiment provides an RSV-specific antibody producing cell comprising an exogenous nucleic acid sequence encoding BCL6 or a functional part, derivative and/or analogue thereof. An exogenous nucleic acid is defined herein as a nucleic acid sequence which does not naturally belong to the genome of a cell. With such exogenous nucleic acid molecule it is possible to regulate a BCL6 concentration in an antibody producing cell independently from expression of endogenous BCL6. Hence, even if expression of endogenous BCL6 is low or absent, for instance caused by Blimp-1, an exogenous nucleic acid sequence encoding BCL6 or a functional part, derivative and/or analogue thereof =

= is still capable of producing a concentration of BCL6 which is sufficient for influencing the stability of an antibody producing cell. Preferably, said nucleic acid sequence encoding BCL6 or a functional part, derivative and/or analogue thereof is constitutively active, so that BCL6 expression is maintained even when endogenous BCL6 expression of said cell is inhibited by an endogenous repressor such as Blimp-1. Most preferably, expression of said nucleic acid sequence encoding BCL6 or a functional part, derivative and/or analogue thereof is regulated by an exogenous inducer of repressor, so that the extent of BCL6 expression is regulated at will.
Preferably, as outlined below in more detail, an RSV-specific antibody producing cell according to the invention comprises an exogenous nucleic acid sequence encoding Bc1-xL or a functional part, derivative and/or analogue thereof.
If Bc1-xL or a functional part, derivative and/or analogue thereof is present, it is possible to grow plasmablasts under conditions of low cell density. Expression of said nucleic acid sequence encoding Bc1-xL or a functional part, derivative and/or analogue thereof is preferably regulated by an exogenous inducer of repressor, so that the extent of Bc1-xL expression is regulated at will. A preferred embodiment therefore provides an RSV-specific antibody producing cell comprising:
- an exogenous nucleic acid sequence encoding BCL6 or a functional part, derivative and/or analogue thereof, and/or - an exogenous nucleic acid sequence encoding Bc1-xL or a functional part, derivative and/or analogue thereof. Said RSV-specific antibody producing cell preferably comprises both an exogenous nucleic acid sequence encoding BCL6 -or a functional part, derivative and/or analogue thereof - and an exogenous nucleic acid sequence encoding Bc1-xL - or a functional part, derivative and/or analogue thereof. Preferably, expression of said nucleic acid sequence encoding BCL6, Bc1-xL or a functional part, derivative and/or analogue of BCL6 or Bc1-xL is regulated by an activator and/or repressor that is inducible by an exogenous compound. For instance, an inducible promoter system is used such as a Tet-on or Tet-off system.
A stable RSV-specific antibody producing cell according to the invention is preferably generated by co-expressing BCL6 and Blimp-1 in an RSV-specific antibody producing cell. An RSV-specific antibody producing cell is preferably =

obtained from an individual who has been exposed to RSV. Methods for isolating antibody producing cells are well known in the art. For instance, RSV-derived compounds that are marked with a label and/or tag are incubated with a sample of an individual who has been exposed to RSV, which sample comprises antibody producing cells. RSV-specific antibody producing cells that recognize the tagged RSV-derived compounds are isolated while unbound cells are washed away. The resulting RSV-specific antibody producing cells are subsequently stabilized by co-expressing BCL6 as well as Blimp-1.
One embodiment involves first stabilizing total antibody-producing cells from an RSV exposed donor and then isolating cells that recognize the tagged RSV-derived compound. In another embodiment antibody producing cells are equipped with a (fluorescent) marker downstream their B cell receptor (BCR, membrane expressed form of the antibody) that signals when the antibody producing cell binds an un-tagged/unlabeled antigen via the BCR. Antibody producing cells in which the marker is turn are selected and are subsequently stabilized by co-expressing BCL6 as well as Blimp-1. In another embodiment, when there are no antigen-derived compounds available but when there are assays available to screen for unique antibodies, total/bulk antibody producing cells are stabilized by co-expressing BCL6 as well as Blimp-1 and, optionally, also Bel-XL. According to this embodiment, cells are cultured at low densities, preferably between 10 and 100 cells per 96-well, in the presence of L-cells (mini bulk cultures, MBC). Culture supernatants can be used directly in screenings assays, like ELISA, Western blot or functional assays like ELISPOT, neutralization assays or cell migration assays.
In one embodiment MBC are selected and, to obtain monoclonal cell lines of the antibody producing cell of interest, limiting dilution cultures are preformed and, preferably 2-3 weeks later, supernatants of those cultures are screened again in the preferred assay.
As is well known by the skilled person, many alternative methods are available in the art. The above mentioned embodiments are non-limiting.
Further provided is therefore a method for producing an antibody producing cell, which is stable for at least three months and which is capable of producing RSV-specific antibodies or functional equivalents thereof, the method =

comprising:
- increasing an expression level of Blimp-1 in a cell which is capable of producing RSV-specific antibodies or functional equivalents thereof; and - increasing and/or maintaining a BCL6 expression level in said cell.
With a method according to the invention it has become possible to convert an RSV-specific memory B cell into a plasmablast-like cell and to stabilize said cell, so that rapid differentiation into a plasma cell does not occur.
This is contrary to natural development of plasma cells, wherein expression of Blimp-1 in a memory B cell results in rapid development into a plasma cell, thereby inhibiting BCL6 expression so that the resulting plasma cell hardly expresses BCL6. One embodiment of the present invention thus involves co-expression of both BCL6 and Blimp-1 in an RSV-specific B cell, resulting in a cell that is capable of both proliferating and producing antibody. The BCL6 expression level in said RSV-specific B-cell is preferably brought to, and maintained at, essentially the same level or at a higher level as compared to a plasmablast. This way a stable culture of RSV-specific B cells is generated, which cells remain capable of producing RSV-specific antibodies. These RSV-specific B
cells that co-express BCL6 and Blimp-1 are preferably further stabilized through the addition of the anti-apoptotic gene Bc1-xL. With the introduction of Bc1-xL it is now possible to grow plasmablasts under conditions of low cell density.
Hence, the invention also provides a method to culture plasmablasts under conditions of low cell density comprising generating an RSV-specific antibody producing cell with expression levels of BCL6, Blimp-1 and Bc1-xL with any of the herein described methods.
The amount of BCL6 expression product (preferably a BCL6 protein) in an RSV-specific antibody producing cell is regulated in a variety of ways.
In one embodiment an antibody producing cell is provided with a compound capable of directly or indirectly influencing BCL6 expression. An antibody producing cell is preferably provided with a compound capable of enhancing BCL6 expression, in order to counteract downregulation of BCL6 during expression of Blimp-1. Such compound preferably comprises a Signal Transducer of Activation and Transcription 5 (STAT5) protein or a functional part, derivative and/or analogue thereof, and/or a nucleic acid sequence coding = therefore. STAT5 is a signal transducer capable of enhancing BCL6 expression.
There are two known forms of STAT5, STAT5a and STAT5b, which are encoded by two different, tandemly linked genes. Administration and/or activation of STAT5 results in enhanced BCL6 levels. Hence, downregulation of BCL6 by Blimp-1 is at least in part compensated by upregulation expression of BCL6 by STAT5 or a functional part, derivative and/or analogue thereof. Hence, STAT5 or a functional part, derivative and/or analogue thereof is capable of directly influencing BCL6 expression. It is also possible to indirectly influence BCL6 expression. This is for instance done by regulating the amount of a compound which in turn is capable of directly or indirectly activating STAT5 and/or regulating STAT5 expression. Hence, in one embodiment the expression and/or activity of endogenous and/or exogenous STAT5 is increased. It is for instance possible to indirectly enhance BCL6 expression by culturing an antibody producing cell in the presence of interleukin (IL) 2 and/or IL 4 which are capable of activating STAT5.
In one embodiment, an RSV-specific antibody producing cell is provided with a nucleic acid sequence encoding STAT5 or a functional part, derivative and/or analogue thereof, wherein said nucleic acid sequence is constitutively active, meaning that STAT5 is continuously expressed, independent of the presence of (endogenous) regulators. In case that endogenous STAT5 expression is low, or absent, an exogenous constitutively active nucleic acid sequence encoding STAT5 or a functional part, derivative and/or analogue thereof is preferably applied resulting in a concentration of STAT5 or a functional part, derivative and/or analogue thereof which is sufficient to enhance BCL6 expression. Most preferably, an RSV-specific antibody producing cell is provided with a nucleic acid sequence encoding a compound comprising STAT5 or a functional part, derivative and/or analogue thereof, preferably a fusion protein, whose activity is regulated by an exogenous inducer of repressor, so that the extent of activation of BCL6 expression is regulated at will. Another system that allows for induction of BCL-6 is provided by a Tet-on system in which addition of tetracycline and/or derivatives of tetracycline induce activity of a transactivator that induced BCL6 gene transcripotion followed by BCL protein synthesis. In one preferred embodiment, an antibody producing cell is provided with a nucleic acid sequence encoding an estrogen receptor (ER) and STAT5 as a fusion protein ER-STAT5. This fusion protein is inactive because it forms a complex with heat shock proteins in the cytosol. This way, STAT5 is unable to reach the nucleus and BCL6 expression is not enhanced. Upon administration of the exogenous inducer 4 hydroxy--tamoxifen (4HT), the fusion protein ER-STAT5 dissociates from the heat shock proteins, so that STAT5 is capable of entering the nucleus and activating BCL6 expression.
Additionally, or alternatively, BCL6 expression in an RSV-specific antibody producing cell is enhanced by culturing said antibody producing cell in the presence of a compound capable of directly or indirectly enhancing BCL6 expression.
One embodiment therefore provides a method for producing an RSV-specific antibody producing cell comprising:
- providing an RSV-specific antibody producing cell with a compound capable of directly or indirectly enhancing BCL6 expression; and/or - culturing an RSV-specific antibody producing cell in the presence of a compound capable of directly or indirectly enhancing BCL6 expression. Said compound capable of directly or indirectly enhancing BCL6 expression preferably comprises STAT5 or a functional part, derivative and/or analogue thereof. Provided is therefore a method according to the invention comprising providing said RSV-specific antibody producing cell with STAT5 or a functional part, derivative and/or analogue thereof, or with a nucleic acid sequence encoding STAT5 or a functional part, derivative and/or analogue thereof. In one embodiment said antibody producing cell is cultured after introduction of a nucleic acid sequence encoding STAT5 or a functional part, derivative and/or analogue thereof into said cell. Said nucleic acid sequence is for instance introduced into said cell by transfection and/or virus-mediated gene transfer. Many alternative methods for introducing a nucleic acid sequence into a cell are available in the art which need no further explanation here.
With a compound capable of directly or indirectly enhancing BCL6 expression it is possible to enhance expression of endogenous BCL6. In one preferred embodiment however an antibody producing cell is provided with a nucleic acid sequence encoding BCL6 or a functional part, derivative and/or analogue thereof. As explained herein before, an exogenous nucleic acid encoding BCL6 is preferred because this allows regulation of a BCL6 concentration within a cell independently from expression of endogenous BCL6. Hence, even if expression of endogenous BCL6 is low or absent, for instance caused by Blimp-1, an exogenous nucleic acid sequence encoding BCL6 or a functional part, derivative and/or analogue thereof is still capable of producing a concentration of BCL6 which is sufficient for influencing the stability of an antibody producing cell. Also provided is therefore a method according to the invention comprising providing an RSV-specific antibody producing cell with a nucleic acid sequence encoding BCL6 or a functional part, derivative and/or analogue thereof.
Preferably, said antibody producing cell is provided with a constitutively active nucleic acid sequence encoding BCL6 or a functional part, derivative and/or analogue thereof, so that BCL6 expression is maintained even when endogenous BCL6 expression of said cell is inhibited by an endogenous repressor such as Blimp-1. Most preferably, expression of said nucleic acid sequence encoding BCL6 or a functional part, derivative and/or analogue thereof is regulated by an exogenous inducer of repressor, so that the extent of BCL6 expression is regulated at will. For instance, an inducible promoter system is used such as a Tet-on or Tet-off system, as already described.
In another preferred embodiment, the invention provides a method wherein the amount of BCL6 is indirectly regulated by providing an RSV-specific antibody producing cell with a nucleic acid sequence encoding E47 or a functional part, derivative and/or analogue thereof. E47 encodes a transcription factor that belongs to a family of helix-loop-helix proteins, named E-proteins. There are four E-proteins, E12, E47, E2-2 and HEB, which are involved in lymphocyte development. El2 and E47 are encoded by one gene, named E2A, which is spliced differently. E-proteins can be inhibited by the E protein inhibitor Id2, and Id3, and by ABF-1 (Mathas S., 2006). E proteins have been described as tumor suppressors and overexpression has been shown to induce apoptosis. One of the specific targets of E47 are the Socsl and Socs3 genes. Those Socs genes are known as negative regulators of STAT5b and thus indirectly of BCL6. In other words, expression of E47 within a B cell enhances Blimp-1 expression which results in B-cell differentiation towards an antibody producing phenotype (plasmacell).

" WO 2008/147196 The amount of Blimp-1 expression in an RSV-specific antibody producing cell is also regulated in a variety of ways. In one embodiment an RSV-specific antibody producing cell is provided with a compound capable of directly or indirectly influencing Blimp-1 expression. Additionally, or alternatively, an antibody producing cell is cultured in the presence of a compound capable of directly or indirectly influencing Blimp-1 expression. Further provided is therefore a method according to the invention comprising providing an RSV-specific antibody producing cell with a compound capable of directly or indirectly influencing Blimp-1 expression. Further provided is a method according to the invention comprising culturing said antibody producing cell in the presence of a compound capable of directly or indirectly influencing Blimp-expression. Preferably, a compound is used that is capable of enhancing Blimp-expression in order to counteract downregulation of Blimp-1 during expression of BCL6. Said compound most preferably comprises 1L-21.
In one preferred embodiment said compound capable of directly or indirectly influencing Blimp-1 expression comprises a Signal Transducer of Activation and Transcription 3 (STAT3) protein or a functional part, derivative and/or analogue thereof, and/or a nucleic acid sequence coding therefore.

is a signal transducer which is involved in B cell development and differentiation.
STAT3 is capable of upregulating Blimp-1 expression. Further provided is therefore a method according to the invention wherein said compound capable of directly or indirectly influencing Blimp-1 expression comprises STAT3 or a functional part, derivative and/or analogue thereof, or a nucleic acid sequence encoding STAT3 or a functional part, derivative and/or analogue thereof. Most preferably, expression of said nucleic acid sequence encoding STAT3 or a functional part, derivative and/or analogue thereof is regulated by an exogenous inducer of repressor, so that the extent of STAT3 expression is regulated at will.
For instance, an inducible promoter system is used such as for instance a Tet-on or Tet-off system. In one embodiment a fusion product comprising of STAT3, a derivative or analogue, and ER is introduced in said cell allowing regulation of STAT3 expression by hydroxytamoxifen.
Since STAT3 is capable of influencing Blimp-1 expression, it is also possible to indirectly regulate Blimp-1 expression by administering a compound = capable of directly or indirectly regulating the activity and/or expression of STAT3. In one embodiment an antibody producing cell is provided with a compound that is capable of enhancing the activity of STAT3, so that Blimp-1 expression is indirectly enhanced as well. Further provided is therefore a method according to the invention, wherein an antibody producing cell is provided with a compound capable of directly or indirectly enhancing activity of STAT3.
Hence, in one embodiment an antibody producing cell is provided with a compound capable of directly or indirectly activating STAT3, in order to enhance Blimp-1 expression.
STAT3 is activated in a variety of ways. Preferably, STAT3 is activated by providing an antibody producing cell with a cytokine. Cytokines, being naturally involved in B cell differentiation, are very effective in regulating STAT
proteins.
Very effective activators of STAT3 are IL-21 and IL-6, but also IL-2, IL-7, IL-10, IL-15 and IL-27 are known to activate STAT3. Moreover, Toll-like receptors (TLRs) which are involved in innate immunity are also capable of activating STAT3. One embodiment therefore provides a method of the invention, wherein said compound capable of directly or indirectly influencing Blimp-1 expression comprises IL-21, IL-2, IL-6, IL-7, IL-10, IL-15 and/or IL-27. Most preferably IL-21 is used, since IL-21 is particularly suitable for influencing the stability of an antibody producing cell. IL-21 is capable of upregulating Blimp-1 expression even when Blimp-1 expression is counteracted by BCL6.
Additionally, or alternatively a mutated Janus kinase (JAK) is used in order to activate STAT3. Naturally, a JAK is capable of phosphorylating STAT3 after it has itself been activated by at least one cytokine. A mutated Janus kinase capable of activating STAT3, independent of the presence of cytokines, is particularly suitable in a method according to the present invention.
As already explained before, a compound capable of enhancing Blimp-1 expression in one embodiment comprises a nucleic acid sequence encoding STAT3 or a functional part, derivative and/or analogue thereof. The presence of an exogenous nucleic acid sequence encoding STAT3 or a functional part, derivative and/or analogue thereof allows for a continuous presence of STAT3 or a functional part, derivative and/or analogue thereof even when expression of endogenous STAT3 is very low or absent.

=

It is also possible to decrease expression and/or activity of STAT5 in order to upregulate Blimp-1. If the amount and/or activity of STAT5 is decreased, activation of BCL6 expression is decreased as well, which results in a decreased amount of BCL6 expression product. Since BCL6 and Blimp-1 counteract each other's expression, a decreased amount of BCL6 expression product results in an increased amount of Blimp-1 expression product. Compounds capable of downregulating the activity of STAT5 are thus capable of indirectly upregulating Blimp-1. Such compounds for instance comprise members of the suppressor of cytokine signalling (SOCS) proteins. In one embodiment the amount of Blimp-1 expression product in an RSV-specific antibody producing cell is therefore upregulated by providing said cell with a SOCS protein, and/or by activating a SOCS protein within said cell.
In one preferred embodiment the expression and/or activity of STAT5 is decreased when an RSV-specific antibody-producing cell is provided with a nucleic acid sequence encoding E47 or a functional part, derivative and/or analogue thereof. Expression of E47 within B cells expressing high levels of STAT5b intervenes with differentiation and proliferation, i.e. blocking of via E47 and SOCS results in decreased BCL6 levels and subsequently in increased Blimp-1 levels. Upregulated levels of Blimp-1 result in a decreased proliferation and in a differentiation of the involved cell towards an antibody-producing cell. In other words, expression of E47 within a B cell enhances Blimp-1 expression which results in B-cell differentiation towards an antibody producing phenotype (plasma cell).
By at least a functional part of a STAT5 protein, a STAT3 protein, Bc1-xL
and/or BCL6 is meant a proteinaceous molecule that has the same capability -in kind, not necessarily in amount - of influencing the stability of an antibody producing cell as compared to a STAT5 protein, a STAT3 protein, Bc1-xL and/or BCL6, respectively. A functional part of a STAT5 protein or a STAT3 protein is for instance devoid of amino acids that are not, or only very little, involved in said capability. A derivative of a STAT5 protein, a STAT3 protein, Bc1-xL and/or BCL6 is defined as a protein which has been altered such that the capability of said protein of influencing the stability of an antibody producing cell is essentially the same in kind, not necessarily in amount. A derivative is provided in many ways, for instance through conservative amino acid substitution wherein one amino acid is substituted by another amino acid with generally similar properties (size, hydrophobicity, etc), such that the overall functioning is likely not to be seriously affected. A derivative for instance comprises a fusion protein, such as a STAT5-ER or STAT3-ER fusion protein whose activity depends on the presence of 4 hydroxy-tamoxifen (41-IT). An analogue of a STAT5 protein, a STAT3 protein, Bc1-xL and/or BCL6 is defined as a molecule having the same capability of influencing the stability of an antibody producing cell in kind, not necessarily in amount. Said analogue is not necessarily derived from said protein, STAT3 protein, Bc1-xL and/or BCL6.
In one preferred embodiment said RSV-specific antibody producing cell is cultured in the presence of IL-21 before said antibody producing cell is provided with a nucleic acid sequence encoding BCL6 or a functional part, derivative and/or analogue thereof. Culturing RSV-specific antibody producing cells, preferably B cells, in the presence of IL-21 before said cell is provided with a nucleic acid sequence encoding BCL6 or a functional part, derivative and/or analogue thereof is preferred, because in these embodiments stability, proliferation and/or antibody production is particularly well improved.
In a preferred embodiment, the invention provides a method for influencing the stability of an RSV-specific antibody producing cell as described herein, further comprising directly or indirectly increasing the amount of Bc1-xL
expression product within said antibody producing cell. This is for example accomplished by providing said antibody producing cell with a nucleic acid sequence encoding Bc1-xL or a functional part, derivative and/or analogue thereof or with nucleic acid sequences encoding other anti-apoptotic genes including but not limited to Bc1-2. In yet another embodiment this is accomplished by providing said antibody producing cell with a compound capable of directly or indirectly enhancing Bc1-xL expression, preferably said compound comprises APRIL, BAFF, CD40, BCR stimulation, cytokines, growth factors or downstream effectors like JNK and AKT (PKB).
Bc1-xL is a member of the anti-apoptotic Bc1-2 family, Bc12-proteins interact with and counteract so-called Bc1-2 homology domain 3 (BH3)-only family members such as Bax, Bak, Bim, and Bad, which induce cytochome c release following intrinsic death stimuli (Boise, L. H., 1993). Thus, protection of mitochondrial membrane integrity through proteins like Bc1-xL is critical for cell survival.
STAT5 activation has been shown to protect cells from cell death. STAT5 has been shown to regulate the expression of Bc1-xL, supporting an anti-apoptotic role for STAT5. STAT5 positively regulates the Bc1-xL expression through STAT
binding elements within the Bc1-xL promoter. In vivo, Bc1-xL expression is absent in bone marrow of STAT5A/B-doubly deficient mice. Furthermore, STAT5-mediated erythroblast survival is dependent upon upregulation of Bc1-xL.
Recently, it has been shown that transgenic overexpression of Bc1-xL in mouse B
cells promotes B cell survival and nonmalignant plasma cell foci.
A method according to the invention is particularly suitable for producing a cell culture comprising RSV-specific antibody producing cells that are capable of proliferating and secreting antibody. In one embodiment, an RSV-specific memory B cell is used in order to produce an ex vivo B cell culture. Said memory B cell is preferably human so that human antibodies are produced. Said B cell preferably originates from an individual, which individual had been previously exposed to Respiratory Syncytial Virus. In one embodiment RSV-specific B cells are isolated from a peripheral blood sample and/or a tonsil sample, using methods known in the art. Memory B cells are for instance isolated by selection (magnetic beads sorting) for the B cell marker CD19 and/or CD22 and (subsequent) selection for cell surface IgG and/or CD27 and/or by negative selection for IgM, IgD and/or IgA. In a germinal center B cell, BCL6 expression is high whereas Blimp-1 expression is low. Natural development into an antibody secreting cell involves upregulation of Blimp-1 expression. Since Blimp-1 represses BCL6 expression, upregulation of Blimp-1 results in downregulation of BCL6 in a natural situation. In a preferred embodiment of the present invention however, Blimp-1 expression is upregulated while BCL6 expression is at least in part maintained. This results in an RSV-specific antibody producing cell wherein BCL6 and Blimp-1 are co-expressed. Said RSV-specific antibody producing cell is capable of proliferating and secreting anti-RSV antibodies and is therefore suitable for use in an ex vivo B cell culture. In a further preferred embodiment, said antibody producing cell is protected by apoptosis by Bc1-xL. An RSV-specific antibody producing cell according to the present invention provides the advantage that it is stable and does not undergo terminal differentiation during a prolonged period. Said antibody producing cell according to the invention is stable for at least one week, preferably for at least one month, more preferably for at least three months, most preferably for at least six months. A B cell according to the invention is preferably cultured in the presence of CD4OL
since replication of most B cells is favoured by CD4OL.
In one embodiment BCL6 expression is maintained at essentially the same level, or at a higher level, as compared to a germinal center B cell since a significant BCL6 expression, together with Blimp-1 expression, results in an antibody producing cell with preferred proliferation and antibody production properties and/or stability. In a preferred embodiment, said BCL6 expression and/or Blimp-1 expression are accompanied by Bc1-xL expression, resulting in even more preferred proliferation and antibody production properties and/or stability.
One embodiment therefore provides a method for producing an RSV-specific antibody producing cell which is stable for at least one week, preferably for at least one month, more preferably for at least three months, more preferably for at least six months, the method comprising:
- providing an RSV-specific memory B cell;
- increasing an expression level of Blimp-1 in said cell; and - increasing and/or maintaining a BCL6 expression level in said cell. An ex vivo method for producing an RSV-specific antibody producing cell comprising increasing an expression level of Blimp-1 in an RSV-specific memory B cell and increasing and/or maintaining a BCL6 expression level in said cell is also provided. Said BCL6 and Blimp-1 expression levels are preferably brought to, and/or maintained at, essentially the same level, or at a higher level, as compared to a plasmablast. In a preferred embodiment said B cell is transduced with BCL6 and Bc1-xL. Further provided is therefore a method for producing an RSV-specific antibody producing cell which is stable for at least three months, comprising:
- providing a B cell capable of producing RSV-specific antibodies with BCL6, or a functional part, derivative and/or analogue thereof; and - providing said B cell with Bc1-xL or a functional part, derivative and/or analogue thereof; and - culturing said B cell.

= Said B cell is preferably provided with a nucleic acid sequence encoding BCL6, or a functional part, derivative and/or analogue thereof, and with a nucleic acid sequence Bc1-xL or a functional part, derivative and/or analogue thereof.

Said B cell is preferably cultured in the presence of a compound capable of enhancing Blimp-1 expression, such as for instance IL-21, IL-2, IL-6, 11-7, IL-10, IL-15, IL-27, or a mutated Janus kinase. Preferably, IL-21 is used because this cytokine is particularly suitable for enhancing Blimp-1 expression and stabilizing an antibody producing cell with a method according to the present invention.
Moreover, in order to enhance transduction efficacy, said B cell is preferably cultured in the presence of IL-21 before said B cell is transduced with a nucleic acid sequence encoding BCL6 and/or Bel-xL, or a functional part, derivative and/or analogue thereof.
In one embodiment said B cell is provided with a SOCS protein or a functional part, derivative and/or analogue thereof, or a nucleic acid coding therefore, since a SOCS protein or a functional part, derivative and/or analogue thereof is capable of indirectly enhancing Blimp-1 expression. In another alternative or additional embodiment, said B-cell is provided with E47 or a functional part, derivative and/or analogue thereof, or a nucleic acid coding therefore. As already outlined earlier, as a result of an increased level of E47 or a functional part, derivative and/or analogue thereof, Socs protein function is enhanced and Blimp-1 expression is indirectly increased.
In the Examples particularly preferred embodiments are shown.
According to one particularly preferred embodiment, RSV-specific B cells are firstly cultured in the presence of IL-21. Subsequently the B cells are subjected to a transduction reaction using a nucleic acid encoding BCL6 and a nucleic acid encoding Bel-xL. Preferably spin transduction is used. Most preferably, B
cells and virus comprising at least one nucleic acid of interest are mixed, where after the mixture is spinned in order to achieve a high transduction efficacy. After transduction, the B cells are cultured in the absence of 11-21 and in the presence of 11-4 and L-cells during 3-5 days in order to allow BCL6 expression.
Subsequently, according to this preferred embodiment, the B cells are subjected again to a transduction reaction using a nucleic acid encoding BCL6 and a nucleic acid encoding Bc1-xL. Afterwards, the B cells are again cultured in the absence of 11-21 and in the presence of 11-4 and L-cells during 3-5 days in order to allow BCL6 expression. Subsequently, cells expressing BCL6 and Bc1-xL are isolated and IL-21 is administered again to the culture in order to enhance replication and antibody production. Antibodies that are secreted by Bc1-6, Blimp1 and Bel-XL expressing cells in the culture supernatant are preferably screened for in vitro neutralizing capacity/activity/reactivity to RSV.
Antibody producing cells that produce those antibodies are preferably further selected, for instance by limiting dilution culture. Stable RSV-specific B cells are thus obtained wherein BCL6 and Blimp-1 are co-expressed. Said B cells are capable of replicating and producing antibody in an in vitro culture during at least six months.
One embodiment provides a method according to the invention further comprising selecting and/or isolating an RSV-specific antibody or a functional equivalent thereof. In one embodiment IgM producing cells and IgG producing cells are selected and/or isolated. Preferably an IgG producing cell is selected and/or isolated.
RSV-specific antibody producing cells generated with a method according to the invention are suitable for producing antibodies against RSV. In one preferred embodiment however, the genes encoding the Ig heavy and/or light chains are isolated from said cell and expressed in a second cell, such as for instance cells of a Chinese hamster ovary (CHO) cell line or 293(T) cells.
Said second cell, also called herein a producer cell, is preferably adapted to commercial antibody production. Proliferation of said producer cell results in a producer cell line capable of producing RSV-specific antibodies. Preferably, said producer cell line is suitable for producing compounds for use in humans. Hence, said producer cell line is preferably free of pathogenic agents such as pathogenic micro-organisms.
A method according to the invention is preferably used for generating an antibody producing cell that is stable for at least one week, preferably at least one month, more preferably at least three months, more preferably at least six months so that commercial antibody production has become possible. Most preferably a stable cell line capable of producing monoclonal antibodies is produced. This is preferably performed by using memory B cells that have for instance been isolated from a sample by selection for CD19 and/or CD22 (B cell marker) and cell surface IgG and/or CD27 (to mark memory cells) and/or by negative selection for IgM, IgD and/or IgA. Furthermore, an RSV-specific antibody producing cell is for instance selected in a binding assay using RSV
or a component derived from RSV, such as for instance the RSV F protein, G protein and/or SR protein. Subsequently, according to this preferred embodiment Blimp-1 and BCL6 are co-expressed in said RSV-specific antibody producing cell, resulting in a culture of cells capable of specifically binding (a component of) RSV.
In yet another preferred embodiment, said B cell is further provided with Bc1-xL
or a functional part, derivative and/or analogue thereof.
If only one memory cell is used, a cell line according to the invention which produces monoclonal antibodies is obtained. It is also possible to generate a monoclonal antibody producing cell line starting with B cells capable of producing antibodies against RSV. After a stable B cell culture has been produced with a method according to the invention, a B cell capable of producing antibodies against a specific antigen of RSV is isolated and at least a functional part of a gene encoding the Ig heavy chain and/or light chain from said B cell is preferably expressed in a second cell line. Preferably at least a functional part of the gene encoding the Ig heavy chain and at least a functional part of the gene encoding the Ig light chain from said B cell are expressed in a second cell line.
In one embodiment an antibody producing cell, preferably but not necessarily a memory B cell, that has been obtained from an individual which had been previously exposed to RSV, is used in a method according to the invention. This way, it has become possible to produce human antibodies of interest ex vivo.
Further provided is therefore a method for producing antibodies which are capable of specifically binding and/or neutralizing Respiratory Syncytial Virus, the method comprising:
- producing an antibody producing cell capable of producing RSV-specific antibodies with a method according to the invention; and - obtaining antibodies produced by said antibody producing cell.
An isolated or recombinant antibody, as well as an isolated or recombinant antibody producing cell, obtainable by a method according to the invention, or a functional equivalent thereof, is also provided. Said antibody = preferably comprises antibody D25, AM14, AM16 and/or AM23, or a functional part, derivative or analogue thereof.
Once an RSV-specific antibody producing cell according to the invention is obtained, at least a functional part of a gene encoding the Ig heavy chain and/or light chain of said cell is preferably isolated and/or generated artificially.
In one embodiment a nucleic acid sequence comprising at least a functional part of a nucleic acid sequence as depicted in Figure 11, Figure 12, Figure 14A, Figure and/or Figure 14C is provided. Said functional part preferably comprises at least one nucleic acid sequence as depicted in Figure 11D, Figure 12, Figure 14A, Figure 14B and/or Figure 14C. Said functional part preferably encodes at least one CDR as depicted in Figure 11D, Figure 12, Figure 14A, Figure 14B and/or Figure 14C.
Further provided is an isolated, synthetic or recombinant nucleic acid sequence comprising a heavy chain sequence which is at least 70%, preferably at least 80%, more preferably at least 90% homologous to at least part of the sequence CAGGTGCAGCTGGTACAGTCTGGGGCTGAAGTGAAGAAGCCTGGGTCCTCGGTGATGGTCTC
CTGCCAGGCCTCTGGAGGCCCCCTCAGAA, ACTATATTATCAAC, TGGCTACGACAGGCCCCTGGACAAGGCCCTGAGTGGATGGGA, GGGATCATTCCTGTCTTGGGTACAGTACACTACGCACCGAAGTTCCAGGGC, AGAGTCACGATTACCGCGGACGAATCCACAGACACAGCCTACATCCATCTGATCAGCCTGAG
ATCTGAGGACACGGCCATGTATTACTGTGCGACG, GAAACAGCTCTGGTTGTATCTACTACCTACCTACCACACTACTTTGACAAC, TGGGGCCAGGGAACCCTGGTCACCGTCTCCTCAG, and/or CAGGTGCAGCTGGTACAGTCTGGGGCTGAAGTGAAGAAGCCTGGGTCCTCGGTGATGGTCTC
CTGCCAGGCCTCTGGAGGCCCCCTCAGAAACTATATTATCAACTGGCTACGACAGGCCCCTG
GACAAGGCCCTGAGTGGATGGGAGGGATCATTCCTGTCTTGGGTACAGTACACTACGCACCG
AAGTTCCAGGGCAGAGTCACGATTACCGCGGACGAATCCACAGACACAGCCTACATCCATCT
GATCAGCCTGAGATCTGAGGACACGGCCATGTATTACTGTGCGACGGAAACAGCTCTGGTTG
TATCTACTACCTACCTACCACACTACTTTGACAACTGGGGCCAGGGAACCCTGGTCACCGTC
TCCTCAG, said part having at least 15 nucleotides. Said heavy chain sequence is preferably derived from antibody D25. Said heavy chain sequence preferably comprises a sequence which is at least 70%, preferably at least 80%, more preferably at least 90% homologous to a sequence as depicted in Figure 11D. An isolated, synthetic or recombinant nucleic acid sequence comprising a heavy chain sequence consisting of any of the above mentioned heavy chain sequences is also herewith provided.
An isolated, synthetic or recombinant nucleic acid sequence comprising a light chain sequence which is at least 70%, preferably at least 80%, more preferably at least 90% homologous to a least part of the sequence GACATCCAGATGACCCAGTCTCCATCCTCCCTGTCTGCAGCTGTAGGAGACAGAGTCACCAT
CACTTGC, CAGGCGAGTCAGGACATTGTCAACTATTTAAAT, TGGTATCAACAGAAACCAGGGAAAGCCCCTAAGCTCCTGATCTAC, GTTGCATCCAATTTGGAGACA, GGGGTCCCATCAAGGTTCAGTGGAAGTGGATCTGGGACAGATTTTAGTCTCACCATCAGCAG
CCTGCAGCCTGAAGATGTTGCAACATATTATTGT , CAACAATATGATAATCTCCCA, CTCACATTCGGCGGAGGGACCAAGGTTGAGATCAAAAGA and/or GACATCCAGATGACCCAGTCTCCATCCTCCCTGTCTGCAGCTGTAGGAGACAGAGTCACCAT
CACTTGCCAGGCGAGTCAGGACATTGTCAACTATTTAAATTGGTATCAACAGAAACCAGGGA
AAGCCCCTAAGCTCCTGATCTACGT TGCATCCAATTTGGAGACAGGGGTCCCATCAAGGTTC
AGTGGAAGTGGATCTGGGACAGATT TTAGTCTCACCATCAGCAGCCTGCAGCCTGAAGATGT
TGCAACATATTATTGTCAACAATATGATAATCTCCCACTCACATTCGGCGGAGGGACCAAGG
TTGAGATCAAAAGA, said part having at least 15 nucleotides, is also provided.
Said light chain sequence is preferably derived from antibody D25.
Said light chain sequence preferably comprises a sequence which is at least 70%, preferably at least 80%, more preferably at least 90% homologous to a sequence as depicted in Figure 11D. An isolated, synthetic or recombinant nucleic acid sequence comprising a heavy chain sequence consisting of any of the above mentioned light chain sequences is also herewith provided.
Further provided is an isolated, synthetic or recombinant nucleic acid sequence comprising a heavy chain sequence which is at least 70%, preferably at least 80%, more preferably at least 90% homologous to at least part of the sequence GAGGTGCAGCTGGTGGAGTCTGGGGGAGGCGTGGTCCAGCCTGGGAGGTCCCTGAGACTCTC
CTGTGCGGCCTCT, GGATTCAGCTTCAGTCACTATGCC, ATGCACTGGGTCCGCCAGGCTCCAGGCAAGGGACTGGAGTGGGTGGCAGTT, ATATCTTATGATGGAGAAAATACA, TATTACGCAGACTCCGTGAAGGGCCGATTCTCCATCTCCAGAGACAATTCCAAGAACACAGT
GTCTCTGCAAATGAACAGCCTGAGACCTGAGGACACGGCTCTATATTACTGT, GCGAGAGACCGCATAGTGGACGACTACTACTACTACGOTATGGACGTC, TGGGGCCAAGGGGCCACGGTCACCGTCTCCTCAG and/or GAGGTGCAGCTGGTGGAGTCTGGGGGACGCGTGGTCCAGCCTGGGAGGTCCCTGAGACTCTC
CTGTGCGGCCTCTGGATTCAGCTTCAGTCACTATGCCATGCACTGGGTCCGCCAGGCTCCAG
GCAAGGGACTGGAGTGGGTGGCAGTTATATCTTATGATGGAGAAAATACATATTACGCAGAC
TCCGTGAAGGGCCGATTCTCCATCTCCAGAGACAATTCCAAGAACACAGTGTCTCTGCAAAT
GAACAGCCTGAGACCTGAGGACACGGCTCTATATTACTGTGCGAGAGACCGCATAGTGGACG
ACTACTACTACTACGGTATGGACGTCTGGGGCCAAGGGGCCACGGTCACCGTCTCCTCA, said part having at least 15 nucleotides. Said heavy chain sequence is preferably derived from antibody AM14. An isolated, synthetic or recombinant nucleic acid sequence comprising a heavy chain sequence consisting of any of the above mentioned heavy chain sequences is also herewith provided.
An isolated, synthetic or recombinant nucleic acid sequence comprising a light chain sequence which is at least 70%, preferably at least 80%, more preferably at least 90% homologous to a least part of the sequence GACATCCAGATGACCCAGTCTCCATCTTCCCTGTCTGCATCTGTAGGAGACAGAGTCACCAT
CACTTGCCAGGCGAGT, CAGGACATTAAGAAGTAT, TTAAATTGGTATCATCAGAAACCAGGGAAAGTCCCTGAGCTCCTGATGCAC, GATGCATCC, AATTTGGAAACAGGGGTCCCATCAAGGTTCAGTGGCAGGGGATCTGGGACAGATTTTACTCT
CACCATTAGCAGCCTGCAGCCTGAAGATATTGGAACATATTACTGT, CAACAGTATGATAATCTGCCTCCGCTCACT, TTCGGCGGAGGGACCAAGGTGGAGATCAAAC and/or GACATCCAGATGACCCAGTCTCCATCTTCCCTGTCTGCATCTGTAGGAGACAGAGTCACCAT
CACTTGCCAGGCGAGTCAGGACATTAAGAAGTATTTAAATTGGTATCATCAGAAACCAGGGA
AAGTCCCTGAGCTCCTGATGCACGATGCATCCAATTTGGAAACAGGGGTCCCATCAAGGTTC
AGTGGCAGGGGATCTGGGACAGATTTTACTCTCACCATTAGCAGCCTGCAGCCTGAAGATAT
TGGAACATATTACTGTCAACAGTATGATAATCTGCCTCCGCTCACTTTCGGCGGAGGGACCA
AGGTGGAGATCAAACGAACTGTG, said part having at least 15 nucleotides, is also provided. Said light chain sequence is preferably derived from antibody AM14.
An isolated, synthetic or recombinant nucleic acid sequence comprising a heavy chain sequence consisting of any of the above mentioned light chain sequences is also herewith provided.
Further provided is an isolated, synthetic or recombinant nucleic acid sequence comprising a heavy chain sequence which is at least 70%, preferably at least 80%, more preferably at least 90% homologous to at least part of the sequence GAGGTGCAGCTGGTGGAGACCGGGGGAGGCCIGGCCCAGCCTGGGGGGTCCCTGAGACTCTC
CTGTGCAGCCTCT, GGATTCACATTCAGTAGTTATAAC, ATGAACTGGGTCCGCCAGGCTCCAGGGAAGGGGCTGGAGTGGGTCTCACAC, ATTAGTGCGGGTAGTAGTTACATA, TACTACTCAGACTCAGTGAAGGGCCGATTCACCGTCTCCAGAGACAACGTCAGGAACTCAGT
ATATCTGCAAATGAACAGCCTGAGAGCCGCTGACACGGCTGTGTATTACTGT, GCGAGAGAGGATTATGGTCCGGGAAATTATTATAGTCCTAACTCGTTCGACCCC, TGGGGCCAGGGAACCCTGGTCACCGTCTCCTCAG and/or GAGGTGCAGCTGGTGGAGACCGGGGGAGGCCTGGCCCAGCCTGGGGGGTCCCTGAGACTCTC
CTGTGCAGCCTCTGGATTCACATTCAGTAGTTATAACATGAACTGGGTCCGCCAGGCTCCAG
GGAAGGGGCTGGAGTGGGTCTCACACATTAGTGCGGGTAGTAGTTACATATACTACTCAGAC
TCAGTGAAGGGCCGATTCACCGTCTCCAGAGACAACGTCAGGAACTCAGTATATCTGCAAAT
GAACAGCCTGAGAGCCGCTGACACGGCTGTGTATTACTGTGCGAGAGAGGATTATGGTCCGG
GAAATTATTATAGTCCTAACTGGTTCGACCCCTGGGGCCAGGGAACCCTGGTCACCGTCTCC
TCA, said part having at least 15 nucleotides. Said heavy chain sequence is preferably derived from antibody AM16. An isolated, synthetic or recombinant nucleic acid sequence comprising a heavy chain sequence consisting of any of the above mentioned heavy chain sequences is also herewith provided.
An isolated, synthetic or recombinant nucleic acid sequence comprising a light chain sequence which is at least 70%, preferably at least 80%, more preferably at least 90% homologous to a least part of the sequence CAGTCTGTCGTGACGCAGCCGCCCTCAGTGTCTGGGGCCCCAGGGCAGAGAGTCACCATCTC
CTGCACTGGGAGC, AGCTCCAACATCGGGGCAGGTTATGAT, GTACACTGGTACCAGCAGCTTCCAGGAACAGCCCCCAAACTCCTCATCTAT, GGCAACACT, AATCGGCCCTCAGGGGTCTCCGACCGATTCTCTGGCTCCAAGTCTGGCACCTCAGCCTCCCT
GGCCATCACTGGACTCCAGGCTGAGGATGAGGCTGATTATTACTGC, CACTCCTATGACAGAAGCCTGAGTGGT, TCAGTATTCGGCGGAGGGACCAAGCTGACCGTCCTAG and/or CAGTCTGTCGTGACGCAGCCGCCCTCAGTGTCTGGGGCCCCAGGGCAGAGAGTCACCATCTC
CTGCACTGGGAGCAGCTCCAACATCGGGGCAGGTTATGATGTACACTGGTACCAGCAGCTTC
CAGGAACAGCCCCCAAACTCCTCATCTATGGCAACACTAATCGGCCCTCAGGGGTCTCCGAC
CGATTCTCTGGCTCCAAGTCTGGCACCTCAGCCTCCCTGGCCATCACTGGACTCCAGGCTGA
GGATGAGGCTGATTATTACTGCCACTCCTATGACAGAAGCCTGAGTGGTTCAGTATTCGGCG
GAGGGACCAAGCTGACCGTC, said part having at least 15 nucleotides, is also = provided. Said light chain sequence is preferably derived from antibody AM16.
An isolated, synthetic or recombinant nucleic acid sequence comprising a heavy chain sequence consisting of any of the above mentioned light chain sequences is also herewith provided.
Further provided is an isolated, synthetic or recombinant nucleic acid sequence comprising a heavy chain sequence which is at least 70%, preferably at least 80%, more preferably at least 90% homologous to at least part of the sequence CAGGTGCAACTGGTGGAGTCTGGGGGAAATGTGGTCAAGCCTGGGACGTCCCTGAGACTGTC
CTGTGCAGCGACT, GGATTCAACTTCCATAACTACGGC, ATGAACTGGGTCCGCCAGGCTCCAGGCAAGGGGCTGGAGTGGGTGGCGGTT, GTTTGGTATGATGGAAGTAAGAAA, TACTATGCAGACTCCGTGACGGGCCGATTCGCCATCTCCAGAGACAATTCCAAGAACACTCT
GTATCTGCAAATGAACAGCCTGAGAGTCGAGGACACGGCTGTTTATTATTGT, GTGAGAGATAAAGTGGGACCGACTCCCTACTTTGACTCC, TGGGGCCAGGGAACCCTGGTCACCGTATCCTCAG and/or GAGGTGCAGCTGGTGGAGTCTGGGGGAAATGTGGTCAAGCCTGGGACGTCCCTGAGACTGTC
CTGTGCAGCGACTGGATTCAACTTCCATAACTACGGCATGAACTGGGTCCGCCAGGCTCCAG
GCAAGGGGCTGGAGTGGGTGGCGGTTGTTTGGTATGATGGAAGTAAGAAATACTATGCAGAC
TCCGTGACGGGCCGATTCGCCATCTCCAGAGACAATTCCAAGAACACTCTGTATCTGCAAAT
GAACAGCCTGAGAGTCGAGGACACGGCTGTTTATTATTGTGTGAGAGATAAAGTGGGACCGA
CTCCCTACTTTGACTCCTGGGGCCAGGGAACCCTGGTCACCGTCTCGAGT,saidpart having at least 15 nucleotides. Said heavy chain sequence is preferably derived from antibody AM23. An isolated, synthetic or recombinant nucleic acid sequence comprising a heavy chain sequence consisting of any of the above mentioned heavy chain sequences is also herewith provided.
An isolated, synthetic or recombinant nucleic acid sequence comprising a light chain sequence which is at least 70%, preferably at least 80%, more preferably at least 90% homologous to a least part of the sequence TCCTATGTGCTGACTCAGCCACCCTCGGTGTCACTGGCCCCAGGAGGGACGGCCGCGATCAC
CTGTGGAAGAAAC, AACATTGGAAGTGAAACT, GTGCACTGGTACCAGCAGAAGCCAGGCCAGGCCCCTGTGCTGGTCGTCTAT, GATGATGAC, GACCGGCCCTCAGGGATCCCTGAGCGATTCTCTGGCTCCAACTCTGGGAACACGGCCACCCT
GACCATCAGCAGGGTCGAGGCCGGGGATGAGGCCGACTATTACTGT, = CAGGTGTGGGATAGGAGTAATTATCATCAGGTA, TTCGGCGGAGGGACCAAGTTGACCGTCCTAG and/or TCCTATGTGCTGACTCAGCCCCCCTCGGTGTCACTGGCCCCAGGAGGGACGGCCGCGATCAC
CTGTGGAAGAAACAACATTGGAAGTGAAACTGTGCACTGGTACCAGCAGAAGCCAGGCCAGG
CCCCTGTGCTGGTCGTCTATGATGATGACGACCGGCCCTCAGGGATCCCTGAGCGATTCTCT
GGCTCCAACTCTGGGAACACGGCCACCCTGACCATCAGCAGGGTCGAGGCCGGGGATGAGGC
CGACTATTACTGTCAGGTGTGGGATAGGAGTAATTATCATCAGGTATTCGGCGGAGGGACCA
AGCTGACCGTC, said part having at least 15 nucleotides, is also provided. Said light chain sequence is preferably derived from antibody AM23. An isolated, synthetic or recombinant nucleic acid sequence comprising a heavy chain sequence consisting of any of the above mentioned heavy chain sequences is also herewith provided.
A nucleic acid sequence encoding an amino acid sequence which is at least 70%, preferably at least 80%, more preferably at least 90% identical to at least a functional part of an amino acid sequence as depicted in Figure 11, Figure 14A, Figure 14B and/or Figure 14C, said part having at least 5 amino acid residues is also provided. Said nucleic acid sequence preferably encodes an amino acid sequence which is at least 80% identical to heavy chain CDR sequence 1, 2 and/or 3 and/or light chain CDR sequence 1 or 2 depicted in Figure 11D. In another preferred embodiment said nucleic acid sequence encodes an amino acid sequence which is at least 80% identical to at least one of the CDR sequences depicted in Figure 14A, in Figure 14B and/or in Figure 14C. In one preferred embodiment said nucleic acid sequence encodes an amino acid sequence which is at least 70%
identical to a heavy chain sequence depicted in Figure 11A, to a heavy chain sequence depicted in Figure 14A, to a heavy chain sequence depicted in Figure 11B, to a heavy chain sequence depicted in Figure 14C, to a light chain sequence depicted in Figure 11A, to a light chain sequence depicted in Figure 14A, to a light chain sequence depicted in Figure 14B, and/or to a light chain sequence depicted in Figure 14C.
Further provided is therefore an isolated, synthetic or recombinant nucleic acid sequence comprising a sequence encoding an amino acid sequence which is at least 70%, preferably at least 80%, more preferably at least 85% identical to an amino acid sequence as depicted in Figure 11A-D. Said nucleic acid sequence preferably encodes an amino acid sequence which is at least 80% identical to heavy chain CDR sequence 1, 2 and/or 3 and/or light chain CDR sequence 1 or 2 as depicted in Figure 11A-D. One embodiment provides an isolated, synthetic or recombinant nucleic acid sequence comprising a sequence encoding an amino acid sequence which is at least 70% identical to the amino acid sequence NYIIN, and/or at least 75% identical to the sequence GHPVLGTVHYAPKFQG, and/or at least 70% identical to the sequence ETALVVSTTYLPHYFDN, and/or at least 85% identical to the sequence QASQDIVNYLN, and/or at least 70% identical to the sequence VASNLET, and/or at least 70% identical to the sequence QVQLVQSGAEVKKPGSSVMVSCQASGGPLRNYIINWLRQAPGQGPEWMGGII
PVLGTVHYAPKFQGRVTITADESTDTAYIHLISLRSEDTAMYYCATETALVVST
TYLPHYFDN WGQGTLVTVSS, and/or at least 70% identical to the sequence DIQMTQSPSSLSAAVGDRVTITCQASQDIVNYLNWYQQKPGKAPKLLIYVASN
LETGVPSRFSGSGSGTDFSLTISSLQPEDVATYYCQQYDNLPLTFGGGTKVEIK
RTV.
A nucleic acid sequence according to the invention is preferably at least 80%, more preferably at least 85%, more preferably at least 90%, most preferably at least 95% homologous to any of the above recited sequences.
Further provided is an isolated, synthetic or recombinant nucleic acid sequence comprising a sequence encoding an amino acid sequence which is at least 70%, preferably at least 80%, more preferably at least 85% identical to an amino acid sequence as depicted in Figure 14A-C. Said nucleic acid sequence preferably encodes an amino acid sequence which is at least 70% identical to a CDR sequence as depicted in Figure 14A, 14B and/or 14C. One embodiment provides an isolated, synthetic or recombinant nucleic acid sequence comprising a sequence encoding an amino acid sequence which is at least 70% identical to an amino acid sequence selected from the group consisting of: GFSFSHYA, ISYDGENT, ARDRIVDDYYYYGMDV, QDIKKY, DAS, QQYDNLPPLT, EVQLVESGGGVVQPGRSLRLSCAASGFSFSHYAMHWVRQAPGKGLEWVAVIS
YDGENTYYADSVKGRFSISRDNSKNTVSLQMNSLRPEDTALYYCARDRIVDD
YYYYGMDVWGQGATVTVSS, DIQMTQS PS SLSASVGDRVT I TCQASQDIKKYLNWYHQKPGKVPELLMHDASNLE TGVPSRF
SGRGSGT DFTLT I SSLQPEDI GT YYCQQY DNLP PLTFGGGTKVEI KRTV , GFTFSSYN, ISAGSSYI, AREDYGPGNYYSPNWFDP, SSNIGAGYD, GNT, HSYDRSLSG, EVQLVETGGGLAQPGGSLRLSCAASGFT FSSYNMNWVRQAPGKGLEWVSHI SAGS SYIYYS D

= SVKGRFTVSRDNVRNSVYLQMNS LRAADTAVYYCARE DYGPGNYYS PNWFD PWGQGTLVTVS
, QSVVTQPPSVSGAPGQRVT I SCT GSSSNIGAGYDVHWYQQL PGTAPKLL I YGNTNRPSGVS D
RFSGSKSGTSASLAI TGLQAEDEADYYCHSYDRSLSGSVFGGGTKLTV, GFNFHNYG, VWYDGSKK, VRDKVGPTPYFDS, NIGSET, DDD, QVWDRSNYHQV, EVQLVESGGNVVKPGTSLRLSCAATGENFHNYGMNWVRQAPGKGLEWVAVVWYDGSKKYYAD
SVTGRFAI S RDNSKNTLYLQMNSLRVE DTAVYYCVRDKVGPT PY FDSWGQGT LVTVSS and SYVLTQPPSVSLAPGGTAAITCGRNNI GSETVHWYQQKPGQAPVLVVYDDDDRPSGI PERFS
GSNSGNTAT LT I SRVEAGDEADYYCQVWERSNYHQVFGGGTKLTV
A nucleic acid sequence according to the invention is preferably at least 80%, more preferably at least 85%, more preferably at least 90%, most preferably at least 95% homologous to any of the above recited sequences.
As already explained herein before, nucleic acid sequences according to the present invention are particularly suitable for expressing an antibody or a functional part, derivative or analogue thereof according to the invention, preferably D25, AM14, AM16, AM23 or a functional part, derivative and/or analogue thereof, in a nucleic acid expression system. A nucleic acid sequence according to the present invention is preferably expressed in a cell, more preferably in a producer cell adapted for antibody production.
The invention is further explained in the following examples. These examples do not limit the scope of the invention, but merely serve to clarify the invention.

=

= Examples MATERIALS AND METHODS
Maintenance and isolation of human B cells Using standard procedures, CD19 positive human B cells were isolated from bloodbank derived huffy coat (other sources can be fresh blood with an anti-coagulation factor, or a lymphoid organ for example tonsil or spleen). In brief, total peripheral blood mononuclear cells (PBMC) were isolated using ficoll density separation (Amersham, Buckinghamshire, UK). CD22 labeled beads were used to positively selected B cells by MACS cell sorting technique as described by the manufacturer (Miltenyi, Utrecht, Netherlands). Cells were subsequently stained with appropriate combinations of monoclonal antibodies (mAbs) to CD19, CD27, IgD, IgM and IgA (Becton Dickinson (BD), Franklin Lakes, NJ, USA).
Memory B cells that are positive for CD19 and CD27 and negative for IgM, IgA
and IgD were then sorted using the FACSAria (BD) (Figure 1). Besides memory B cells, other B cells subsets, like naive, nalve, follicular, memory, antibody producing, centroblast, centrocyte, germinal center, plasma blast, plasma cell, marginal zone, perisinusoidal or transitional B cells (many of those subsets have only been determined in mice) can be isolated using appropriate markers.
Cell Culture Sorted cells were washed and cultured in 24 well plates (1.5 to 2x105cells/m1) on 80 Gray, irradiated CD4OL-expressing L-cells (5x104 cells/ml; provided by DR.
J.
Banchereau, Schering Plough France, Dardilly France), in complete medium (Iscove's Modified D Minimal Essential Medium containing 8% fetal calf serum (FCS) and Penicillin/Streptomycin). Unless mentioned otherwise, these CD4OL-expressing L-cells are always present in the cultures in combination with 8%
FCS. To prepare the B cell for retroviral transduction cells were cultured for hours in the presence of mouse IL-21 (50 ng/ml, R&D, Minneapolis, MN, USA).
After transduction cells are preferentially cultured in the presence of IL-21, however cells do respond to IL-4, IL-15 and IL-10 (not excluding other cytokines).
For example, IL-4 induced B cell expansion is lower compared to IL-21 and lower levels of cell division can be required in some experiments.

Retroviral constructs and production of recombinant retrovirus Constitutive active mutants of STAT5a and b have been described previously.
DNAs encoding these mutants and wildtype STAT5b were obtained from T.
Kitamura (IMSUT, Tokyo, Japan). Bc1-6 was identified in a senescence rescue screen in murine fibroblasts as an inhibitor of anti-proliferative p19ARF-p53 signaling. Bcl-XL was identified as an anti-apoptose factor, which was kindly provided by Dr Korsmeyer (Howard Hughes Medical Institute, Boston, US).
These DNAs were ligated into LZRS-linker-IRES-GFP (or IRES-YFP or IRES-NGFR) vector that was described previously (Heemskerk et al., 1997; Heemskerk et al., 1999). Instead of the IRES-GFP (Green Fluorescent Protein) marker also an IRES-YFP (Yellow Fluorescent Protein) or an IRES-NGFR (Nerve Growth Factor Receptor) was used. NGFR is a signaling-incompetent mutant of the NGFR, kindly provided by Dr. C. Bonini. A monoclonal antibody against NGFR
(Chromaprobe, Mountain View, CA, US or Miltenyi) was used to visualize NGFR-expressing cells.
For production of recombinant retrovirus, the retroviral plasmids were transfected into a helper-virus free amphotropic producer cell line Phoenix-A, a derivative of the human embryonic kidney cell line 293 (Kinsella and Nolan, 1996) (a kind gift of Dr. G. Nolan, Stanford University, Palo Alto, CA), using Fugene-6 (Roche Diagnostics Netherlands, Almere, Netherlands) according to manufacturers protocols. Two days later selection of transfected cells started by the addition of 2 j.tg/m1puromycin (Becton Dickinson Clontech Laboratories, Palo Alto, CA). Ten to 14 days after transfection 6 x 106 cells were plated per 10 cm petridish (Becton Dickinson Discovery Labware, Bedford, MA) in 10 ml complete medium without puromycin. The next day the medium was refreshed and on the following day retroviral supernatant was harvested, centrifuged and frozen in cell free aliquots at -70 C. This approach affords a reproducible rapid, large scale and high titer retroviral production of over 3 x 106 infectious virus particles/ml.
Retroviral Transduction The recombinant human fibronectin fragments CH-296 transduction procedure (RetroNectinTM; Takara, Otsu, Japan) was performed as described previously (Heemskerk et al., 1997; Heemskerk et al., 1999). Non-tissue culture-treated wells plates (Costar, Badhoevedorp, Netherlands) were coated with 0.3 ml of 30 =

1.1g/m1 recombinant human fibronectin fragment CH-296 at room temperature for 2 hours or overnight at 4 C. When different sized non-tissue culture plates were used, reagents were used proportionately. The CH-296 solution was removed, followed by incubation with 2% human serum albumin (HSA) in phoshate buffered saline (PBS) for 30 min at room temperature, followed by washing once with PBS. 5x105B cells, which were prepared for retroviral transduction were plated in 0.25 ml KEN/II without FCS and L-cells and mixed with 0.25 ml of thawed retroviral supernatant. For the Bc1-6 Bcl-XL double transduction 125 1 of Bc1-6-IRES-NGFR (or IRES-YFP) (Shvarts A. et al. Genes Dev., 2002) and 125 ill of Bcl-XL-IRES-GFP (provided by S. Korsmeyer, Howard Hughes Medical Institute, Childrens Hospital, Boston, USA) were mixed and added to the cells.

The culture was subsequently centrifuged at 1800 rpm at 25 C for 60 minutes and incubated for 6 hours at 37 C. Next 0.25 ml of supernatant was removed and 0.25 ml of fresh retroviral supernatant was added. The culture was again centrifuged at 1800 rpm at 25 C for 60 minutes and incubated at 37 C
overnight.
The next morning cells were transferred to 24 wells tissue culture treated plate (Costar) and cultured for 3-5 days under normal conditions in the presence of human IL-4 (50 ng/ml) or mouse IL-21 (50 ng/ml, R&D, Minneapolis, MN, USA).
Transduction efficiency was determined by antibody staining of a truncated, signaling incompetent mutant of Nerve Growth Factor Receptor (ANGFR, provided by C. Bonini, St. Raphael Hospital, Milan, Italy) or (co)expression of GFP and or YFP. The cells containing the transgene(s) of interest are then selected for further experiments.
Flowcytometry Antibodies against the human molecules IgD, IgG, CD3, CD19, CD20, CD27, CD38, CD40, CD45, CD56, CD 70, CD80, CD 86, 1-ILA-DR (BD) directly labeled with FITC, PE, PERCY, PE-Cy5, APC or APC-Cy7 and IgM, kappa light chain, lambda light chain, CD138, directly labeled with PE (DAKO) were used for flowcytometry analysis. Stained cells were analyzed using a LSRII (BD) and FACS data was processed with FlowJo computer software (Tree Star, Inc).
Proliferation experiment Naive and memory B cells were isolated from fresh PBMC on the FACSAria:
Naïve B cells: CD19-Pe-Cy7 pos, CD27-APC neg, IgD-PE pos Memory B cells: CD19-Pe-Cy7 pos, CD27-APC pos, IgD-PE neg, IgA-FITC neg Cells were washed in PBS and resuspended in 0.5 ml RPMI (37 C) without FCS.
An equal amount of IMDM containing 2 M Carboxyfluorescein succinimidyl ester (CFSE) was added to the cell mixture and incubated for 7 min at 37 C. Up labeling of the cells was stopped by washing the cell with cold FCS. Cells were resuspended in 500111 IMDM-8% FCS and cultured with L-cells and in the absence or presence of IL-21. Non-labeled cells were used as control.
After 36 hrs (immediately before transduction) a proportion of cells was analyzed for their CFSE content. Remaining cells were spin transduced with Bc1-6-IRES-NGFR, cultured for 3 days, and analyzed for their CFSE content using the LSRII.
Data was analyzed using FlowJo software (Treestar) Isolation of antigen specific human B cells using high speed single cell sorting In addition to the memory B cell isolation method described above starting with MBC (i.e. 100 cell/well cultures), human memory B cells can also be incubated with a fluorescent labeled antigen and sorted based on antigen recognition. An example is the isolation of B cells that bind phycoerythrin (PE) labeled Tetanus Toxoid (provided by A. Radbruch, Berlin, Germany) (Figure 4). Cells were cultured at 1 cell/well and checked for TT binding. Notwithstanding that any other labeled antigen can be used.
Determining the B cell receptor (BCR) expression alter long term culture of Bc1-6 and Bcl-XL transduced cells It is known that B cells that differentiate during in vitro culture lose their BCR
membrane expression, which is also observed in EBV transformed B cells.
Therefore B cells transduced with Bc1-6 and Bcl-XL and cultured in the presence of IL-21 were stained for GFP, NGFR, CD19, Kappa and /or Lambda or IgG or with labeled Tetanus Toxoid. To show the usefulness of the BCR expression we sorted TT-PE (Radbruch) binding cells using the FACSAria (BD) at 1 cell/well in 96-well plates, which were seeded with L-cells and IL-21 containing culture medium. After three weeks Tetanus Toxoid binding of outgrowing clones was checked using the FACS Canto (BD). Therefore cells were harvested and stained in 96-well plates with GFP, NGFR, CD19 and TT-PE.

Development of Bel-6 and Bc1-XL double positive B cell lines that secrete antibodies B cell lines were created that produces monoclonal antibodies and are 100% Bc1-and Bel-XL double positive. First this was achieved by inducing proliferation and differentiation using IL-21. Meanwhile these cells are transduced with the Bc1-IRES-NGFR and Bcl-XL-IRES-GFP retroviruses. The cells are maintained on IL-4 for 3-4 days. The cells that are transduced with either one or both retroviruses then express the transgene and will therefore express the NGFR or GFP protein.

The expression of NGFR and/or GFP can be visualized by using the LSRII (BD).
If necessary, cells can be transduced again to obtain higher numbers of cells expressing both transgenes. Irrespective of a second transduction the cells that express both transgenes are sorted using the FACS Aria (BD) and cultured at a cell density ranging from 10-500 cells/well in 96-well plates in the presence of IL-21 and 2500 to 5000 L-cells/well. These mini-bulk-cultures (MBC) secrete relatively large amounts of antibody in the culture supernatant already at day
5 which then can be used for screening purposes. Screening can be based on techniques available for the antigen of interest e.g. ELISA/EIA/RIA, Western blot or direct functional assays like neutralization of cytokine blocking experiments.
After screening and selection of MBC that recognize the antigen of interest (TT
and RSV in our experiments), cells are subcloned at 0.5 ¨ 1 cell/well in 96 well in the presence of IL-21. Subcloning normally takes 2-3 weeks and can be performed by limiting dilution (LD) cultures or single cell sorting using flow cytometry (FACSAria).
RSV A-2 virus stock and HEp2 cell line The RSV A-2 virus (kindly provided by G. van Bleek, WKZ, Utrecht) and HEp2 cell line (Clinical Laboratory, AMC, Amsterdam), were cultured in large quantities and frozen in liquid nitrogen.
The adherent HEp2 cell line was cultured in normal medium in T175 Falcon bottles before aliquots were frozen.
To obtain a high titer RSV stock, HEp2 cells were seeded and cultured to reach 50-60% confluence. The original RSV stock was added (1/20 dilution total volume 5 ml) for 45' at RT on the HEp2 cells. 15 ml fresh medium was added and cells were left o/n at 37 C, 5% CO2 with the coverlid open. The next morning culture supernatant was carefully removed and 15 ml medium containing 1% FCS was
6 added. Cells were left for 24 to 36 hours at 37 C, 5% CO2 with the coverlid closed.
When RSV induced syncytia were clearly visible and the majority of the syncytia were still intact, the medium was harvested, filtered (0.22 [um) and spin at rpm at RT before samples were snap frozen and stored in liquid nitrogen. A
second harvest can be obtained by immediately adding new medium containing 1% FCS and freezing this batch 4-6 hours later.
RSV lysate for ELISA
HEp2 cells that were infected with RSV A-2 to obtain virus stocks were used to isolate RSV proteins. First cell were carefully washed with PBS and trypsinized.
Trypsin (Gibco) was washed away and the cell pellet was lysed with 1%
octylglucoside (cell pellet of one T175 flask was treated with 2 ml octylglucoside).
Suspension was homogenized with syringe and needle (10 times up and down), incubated for 1 hour on ice and then dialyzed against 2L TBS buffer pH 7.4, o/n at 4 C. Supernatant was obtained after spin down of cell debris. The protein content was determined at 3.6 mg/ml and was used at 20 pg/m1 (50 1) in ELISAs.
Determining TCID50 and PFU of RSV stocks To determine the TCID50, 101 HEp2 were seeded in 96 well plates and infected with a 2 or 10 step serial dilution of RSV virus in 4-plo. 2-3 days later culture supernatant were removed and cells were fixed with 80% acetone for 10' at RT.
After removal of the acetone, the fixed cell layer was dried and kept at 4 C
or frozen at -20 C. To stain RSV HEp2 cells the plates were first blocked with 5%

milkpower in PBS 0.1% Tween 20. Then plates were washed 3 times before being incubated for 3-5 hours at 37 C with polyclonal goat anti-RSV-HRP (1:500, Biodesign, Saco, ME, US) and washed extensively. Next the wells were incubated with AEC substrate for 30' at RT. Infected foci stain red and can be observed by eye using a light microscope and can be counted. Standard Excel software was used to determine the TCID5o.
To determine the amount of plaque forming units (PFU) of the virus, 1x105/m1 of HEp2 cells in 24 well plates were incubated with 10-fold serial dilutions (10-3 ¨
10-7) of RSV virus stock in medium with 1% FCS at 37 C for 45' (200 I.L1) before cells and virus were covered with 0.5 ml hand warm 0.25% seaplaque agar (Biozyme). The agarose layer prevents the spreading of the virus to uninfected cells through the culture medium. Thereby the virus can infect only neighboring cells, which eventually are killed by the virus creating plaques in the monolayer of HEp2 cells. Those plaques can best be visualized by staining the fixed cells (96% ethanol ¨ 100% acetic acid ¨ 10% formalin 6:2:1) with 1% crystal violet solution. Plaques are counted (by at least two different individuals) and the PFU
value can be determined.
Selection of Respiratory Syncytial Virus (RSV) neutralizing antibodies To obtain anti-respiratory syncytial virus (RSV) B cell clones, peripheral blood cells (PBMC) from two donors were isolated from bloodbank derived huffy coats (donor B62 and B63). Before sorting CD19P sIgM"egIgD1 gIgA"egCD27P0- cells using the FACSAria (BD)(Figure 1), CD22+ cells were isolated using MACS beads and columns (Miltenyi). Only if mentioned differently, cells were cultured with L-cells.
Cells were cultured for 36 hours in the presence IL-21 before being transduced with Bc1-6-IRES-NGFR only. After 12h cells were harvested and cultured for 3 days in the presence of IL-4 before NGFR expressing cells were sorted using MACS beads (Miltenyi) and immediately transduced with Bc1-XL-IRES-GFP. The B cells that did not bind to the MACS beads were washed and transduced with Bc1-6 and Bel-XL at the same time. After 12h cells were harvested, pooled and cultured for 3 days in the presence of IL-4 before being sorted on GFP and NGFR
expression on the FACSAria. Cells were washed and cultured at 100 cell/well density in 96 well plates (Costar) in the presence of IL-21.
The double transduced Bc1-6 and Bel-XL B cell cultures were screened for RSV
binding using a RSV-infected HEp2 cell lysate ELISA and were tested in parallel using a RSV microneutralization experiment. In brief, 10 HEp2 cells are seeded in flat bottom 96 well plates (Costar) in complete medium. The next day medium is replaced for lh at RT with the mixture of RSV virus and cell culture supernatant which have been pre-incubated for 30 min at 37 C. The total volume is 25 )11 and the RSV end concentration is 0.1 MOI. After 1h the virus supernatant mixture is 9 times diluted with PBS and replaced with 100 1 IMDM/5%FCS. After 2 days cells are fixed with 80% acetone and stained with polyclonal anti-RSV-HRP (Biodesign). Using H202 and AEC cells infected with RSV develop a red stain. Using light microscopy infected cells can be observed and counted if necessary. As a control for RSV neutralization a goat polyclonal anti-RSV (Abeam, Cambridge, MA) is used.

RT-PCR and cloning of VH and VL regions Total RNA was isolated from ¨5x105 B cells with the KNeasy mini kit (Qiagen, Venlo, The Netherlands). 250 ng of total RNA was reverse transcribed in a volume of 20 pi containing 1X first strand buffer, 500 !..LIVI dNTP, 250 ng random hexamers, 5 mM DTT, 40 U RNasin (Promega) and 200 U SuperScript III RT
(Invitrogen). The cDNA was diluted 10X in Ultrapure water and 2.5 I of cDNA
was subjected to PCR in a 50 pl solution containing 20 mM Tris-HCL, 50 mM
KCL, 2.5 mM MgC12, 250 M dNTP, 1 U AmpliTaq Cold DNA poly-merase (Applied Biosystems Inc.), and 25 pmol of each primer. PCR conditions were as follows: 8 min denaturing step at 96 C followed by 35 cycles of 30 sec at 96 C, 30 sec at 60 C, 1 min at 72 C, and a final 10 min extension at 72 C.
PCR products were run on agarose gels, purified and cloned into the pCR2.1 TA
cloning vector according to manufacturers' recommendations. Sequence analysis was performed using Bignye Terminator chemistry (Applied Biosystems Inc.) and Vector-NTI software (Invitrogen).
To rule out reverse transcriptase andJor DNA polymerase induced mutations, several independent cDNA conversions and PCR reactions were performed and individually cloned and sequence analyzed. Consensus sequences were determined with Vector-NTI Contig Express software.
For recombinant protein antibody expression in 293T cells full length heavy and light chain constructs were generated in pCDNA3.1(+)Zeo (Invitrogen). The heavy chain expression vector was constructed by PCR amplification of the heavy chain leader sequence and VH region of clone D25 introducing a 5'-NheI site and a 3'-XhoI site. The IgG1 constant region (CH1-hinge-CH2-CH3) was amplified from the same cDNA while introducing a 5'-XhoI and a 3'-NotI site. The full length heavy chain expression vector was obtained by three point ligation into NheI/NotI digested pCDNA3.1(+)Zeo. The full length light chain expression construct was generated by PCR amplification of the light chain leader sequence, VL region and light chain constant region with primers introducing a 5'-NheI
and 3'-NotI site. The latter product was cloned into NheI/NotI digested pCDNA3.1(+)Zeo to obtain a full length light chain expression vector.
Sequence analysis was performed to confirm correctness of the expression constructs.

Transient double transfection (Fugene-6, Roche, Germany or Lipofectamine LTX, Invitrogen) of 293T cells with both heavy and light chain expression vectors was performed to produce recombinant monoclonal antibody. A FACS staining with the resulting culture supernatant (48 hours) on RSV infected Hep2 cells was performed to show functional binding of the antibody to the RSV F-protein.
The oligonucleotides used for PCR amplifications were:
VH regions:
VH1-For 5'-AAATCGATACCACCATGGACTGGACCTGGAGG-3' VH1B-For 5'-AAATCGATACCACCATGGACTGGACCTGGAGM-3' VH2A-For 5'-AAATCGATACCACCATGGACACACTTTGCTMCAC-3' VH2B-For 5'-AAATCGATACCACCATGGACATACTTTGTTCCAAC-3' VH3-For 5f-AAATCGATACCACCATGGAGTTTGGGCTGAGC-31 VH3B-For 5'-AAATCGATACCACCATGGARYTKKGRCTBHGC-3' VH4-For 5f-AAATCGATACCACCATGAAACACCTGTGGTTCTT-3' VH5-For 5f-AAATCGATACCACCATGGGGTCAACCGCCATC-3' VH6- For 5'-AAATCGATACCACCATGTCTGTCTCCTTCCTC-3' Cgamma-Rev 5'-GGGTCTAGACAGGCAGCCCAGGGCCGCTGTGC-3' Vkappa regions:
Vkl-For 5'-AAATCGATACCACCATGGACATGAGGGTCCCY-3' VklB-For 5f-AAATCGATACCACCATGGACATGAGRGTCCYY-3' Vk2-For 5'-AAATCGATACCACCATGAGGCTCCCTGCTCAG-3' Vk3-For 5'-AAATCGATACCACCATGGAARCCCCAGCGCA-3' Vk4-For 5'-AAATCGATACCACCATGGTGTTGCAGACCCAG-3' Ck-Rev 5'-GATCGCGGCCGCTTATCAACACTCTCCCCTGTTGAAGCTCTT-3' Vlambda regions:
V1laecb 5'-AAATCGATACCACCATGGCCTGGTCCCCTCTCCTCC-3' V11g 5'-AAATCGATACCACCATGGCCGGCTTCCCTCTCCTCC-3' V12/10 5f-AAATCGATACCACCATGGCCTGGGCTCTGCTCCTCC-3' V13jpah 5'-AAATCGATACCACCATGGCCTGGACCGCTCTCCTGC-3' V15/7 5'-AAATCGATACCACCATGGCCTGGACTCCTCTCCTTC-3' V16/9 5'-AAATCGATACCACCATGGCCTGGGCTCCTCTCCTTC-3' V13rm 5'-AAATCGATACCACCATGGCCTGGATCCCTCTCCTCC-3' V131 5'-AAATCGATACCACCATGGCCTGGACCCCTCTCTGGC-3' V1 3 e 5' -AAATCGATACCACCATGGCCTGGGCCACACTCCTGC- 3 ' V1 4 c 5' -AAATCGATACCACCATGGCCTGGGTCTCCTTCTACC- 3 ' V18a 5' -AAATCGATACCACCATGGCCTGGATGATGCTTCTCC-3' Cl 2 / 7 5' -GATCGCGGCCGCTTATCAWGARCATTCTGYAGGGGCCACTG- 3 ' The oligonucleotides used for expression vector constructions were:
Heavy chain expression vector:
VH1-L-NheI: 5'-GCGGCTAGCCACCATGGACTGGACCTGGAGG-3' JH4/5-XhoI: 5'-GCGCTCGAGACGGTGACCAGGGTTCCCTG-3' CHfw-XhoI: 5'-CGCGCTCGAGTGCCTCCACCAAGGGCCCATCGGTC-3' CHrev-NotI: 5'-GATCGCGGCCGCTTATCATTTACCCGGRGACAGGGAGAGGC-3' Light chain expression vector:
VK1-L-NheI: 5'-GCGGCTAGCCACCATGGACATGAGGGTCCCY-3' CK-NotI: 5'-GATCGCGGCCGCTTATCAACACTCTCCCCTGTTGAAGCTCTT-3' EBV RT-PCR
To test if the strong proliferative response was related to the presence of EBV, an EBV RT-PCR was performed. The RT procedure is described above. The PCR
conditions were as follows: a 7-minute denaturing step at 94 C followed by 30 cycles of 30s at 94 C, 30s at 62 C (HPRT1), 52 C (LMP-1) and 58 C (EBNA1/2) and 30s at 72 C, and a final 7-minute extension at 72 C. The oligonucleotides used for RT-PCR were as follows: HPRT1 forward (5'-TATGGACAGGACTGAACGTCTTGC-3') and HPRT1 reverse (5'-GACACAAACATGATTCAAATCCCTGA-3'); LMP-1 forward: (5'-GCGACTCTGCTGGAAATGAT-3') and LMP-1 reverse (5'-GACATGGTAATGCCTAGAAG-3'); EBNA1/2 forward (5'-AGCAAGAAGAGGAGGTGGTAAG-3') and EBNA1/2 reverse (5'-GGCTCAAAGTGGTCTCTAATGC-3').
In addition to the RT-PCR we performed a PCR directly on cell pellet and supernatant DNA that was isolated using the QIAmp isolation kit (Qiagen).

RESULTS
B cell phenotype The use of human memory B cells as the platform to isolate therapeutics medicines relies on the ability to grow and test these cells for a relative long period of time. Human B cells can be cultured and maintained in a laboratory setting however not long enough to expand, select and clone single B cell lines against an antigen of interest. We developed immortalization techniques based on genetic modifications of human B cells. We studied downstream targets of STAT5. One target besides others is Bc1-6. Bc1-6 inhibits differentiation of B
cells to plasma cells that are arrested in proliferation. Overexpression of Bc1-6 keeps BLIMP' in balance, a transcription factor which expression is strongly enhanced by stimulating B cells with IL-21 (works via STAT3). BLIMP1 is necessary to induce the development of Ig producing cells (CD20-CD38+) whereas Bel-6 can prevent this (cells maintain CD20 expression, the so-called germinal center phenotype).
To study the possible skewing of certain cell populations within the B cell compartment, CFSE labeling prior to stimulation of fresh memory and naive human B cells revealed that all cells start dividing and that all populations of B
cells are equally transduced (Figure 2). Shown are memory B cells transduced with Bc1-6 and cultured in the presence of IL-21 and IL-4. Naive B cells were transduced at a lower level and division rates were lower at 36hrs but were identical to memory B cells after another 3 days of culture (data not shown).
Next we show that Bel-6, together with Bel-XL (anti-apoptotic downstream target of STAT5), CD4OL signaling and in the presence of IL-21, maintain human IgG
memory B cells in the CD2O+CD38dull phenotype for long periods of time (>3 months) (Figure 3). In addition, the Bel-6 Bel-XL B cells have a phenotype corresponding to activated B cells (see 'Fable 1, exemplified by FACS staining of 3 TT+ B cell clones), since these cells have high expression of CD80, CD86 and HLA-DR.

determined on three different BcI-6 Bcl-XL B cell clones cultured with IL-21 and CD4OL signaling staining result staining result CD2 neg CD69 neg CD5 neg CD70 pos CD7 neg CD71 pos CD10 pos CD73 neg CD20 pos CD80 pos/high CD21 pos CD86 pos CD22 pos CD95 pos/high CD23 neg/5% pos CD126 neg CD24 neg CD132 (common gamma) pos CD25 pos CD138 neg/2%pos CD27 neg/low CD154 (CD4OL) 8 Wo pos CD28 neg ICOSL pos CD30 pos(56-74%) IgM neg CD38 pos/intermediate IgG pos CD40 pos H LA-DR pos(high) CD44 pos Kappa pos/neg CD45 pos Lambda pos/neg CD45RA pos/high 1L21-R pos Antibody membrane expression The Bc1-6 Bel-XL transduced, EBV negative cells remained BCR expression positive as determined by antigen binding or Kappa and Lambda staining (Figure 3 and 4). Hence, such cells are particularly suitable for isolating and/or screening after a long period of culture for a desired specificity, for instance using labeled antigen, because such cells will bind said labeled antigen with their BCR.
This was confirmed by single cell sorting of Bc1-6 and Bel-XL double transduced B cells that bind PE labeled TT using the FACSAria. After three weeks single cell sorted clones were stained with appropriate markers and TT-PE in 96 well plates and measured for binding in the FACS Canto (BD) (Figure 4). In conclusion, in cases where the presence of a B cell receptor on B cells is desired, such as for instance in screening assays, the B cells are preferably transduced with Bc1-6 and Bel-XL and not infected with EBV.
Cell division and growth curves Bc1-6 Bel-XL transduced B cells divide on average 0.6 times per day. Division rate varies between donors and cell density of the cultures (Figure 5a). The anti-RSV

clone D25 had a division rate of 0.47 times per day (figure 5b). Cells can be grown at densities below 1 cell/96 well for cloning purposes.
Antibody secretion of Bel-6 Bc1-XL B cells The Bc1-6 Bc1-XL transduced B cells secrete on average one Wall of antibodies, which is enough to grow quantities necessary for pre-clinical tests (Figure 6).
Surprisingly the D25 anti-RSV clone produced three times more antibodies compared to the other cell lines tested.
Determine EBV content EBV RT-PCR on mRNA of Bc1-6 Bel-XL cell lines that were cultured with IL-21 and CD4OL signaling. In the cell lines obtained with this immortalization technique no EBV gene transcript have ever been detected (data not shown).
Selection procedure Due to the stability in growth and expression of the BCR, these cells are well suited to isolate antigen-specific B cells. It gave us the opportunity to use several different selection and cloning procedures. One is to immediately obtain antigen specific cells after introduction of Bel-6 and Bel-XL by FACS or Magnetic Bead sorting using labeled antigen of interest thereby enhancing the probability of generating multiple antigen-specific B cell clones. Another option is to grow purified, bulk Bc1-6 Bel-XL transduced memory (or any other) B cells at low cell densities (for example 100 cells/well). Supernatants from these 100c/w cultures can be collected and tested for their specificity. 100 cell/well cultures that are found positive for antigen recognition, are then subcloned by limiting dilution cultures to obtain monoclonal cell lines. Using both methods we could isolate over 40 Tetanus Toxoid (TT) recognizing B cell clones. Thus these clones were either selected on TT binding to the BCE on the FACSAria or they were selected by ELISA screening of series of cultures till the single anti-TT monoclonal cell line was isolated (not shown).
Selection of RSV neutralizing antibodies From donor B63, 25 100 cell/well cultures completely blocked RSV infection and replication. D10, one of the neutralizing 100 cell/well cultures produced a strong anti-RSV antibody which we cloned by limiting dilution culture. One of the monoclonal antibodies, D25 was used to continue studies. D25, a monoclonal antibody with an IgG1 heavy chain, as determined by commercial ELISA
(Sanquin, Amsterdam, not shown) and a Kappa light chain (Figure 7), very efficiently blocked RSV infection with an IC5o value of between 0.5 and 1.5 ng/ml (+ lOpM) whereas the IC50 of the standard anti-RSV antibody used in the clinic (palivizumab developed by MedImmune) is 0.4531Ag/m1 (3.02nM) (H. Wu et al.
2005 J.Mol.Biol. and A. Mejias et al. 2005 Antimicrob. Agents Chemother.) (Figure 8).
Antigen recognition In addition to the neutralization experiments, the binding of D25 to RSV
infected HEp2 cells was determined. HEp2 cell were infected using the regular virus production protocol. HEp2 cells infected with RSV were trypsinized and incubated with 25-50 t1 culture supernatant. Cells were washed and stained with mouse-anti-human IgG-PE (BD or Jackson) to detect binding of the D25 antibody to the infected cells. The r-Biopharm ELISA control antibody was used as an internal control. Shown in figure 9a is the binding of D25 to intact, RSV
infected HEp2 cells.
Since the RSV envelope (membrane) proteins exist of two proteins namely the G
and F-protein, the binding of D25 was tested against cells infected with the VSV
virus pseudotyped with either no or the RSV F or RSV G protein (kindly provided by John K Rose). As shown in figure 9b, D25 bound strongly to EL-4 cells infected with the VSV-F protein. In an attempt to study the epitope recognized by D25 versus palivizumab, VSV-F protein infected EL-4 cells were incubated with increasing amounts of 1)25 or palivizumab. Cells were washed and stained with a mixture of 3 mouse-anti-RSV-F antibodies (Dako). In contrast to Palivizumab that showed competition for the binding to infected VSV-F cells with the mouse-anti-RSV-F antibody, 1)25 binding was not affected (data not shown).
Figure 9c shows the binding of Palivizumab (Synagis) and 1)25 in a concentration dependent manner to infected HEp2 cells. Since both antibodies bind 1 to 1 to their target protein there is no difference in binding to infected HEp2 cells.

Frequency of RSV antigen binding vs neutralizing clones We calculated that the frequency of antigen specific memory B cells that bind RSV was 17% and the frequency of antigen specific cells that neutralize RSV
was 6%, as determined for donor B63. D25 binds to a conformational epitope that is different then the epitope recognized by palivizumab. This is illustrated in figure in which 1)25 does not bind to denatured, linear epitopes presented by lysed RSV infected cell lysate coated on ELISA plates while palivizumab does bind to denatured (F) protein.
10 Isolation and purification of antibody fragments From several B cell lines including the highly RSV neutralizing clone D25 we were able to grow volumes as much as 500 ml. These culture supernatants contain at least 2 g/ml, therefore we should be able to obtain enough purified antibody to perform pre-clinical (animal) studies. The purification is performed using Montage Antigen Purification Kit (Millipore, Billerica, MA, USA) and HiTrap Protein A HP columns (GE Healthcare, Diegem, Belgium).
In addition, 293T cells were transfected with the heavy and light chain of D25 that were subcloned in pCDA3.1 protein expression vectors using lip ofectamine LTX (Invitrogen). The amount of IgG that were present in the supernatant was approximately 2214/m1 (total volume 50 ml). This antibody derived from the cloned nucleotide sequence of the antibody expressed by the 1)25 B cell line did also recognized infected HEp2 cells (data not shown).
Antibody sequence Figure 11a shows the heavy and light chain nucleotide and amino acid sequence of the B63D10-D25 clone. By using standard RT-PCR and antibody specific primers, the heavy (Vh1-69) and light (VkI 08/018) chain sequences were determined. The whole antibody sequence was cloned by using TOPO vectors and after sequence control, suhcloned into the pCDNA3.1 mammalian protein expression vector (Invitrogen). Figure lib and 11c depict the VH and VL4 chain of the clone, Astricks indicate mutations compared to the germline sequence of the Vh1-69 that must have occurred during affinity maturation and further B
cell selection.

To summarize, we here show the isolation, characterization and long-term culture of human memory B cells using the transgenes Bc1-6 and Bel-XL. They give us the tool necessary to isolate antibodies with unique properties, like the anti-RSV monoclonal antibody B63D10-B25. Since the B cells are from a human origin, they can readily be deployed as a therapeutic medicine.

The 1325 heavy and light chain were cloned into standard expression vectors as described before (p44 'antibody sequence). To create an expression construct that allows for maximum protein expression the 1325 heavy and light chain sequences were codon optimized by GENEART (Regensburg, Germany). In this procedure additional restriction sites were created to simplify future cloning procedures but most importantly nucleotide codons that translate into amino acid sequences were optimized for maximum translation into protein. Thus the nucleotide sequence was optimized but the amino acid sequence remained unchanged.
Shown in EXAMPLE 4 is the neutralizing capacity of purified B cell supernatant derived 1125, recombinant 1125 and GENEART optimized 1325. All efficiently neutralize RSV.
The GENEART modifications compared to the original 1125 sequence are depicted in Figure 12.

Next to the in vitro RSV neutralization experiments we tested the D25 monoclonal antibody in in vivo models. The models that have been described for in vivo anti-RSV tests are BALB/c mice and cotton rats (Sigmodon hispidus) (Mejias A et al., Antimicrobial Agents and chemotherapy 2004;p1811, Johnson S
et al., JID 1997;p1215 and Wu H et al., JMB 2007:p652). The BALB/c mouse model is clearly the weakest model but since the cotton rats are difficult to get and maintain, we first set up D25 tests in BALB/c mice.
Protocol: RSV specific antibodies in BALB/c, Day 5 Experimental design:
Day -1. I.P. injection 100111 antibodies Day 0. IN infection lx107 pfu RSV A2 in 50[1.1 Day 1 to 5, check general well being and weigh mice Dag 5, autopsy, collect BAL, blood and lungs Draw blood via vena puncture Collect 2.0m1 BAL via trachea canule Collect lungs Immediately start TCID50 on BAL material (1 ml) Freeze lml BAL material (ELISA cytokine/RT-PCR) -80C
Perform TCID50 on prepared long material (1m1) Freeze 1 ml long material (ELISA cytokine/RT-PCR) -80C
Collect/spin blood for hIgG ELISA on serum en store at -80C
The results are shown in Figure 13:
(A) One day before RSV challenge (1x107 RSV-A2particles) by nasal spray, animals were IP injected with different amounts of Synagis (MedImmune), purified D25 or an IgG1 ctrl antibody (Eureka) (Table 3). (Figure 13B) Human IgG levels were determined in mice sera from day 5 and the drop in antibody serum levels in 5 days; Table 4 shows an overview of the half-life values.
Figure 13D depicts virus titers found in lung lavages (BAL) at day 5 in treated and untreated animals whereas figure 13E depicts T and B cell numbers in peripheral blood of treated and untreated mice. Figure 13F shows the histology of the lungs with bronchi and infiltration of (normally mainly eosinophils) untreated and treated animals.
Conclusion/Result:
An estimate of the D25 half-live is 5 to 9 days based on the (linear) calculation that 60 and 301.tg of antibody was injected on day 0 (2 and 1 mg/kg respectively) and at day 5 33 or 16 g was detected (total volume of mice 1,5). When we started with 0,5mg/kg injection per animal on dO then Ig levels drop from 15 g to 11 g on day 5, which would indicate a 9 day half life (Table 4).
Table 4 mg/kg total administered detected on half-life dO (1.1g) d5 (lig) (days) 2,0 60 33 5,6 1,0 30 16 5,4 0,5 15 11 9,4 Virus titer as determined TCID50 assay shows that in control animals 1x104 PFU can be detected whereas no virus was detected in the Synagis (2mg/kg) or D25 (2, 1 and 0,5mg/kg) treated animals.
Animals treated with Synagis or D25 maintain higher % of peripheral CD4 T
cells and B220 B cells. Animals treated with Synagis (2mg/kg) have lower % of CD4 T cells compared to D25 treated animals. Although this may not be significant it is important to note that animals treat with a low dose of D25 (1 and 0,5 mg/kg) maintain high levels of B and T cells when compared to control treated animals.
Although the histology data (figure 13F) are not quantitative it is clear that Synagis and D25 reduce influx of immune cells into the lungs and around the bronchi compared to control. When D25 and Synagis are compared, then 1125 treated animals seem to have less cellular infiltration into the lungs and around the bronchi.

To test D25 in the Cottonrats, experiments are set up to compare animals pre-treated with Synagis and D25 before challenge with the RSV-X virus at the NVI
(Bilthoven, Netherlands).

In addition to B63-D10-D25, we isolated three new potent RSV neutralizing antibodies (AM14, AM16 and AM23) from the same donor (B63). 100 cell per well bulk B cell cultures that were originally selected for RSV neutralization and were frozen and stored in liquid nitrogen, were thawed and culture supernatant was tested for binding to RSV infected HEp2 cells. We tested for binding to infected Hep2 cells since that is a marker for antibody recognition of native, oligomeric RSV membrane proteins like F and G protein and may serve as a good predictor for neutralization. When binding was detected, cells were single cell cultured and screened for binding to obtain clones. All three antibodies were cloned into the GENEART vector that was originally constructed for D25. In addition like D25 all recognize the RSV-F protein (not shown). After cloning and expression in 293T cells recombinant protein was purified (nucleotide and amino acid sequences are depicted in figure 14A, B and C). Antibodies were tested for neutralization against several primary RSV isolates on Vero and HEp2 cells (Figure 15). All three antibodies are of the IgG1 isotype. AM14 has a Kappa light chain, while AM16 and AM23 have a Lambda light chain. All three antibodies, like D25, contain somatic hypermutations in their antibody variable domains suggesting that they in vivo have undergone affinity maturation during a germinal center reaction, a process that creates unique antibody sequences.
The results are shown in Figures 15-I and 15-II: RS virus neutralization assay with purified B cell line supernatant derived D25 (sD25), recombinant purified D25 (rD25), recombinant GENEART codon optimized D25 (rD25 GA), AM14, AM16, AM23 (all purified recombinant protein) and Synagis. Virus antibody neutralization was tested on two different cell lines (Figure 15-I) Vero and (Figure 15-II) Hep2 cells with different antibodies: A2 (A), X (B) and 2006/1 (C) are RSV subtype A while virus Z (D) and 2007-2 (E) are subtype B. 100TCID50 of each virus was added to serial antibody dilutions in DMEM/1%FCS and incubated for 1 hour at 37 degree before 100u1 Vero or HEp2 cells (1x106/m1) were added. Virus antibody mixture was not washed away. After three days supernatant was removed and cells were fixed with 80% acetone for 10' at RT.
After removal of the acetone, the fixed cell layer was dried and kept at 4 C
or frozen at -20 C. To stain RSV infected HEp2 cells, plates were first blocked with 5% milkpower in PBS 0.1% Tween 20, then plates were washed 3 times before being incubated for 3-5 hours at 37 C with polyclonal goat anti-RSV-HRP
(1:500, Biodesign, Saco, ME, US) and washed extensively. Subsequently all wells were incubated with AEC substrate for 30' at RT. Infected foci stain red and can be observed by eye using a light microscope and can be counted.
Result/conclusion All antibodies neutralize the RSV A and B strains (Table 5). In general the different D25 antibodies neutralize the RSV viruses efficiently, although minor inter-experimental variations can be seen. AM14 is just as potent as D25 while AM16 is just as potent as Synagis. AM23 however does neutralize the RSV A
strains very efficient, while it is less potent in neutralizing RSV B strains, although still comparable to Synagis.
Table 5 IC50 values (ng/ml) Cell line RSV
sD25 rD25 rD25 GA AM14 AM16 A1\423 used subtype Vero A 3.4 1.6 3.2 15.2 304.3 19.4 Vero B 9.0 0.3 1.2 1.1 126.4 168.8 HEp2 A 3.3 2.1 5.3 21.5 285.6 25.0 HEp2 B 14.3 1.9 1.3 6.7 124.8 190.7 The IC50 value for each antibody on RS virus subtype A on Vero or HEp2 cells was calculated as the average 50% neutralization on three virus strains (A2, X and 2006-1). The 1050 value for each antibody on RS virus subtype B on Vero or HEp2 cells was calculated as the average 50%
neutralization on two virus strains (2007-2 and Z). Each of the neutralizations assays was performed in triplo and repeated twice (also shown in figure 15A and B).
sD25 = purified B cell derived culture supernatant rD25 = purified recombinant 1)25 r1325 GA = supernatant of 293T cells with GENEART codon optimized recombinnat Synergistic and blocking effects of anti-RSV antibodies.
To analyze whether D25, Synagis or the new AM antibody set interfere with each other for recognition of the RSV F protein, we pre-incubated RSV infected HEp2 cells with increasing concentrations of unlabeled antibodies till they reached the plateau of maximum binding. We determine for each antibody the plateau phase in which no increase in binding was detected when the amount of Ig was increased. (not shown). After washing, samples were incubated with a standard dose (3 pmol) of PE labeled D25 or APC labeled Synagis. This dose gives also maximum binding.
Result As shown in Figure 16 labeled Synagis and D25 show a reduced binding to RSV
infected HEp2 cells when these cells were pre-incubated with either unlabeled Synagis or D25. Synagis shows furthermore a slight reduction in binding induced by AM16. D25 binding is strongly blocked by AM23 but on the contrary D25 binding is strongly enhanced after pre-incubation with A1VI14. That indicates that the epitope recognized by D25 is normally not even fully exposed but exposure is enhanced after binding of AM14 to its native epitope. That demonstrates that these two antibodies can work together and enhance neutralization.

Brief description of the drawings Figure 1.
Isolation of human, IgG positive, memory B cells. PBMC isolated from buffy coat using Ficoll density separation (Amersham) were incubated with anti-CD22 magnetic beads before being isolated using MACS columns (Miltenyi). CD22 positive cells were then incubated with antibodies against human CD19, CD27, IgM, IgD and IgA (BD). Cells negative for IgM, IgD and IgA and positive for CD19 and CD27 were sorted using high speed single cell sorting (FACSAria, BD).
Figure 2 CFSE staining. Fresh human memory B cells were isolated, labeled with CSFE
and stimulated for 36h with IL-21 before being transduced with Bel-6-IRES-NGFR. Cells were kept an additional 3 days on IL-21 before CFSE content was determined. The CFSE dye is diluted with every cell division.
Figure 3 An example of human B cells transduced with Bc1-6 and Bel-XL or Bcl-XL only.
Cells were maintained on irradiated L cells expressing CD4OL and the cy-tokine IL-21. Shown on the left is the BCR expression as determined by kappa and lambda staining (93% of the kappa lambda positive cells are of the IgG
isotype, not shown). On the right is shown the CD38 expression on the X-axes and CD20 expression on the Y-axes. The CD38d"11CD20+ staining indicates memory or germinal center B cells; the CD38+CD20 staining indicate plasmablasts.
Figure 4 Isolation of immortalized, antigen specific human B cells. Human memory B
cells were isolated as described in figure 1 and subsequently transduced with Bc1-6-IRES-NGFR and Bc1-XL-IRES-GFP. Cells expressing NGFR, GFP and were binding to PE-labeled Tetanus Toxin were isolated using the FACSAria. Cells were single cell cultured in 96 well flat bottom plates in the presence of irradiated L cells and IL-21 before being selected based on TT-PE binding using the FACS Canto (BD).

Figure 5 Cumulative cell growth and division rate of 6XL B cell clones. B cells from (A) two anti-TT clones and (B) one anti-RSV clone (B63D10-D25) were cultured in the presence of IL-21 and irradiated L cells.
Figure 6 Fresh cultures were started with 200,000 cell/24 well in 1,0 ml IMDM with 8%FCS and pen/strep. The FCS used was either normal (HyClone) or Ultralow Bovine IgG FCS (Gibco). After 3 days the culture supernatant was replaced and cell numbers were adjusted to 200,000 cell/ml. Shown is the average IgG
production in 3 days measured in 3 consecutive time points the difference was not significant (p value 0.2).
Figure 7 To determine the light chain phenotype of the 1325 anti-RSV clone, the 1325 B
cell line was stained with either kappa-phycoerythrin or lambda-phycoerythrin (BD) antibodies. Only the kappa- phycoerythrin antibodies bound to the cell line, showing this antibody has a kappa light chain.
Figure 8 From donor B63, 100 cell/well cultures were grown using Bc1-6 Bc1-XL positive human memory B cells. One of those cultures, D10 showed strong neutralization.

LD derived monoclonal cell lines were made, one 1325 neutralized the RSV A-2 virus efficiently. Shown here is D25 compared to palivizumab (synag-is) and a polyclonal goat ant-RSV. Not shown are irrelevant culture supernatants of Bc16 Bcl-XL transduced B cell clones cultured with IL-21 and CD4OL signaling that produce high levels of antibodies but did not block RSV infection The D25 clone was used for further characterization.
Figure 9 In figure 9a: 1-1Ep2 cell were seeded at 10-12e6 cells per T175 flask (Nunc) in IMDM/5910FCS. The next day the medium was replaced with 5m1 of medium with RSV virus (1.0 MOI) and incubated for 45' at RT before 20 ml of fresh medium was added and the cells were cultured o/n at 37 C. The next day the medium was replaced with IMDM/1%FCS and cultured o/n with a closed lid at 37 C. The next day cells were washed with PBS and treated with trypsin. To stain infected cells the primary incubation was performed with culture supernatant. The secondary incubation was done with anti-human IgG-PE (BD). Cells were analyzed using the LSRII (BD). As a positive control the positive control of the commercial ELISA KIT from r-Biopharm was used.
In figure 9b: EL-4 cells were infected with VSV virus pseudotyped with RSV F
or G protein (kindly provided by John Rose) and incubated with D25 culture supernatant. Cells were washed and incubated with anti-human-IgG-PE
(Jackson) to detect binding of D25 to the infected cells. Only binding of D25 to the VSV virus infected cells pseudotyped with the RSV F protein was detected.
Figure 9c shows the binding of Palivizumab (Synagis) and D25 in a concentration dependent manner to infected HEp2 cells. Shown is the mean fluorescence intensity (MF1).
Figure 10 Binding of polyclonal goat anti-RSV (pos ctrl), palivizumab (synagis) and D25 to coated HEp2 infected cell lysate.
Figure 11 Sequence analysis of the D25 clone. 11a shows nucleotide and predicted amino acid sequence of the variable heavy and light chain domains. 11b/c show the heavy and light chain sequence compared to predicted germline. Asterisks indicate mutations that probably occurred during selection and affinity maturation of the B cell clone in vivo.
Figure 12 Cloning and expression of recombinant human antibodies from BCL6 BCL-xL
transduced B cell lines. This has already been described for the D25 antibody (Figure 11). Here are depicted the GENEART nucleotide modifications compared to the original D25 sequence, note that these mutations do not change the amino acid composition of the D25 antibody.
Figure 13 BALB/c mice challenge with purified, B cell supernatant derived D25 and Synagis. (A) One day before RSV challenge (1x107 RSV-A2particles) by nasal spray, animals were IP injected with different amounts of Synagis (MedImmune), purified D25 or an IgG1 ctrl antibody (Eureka)(table 3). (B) human IgG levels were determined in mice sera from day 5 and the drop in antibody serum levels in 5 days (C); table 4 shows an overview of the half-life values. Figure 13D
depicts virus titers found in lung lavages (BAL) at day 5 in treated and untreated animals whereas figure 13E depicts T and B cell numbers in peripheral blood of treated and untreated mice. (F) shows the histology of the lungs with bronchi and infiltration of (normally mainly eosinophils) untreated and treated animals.
Figure 14 Nucleotide and amino acid sequences of three new potent RSV neutralizing antibodies (A) AM14, (B) AM16 and (C) AM23.
Figure 15 RS virus neutralization assay with purified B cell line supernatant derived (sD25), recombinant purified D25 (rD25), recombinant GENEART codon optimized D25 (rD25 GA), AM14, AM16, AM23 (all purified recombinant protein) and Synagis. Virus antibody neutralization was tested on two different cell lines (figure 15-1) Vero and (figure 15-II) Hep2 cells with different antibodies A2 (A), X
(B) and 2006/1 (C) are RSV subtype A while virus Z (D) and 2007-2 (E) are subtype B. 100TCID50 of each virus was added to serial antibody dilutions in DMEM/1%FCS and incubated for 1 hour at 37 degree before 100u1 Vero or HEp2 cells (1x10qm1) were added.
Figure 16 Relative binding of a fixed amount (3pmol) of APC-labeled Synagis and PE-labeled rD25 to RSV infected HEp2 cells that were pre-incubated with increasing concentrations of the indicated unlabeled antibodies.

References Banchereau, J. , de Paoli, P. , Valle, A. , Garcia, E. , Rousset, F., (1991).
Long term human B cell lines dependent on interleukin-4 and antibody to CD40, Science 251, 70-2.
Boise, L. H., M. Gonzalez-Garcia, C. E. Postema, L. Ding, T. Lindsten, L. A.
Turka, X. Mao, G. Nunez, and C. B. Thompson. (1993). Bcl-x, a bc1-2-related gene that functions as a dominant regulator of apoptotic cell death. Cell 74:597.
Dadgostar, H., Zarnegar, B. , Hoffmann, A. , Qin, X. F. , Truong, U. , Rao, G., Baltimore, D. , and Cheng, G. (2002). Cooperation of multiple signaling pathways in CD40-regulated gene expression in B lymphocytes. Proc.Natl.Acad.Sci USA 99, 1497-1502.
Heemskerk et al, 1997: J.Exp.Med. Vol 186, page 1597-1602 Heemskerk et al, 1999: Cell Immunol. Vol 195, page 10-17 Kinsella and Nolan, 1996: Hum. Gene Ther. Vol 7 page 1405-1413 Malisan, F. , Briere, F., Bridon, J.M. , Harindranath, N. , Mills, F. C. , Max, E.
E. , Banchereau, J. , Martinez-Valdez, H. (1996). Interleukin-10 induces immunoglobulin G isotype switch recombination in human CD40-activated naive B lymphocytes, J.Exp.Med. 183, 937-47.
Mathas S, Janz M, Hummel F, Hummel M, Wollert-Wulf B, Lusatis S, Anagnostopoulos I, Lietz A, Sigvardsson M, Jundt F, Johrens K, Bommert K, Stein H, Dorken B (2006). Intrinsic inhibition of transcription factor E2A by HLH
proteins ABF-1 and Id2 mediates reprogramming of neoplastic B cells in Hodgkin lymphoma. Nat Immunol. 7, 207-215.
Mejias A et al., Antimicrobial Agents and chemotherapy 2004;p1811, Johnson S
et al., JID 1997;p1215 Wu H et al., JMB 2007:p652 Shvarts A. et al, 2002: Genes Dev. Vol 16, page 681-686 Traggiai, E. , Becker, S., Subbarao, K. , Kolesnikova, L., Uematsu, Y., Gismondo, M.R. , Murphy, B.R. , Rappuoli, R. , Lanzavecchia, A. (2004). An efficient method to make human monoclonal antibodies from memory B cells:
potent neutralization of SARS coronavirus. Nature Medicine Volume 10, No. 8, 871-875.
Ye, B. H., Cattoretti, G. , Shen, Q. , Zhang, J., Hawe, N., de Waard, R. , Leung, C. , Nouri-Shirazi, M. , Orazi, A., Chaganti, R. S., et al. (1997). The BCL-6 proto-oncogene controls germinal-centre formation and Th2-type inflammation. Nat Genet 16, 161-170.

SEQUENCE LISTING IN ELECTRONIC FORM
In accordance with Section 111(1) of the Patent Rules, this description contains a sequence listing in electronic form in ASCII text format (file: 76433-194D1 Seq 07-NOV-16 vl.txt).
A copy of the sequence listing in electronic form is available from the Canadian Intellectual Property Office.
The sequences in the sequence listing in electronic form are reproduced in the following table.
SEQUENCE TABLE
<110> MedImmune Limited <120> RSV-specific binding molecules and means for producing them <130> 76433-194D1 <140> Division of CA 2,689,290 <141> 2008-05-30 <150> EP07109472.6 <151> 2007-06-01 <160> 160 <170> PatentIn version 3.3 <210> 1 <211> 5 <212> PRT
<213> Homo sapiens <400> 1 Asn Tyr Ile Ile Asn <210> 2 <211> 17 <212> PRT
<213> Homo sapiens <400> 2 Gly Ile Ile Pro Val Leu Gly Thr Val His Tyr Ala Pro Lys Phe Gin Gly <210> 3 <211> 17 <212> PRT
<213> Homo sapiens <400> 3 Glu Thr Ala Leu Val Val Ser Thr Thr Tyr Leu Pro His Tyr Phe Asp Asn <210> 4 <211> 11 <212> PRT
<213> Homo sapiens <400> 4 Gin Ala Ser Gin Asp Ile Val Asn Tyr Leu Asn <210> 5 <211> 7 <212> PRT
<213> Homo sapiens <400> 5 Val Ala Ser Asn Leu Glu Thr <210> 6 <211> 7 <212> PRT
<213> Homo sapiens <400> 6 Gin Gin Tyr Asp Asn Leu Pro <210> 7 <211> 126 <212> PRT
<213> Homo sapiens <400> 7 Gin Val Gin Leu Val Gin Ser Gly Ala Glu Val Lys Lys Pro Gly Ser Ser Val Met Val Ser Cys Gin Ala Ser Gly Gly Pro Leu Arg Asn Tyr Ile Ile Asn Trp Leu Arg Gin Ala Pro Gil/ Gin Gly Pro Glu Trp Met Gly Gly Ile Ile Pro Val Leu Gly Thr Val His Tyr Ala Pro Lys Phe Gin Gly Arg Val Thr Ile Thr Ala Asp Glu Ser Thr Asp Thr Ala Tyr Ile His Leu Ile Ser Leu Arg Ser Glu Asp Thr Ala Met Tyr Tyr Cys Ala Thr Glu Thr Ala Leu Val Val Ser Thr Thr Tyr Lou Pro His Tyr Phe Asp Asn Trp Gly Gin Gly Thr Leu Val Thr Val Ser Ser <210> 8 <211> 110 <212> PRT
<213> Homo sapiens <400> 8 Asp Ile Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ala Val Gly Asp Arg Val Thr Ile Thr Cys Gin Ala Ser Gin Asp Ile Val Asn Tyr Leu Asn Trp Tyr Gin Gin Lys Pro Gly Lys Ala Pro Lys Leu Leu Ile Tyr Val Ala Ser Asn Leu Glu Thr Gly Val Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Ser Lou Thr Ile Ser Ser Lou Gin Pro Glu Asp Val Ala Thr Tyr Tyr Cys Gin Gin Tyr Asp Asn Leu Pro Leu Thr Phe Gly Gly Gly Thr Lys Val Glu Ile Lys Arg Thr Val <210> 9 <211> 379 <212> DNA
<213> Homo sapiens <400> 9 caggtgcagc tggtacagtc tggggctgaa gtgaagaagc ctgggtcctc ggtgatggtc 60 tcctgccagg cctctggagg ccccctcaga aactatatta tcaactggct acgacaggcc 120 cctggacaag gccctgagtg gatgggaggg atcattcctg tcttgggtac agtacactac 180 gcaccgaagt tccagggcag agtcacgatt accgcggacg aatccacaga cacagcctac 240 atccatctga tcagcctgag atctgaggac acggccatgt attactgtgc gacggaaaca 300 gctctggttg tatctactac ctacctacca cactactttg acaactgggg ccagggaacc 360 ctggtcaccg tctcctcag 379 <210> 10 <211> 324 <212> DNA
<213> Homo sapiens <400> 10 gacatccaga tgacccagtc tccatcctcc ctgtctgcag ctgtaggaga cagagtcacc 60 atcacttgcc aggcgagtca ggacattgtc aactatttaa attggtatca acagaaacca 120 gggaaagccc ctaagctcct gatctacgtt gcatccaatt tggagacagg ggtcccatca 180 aggttcagtg gaagtggatc tgggacagat tttagtctca ccatcagcag cctgcagcct 240 gaagatgttg caacatatta ttgtcaacaa tatgataatc tcccactcac attcggcgga 300 gggaccaagg ttgagatcaa aaga 324 <210> 11 <211> 32 <212> DNA
<213> artificial sequence <220>
<223> Primer VH1 <400> 11 aaatcgatac caccatggac tggacctgga gg 32 <210> 12 <211> 32 <212> DNA
<213> artificial sequence <220>
<223> Primer VH1B
<400> 12 aaatcgatac caccatggac tggacctgga gm 32 <210> 13 <211> 34 <212> DNA
<213> artificial sequence <220>
<223> Primer VH2A
<400> 13 aaatcgatac caccatggac acactttgct mcac 34 <210> 14 <211> 35 <212> DNA
<213> artificial sequence <220>
<223> Primer VH2B
<400> 14 aaatcgatac caccatggac atactttgtt ccaac 35 <210> 15 <211> 32 <212> DNA
<213> artificial sequence <220>
<223> Primer VH3 <400> 15 aaatcgatac caccatggag tttgggctga gc 32 <210> 16 <211> 32 <212> DNA
<213> artificial sequence <220>
<223> Primer VH3B
<400> 16 aaatcgatac caccatggar ytkkgrctbh gc 32 <210> 17 <211> 34 <212> DNA
<213> artificial sequence <220>
<223> Primer VH4 <400> 17 aaatcgatac caccatgaaa cacctgtggt tctt 34 <210> 18 <211> 32 <212> DNA
<213> artificial sequence <220>
<223> Primer VH5 <400> 18 aaatcgatac caccatgggg tcaaccgcca tc 32 <210> 19 <211> 32 <212> DNA
<213> artificial sequence <220>
<223> Primer VH6 <400> 19 aaatcgatac caccatgtct gtctccttcc tc 32 <210> 20 <211> 32 <212> DNA
<213> artificial sequence <220>
<223> Primer Cgamma-Rev <400> 20 gggtctagac aggcagccca gggccgctgt gc 32 <210> 21 <211> 32 <212> DNA
<213> artificial sequence <220>
<223> Primer Vkl <400> 21 aaatcgatac caccatggac atgagggtcc cy 32 <210> 22 <211> 32 <212> DNA
<213> artificial sequence <220>
<223> Primer VklB
<400> 22 aaatcgatac caccatggac atgagrgtcc yy 32 <210> 23 <211> 32 <212> DNA
<213> artificial sequence <220>
<223> Primer Vk2 <400> 23 aaatcgatac caccatgagg ctccctgctc ag 32 <210> 24 <211> 31 <212> DNA
<213> artificial sequence <220>
<223> Primer Vk3 <400> 24 aaatcgatac caccatggaa rccccagcgc a 31 <210> 25 <211> 32 <212> DNA
<213> artificial sequence <220>
<223> Primer Vk4 <400> 25 aaatcgatac caccatggtg ttgcagaccc ag 32 <210> 26 <211> 42 <212> DNA
<213> artificial sequence <220>
<223> Primer Ck-Rev <400> 26 gatcgcggcc gcttatcaac actctcccct gttgaagctc tt 42 <210> 27 <211> 36 <212> DNA
<213> artificial sequence <220>
<223> Primer Vllaecb <400> 27 aaatcgatac caccatggcc tggtcccctc tcctcc 36 <210> 28 <211> 36 <212> DNA
<213> artificial sequence <220>
<223> Primer Vllg <400> 28 aaatcgatac caccatggcc ggcttccctc tcctcc 36 <210> 29 <211> 36 <212> DNA
<213> artificial sequence <220>
<223> Primer V12/10 <400> 29 aaatcgatac caccatggcc tgggctctgc tcctcc 36 <210> 30 <211> 36 <212> DNA
<213> artificial sequence <220>
<223> Primer V13jpah <400> 30 aaatcgatac caccatggcc tggaccgctc tcctgc 36 <210> 31 <211> 36 <212> DNA
<213> artificial sequence <220>
<223> Primer V15/7 <400> 31 aaatcgatac caccatggcc tggactcctc tccttc 36 <210> 32 <211> 36 <212> DNA
<213> artificial sequence <220>
<223> Primer V16/9 <400> 32 aaatcgatac caccatggcc tgggctcctc tccttc 36 <210> 33 <211> 36 <212> DNA
<213> artificial sequence <220>
<223> Primer V13rm <400> 33 aaatcgatac caccatggcc tggatccctc tcctcc 36 <210> 34 <211> 36 <212> DNA
<213> artificial sequence <220>
<223> Primer V131 <400> 34 aaatcgatac caccatggcc tggacccctc tctggc 36 <210> 35 <211> 36 <212> DNA
<213> artificial sequence <220>
<223> Primer V13e <400> 35 aaatcgatac caccatggcc tgggccacac tcctgc 36 <210> 36 <211> 36 <212> DNA
<213> artificial sequence <220>
<223> Primer V14c <400> 36 aaatcgatac caccatggcc tgggtctcct tctacc 36 <210> 37 <211> 36 <212> DNA
<213> artificial sequence <220>
<223> Primer V18a <400> 37 aaatcgatac caccatggcc tggatgatgc ttctcc 36 <210> 32 <211> 41 <212> DNA
<213> artificial sequence <220>
<223> Primer C12/7 <400> 38 gatcgcggcc gcttatcawg arcattctgy aggggccact g 41 <210> 39 <211> 31 <212> DNA
<213> artificial sequence <220>
<223> Primer VH1-L-NheI
<400> 39 gcggctagcc accatggact ggacctggag g 31 <210> 40 <211> 29 <212> DNA
<213> artificial sequence <220>
<223> Primer JH4/5-XhoI
<400> 40 gcgctcgaga cggtgaccag ggttccctg 29 <210> 41 <211> 35 <212> DNA
<213> artificial sequence <220>
<223> Primer CHfw-XhoI
<400> 41 cgcgctcgag tgcctccacc aagggcccat cggtc 35 <210> 42 <211> 41 <212> DNA
<213> artificial sequence <220>
<223> Primer CHrev-NotI
<400> 42 gatcgcggcc gcttatcatt tacccggrga cagggagagg c 41 <210> 43 <211> 31 <212> DNA
<213> artificial sequence <220>
<223> Primer VKl-L-NheI
<400> 43 gcggctagcc accatggaca tgagggtccc y 31 <210> 44 <211> 42 <212> DNA
<213> artificial sequence <220>
<223> Primer CK-NotT
<400> 44 gatcgcggcc gcttatcaac actctcccct gttgaagctc tt 42 <210> 45 <211> 24 <212> DNA
<213> artificial sequence <220>
<223> Primer HPRT1 forward <400> 45 tatggacagg actgaacgtc ttgc 24 <210> 46 <211> 26 <212> DNA
<213> artificial sequence <220>
<223> Primer HPRT1 reverse <400> 46 gacacaaaca tgattcaaat ccctga 26 <210> 47 <211> 20 <212> DNA
<213> artificial sequence <220>
<223> Primer LMP-1 forward <400> 47 gcgactctgc tggaaatgat 20 <210> 48 <211> 20 <212> DNA
<213> artificial sequence <220>
<223> Primer LMP-1 reverse <400> 48 gacatggtaa tgcctagaag 20 <210> 49 <211> 22 <212> DNA
<213> artificial sequence <220>
<223> Primer EBNA1/2 forward <400> 49 agcaagaaga ggaggtggta ag 22 <210> 50 <211> 22 <212> DNA
<213> artificial sequence <220>
<223> Primer EBNA1/2 reverse <400> 50 ggctcaaagt ggtctctaat gc 22 <210> 51 <211> 379 <212> DNA
<213> artificial sequence <220>
<223> Anti-RSV clone B63D10-D25 <220>
<221> CDS
<222> (1)..(378) <400> 51 cag gtg cag ctg gta cag tct ggg gct gaa gtg aag aag cct ggg tcc 48 Gin Val Gin Leu Val Gin Ser Gly Ala Glu Val Lys Lys Pro Gly Ser tcg gtg atg gtc tcc tgc cag gcc tct gga ggc ccc ctc aga aac tat 96 Ser Val Met Val Ser Cys Gin Ala Ser Gly Gly Pro Leu Arg Asn Tyr att atc aac tgg cta cga cag gcc cct gga caa ggc cct gag tgg atg 144 Ile Ile Asn Trp Leu Arg Gin Ala Pro Gly Gin Gly Pro Glu Trp Met gga ggg atc att cct gtc ttg ggt aca gta cac tac gca ccg aag ttc 192 Gly Gly Ile Ile Pro Val Leu Gly Thr Val His Tyr Ala Pro Lys Phe cag ggc aga gtc acg att acc gcg gac gaa tcc aca gac aca gcc tac 240 Gin Gly Arg Val Thr Ile Thr Ala Asp Glu Ser Thr Asp Thr Ala Tyr atc cat ctg atc agc ctg aga tct gag gac acg gcc atg tat tac tgt 288 Ile His Leu Ile Ser Leu Arg Ser Glu Asp Thr Ala Met Tyr Tyr Cys gcg acg gaa aca got ctg gtt gta tct act acc tac cta cca cac tac 336 Ala Thr Glu Thr Ala Leu Val Val Ser Thr Thr Tyr Leu Pro His Tyr ttt gac aac tgg ggc cag gga acc ctg gtc acc gtc tcc tca g 379 Phe Asp Asn Trp Gly Gin Gly Thr Leu Val Thr Val Ser Ser <210> 52 <211> 126 <212> PRT
<213> artificial sequence <220>
<223> Synthetic Construct <400> 52 Gin Val Gin Leu Val Gin Ser Gly Ala Glu Val Lys Lys Pro Gly Ser Ser Val Met Val Ser Cys Gin Ala Ser Gly Gly Pro Leu Arg Asn Tyr Ile Ile Asn Trp Leu Arg Gin Ala Pro Gly Gin Gly Pro Glu Trp Met Gly Gly Ile Ile Pro Val Leu Gly Thr Val His Tyr Ala Pro Lys Phe Gin Gly Arg Val Thr Ile Thr Ala Asp Glu Ser Thr Asp Thr Ala Tyr Ile His Leu Ile Ser Leu Arg Ser Glu Asp Thr Ala Met Tyr Tyr Cys Ala Thr Glu Thr Ala Leu Val Val Ser Thr Thr Tyr Leu Pro His Tyr Phe Asp Asn Trp Gly Gin Gly Thr Leu Val Thr Val Ser Ser <210> 53 <211> 324 <212> DNA
<213> artificial sequence <220>
<223> VL region <220>
<221> CDS
<222> (1)..(324) <400> 53 gac atc cag atg acc cag tct cca tcc tcc ctg tct gca gct gta gga 48 Asp Ile Gin Met Thr Gln Ser Pro Ser Ser Leu Ser Ala Ala Val Gly gac aga gtc acc atc act tgc cag gcg agt cag gac att gtc aac tat 96 Asp Arg Val Thr Ile Thr Cys Gin Ala Ser Gin Asp Ile Val Asn Tyr tta aat tgg tat caa cag aaa cca ggg aaa gcc cct aag ctc ctg atc 144 Leu Asn Trp Tyr Gin Gin Lys Pro Gly Lys Ala Pro Lys Leu Leu Ile tac gtt gca tcc aat ttg gag aca ggg gtc cca tca agg ttc agt gga 192 Tyr Val Ala Ser Asn Leu Glu Thr Gly Val Pro Ser Arg Phe Ser Gly agt gga tct ggg aca gat ttt agt ctc acc atc agc agc ctg cag cct 240 Ser Gly Ser Gly Thr Asp Phe Ser Leu Thr Ile Ser Ser Leu Gin Pro gaa gat gtt gca aca tat tat tgt caa caa tat gat aat ctc cca ctc 288 Glu Asp Val Ala Thr Tyr Tyr Cys Gin Gln Tyr Asp Asn Leu Pro Leu aca ttc ggc gga ggg acc aag gtt gag atc aaa aga 324 Thr Phe Gly Gly Gly Thr Lys Val Glu Ile Lys Arg <210> 54 <211> 108 <212> PRT
<213> artificial sequence <220>
<223> Synthetic Construct <400> 54 Asp Ile Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ala Val Gly Asp Arg Val Thr Ile Thr Cys Gin Ala Ser Gin Asp Ile Val Asn Tyr Leu Asn Trp Tyr Gin Gin Lys Pro Gly Lys Ala Pro Lys Leu Leu Ile Tyr Val Ala Ser Asn Leu Glu Thr Gly Val Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Ser Leu Thr Ile Ser Ser Leu Gin Pro Glu Asp Val Ala Thr Tyr Tyr Cys Gin Gin Tyr Asp Asn Leu Pro Leu Thr Phe Gly Gly Gly Thr Lys Val Glu Ile Lys Arg <210> 55 <211> 98 <212> PRT
<213> artificial sequence <220>
<223> VH1-69 germl <400> 55 Gin Val Gin Leu Val Gin Ser Gly Ala Glu Val Lys Lys Pro Gly Ser Ser Val Lys Val Ser Cys Lys Ala Ser Gly Gly Thr Phe Ser Ser Tyr Ala Ile Ser Trp Val Arg Gin Ala Pro Gly Gin Gly Leu Glu Trp Met Gly Gly Ile Ile Pro Ile Phe Gly Thr Ala Asn Tyr Ala Gin Lys Phe Gin Gly Arg Val Thr Ile Thr Ala Asp Glu Ser Thr Ser Thr Ala Tyr Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys Ala Arg <210> 56 <211> 126 <212> PRT
<213> artificial sequence <220>
<223> 863D1O-D25 <400> 56 Gin Val Gin Leu Val Gin Ser Gly Ala Glu Val Lys Lys Pro Gly Ser Ser Val Met Val Ser Cys Gin Ala Ser Gly Gly Pro Leu Arg Asn Tyr Ile Ile Asn Trp Leu Arg Gin Ala Pro Gly Gin Gly Pro Glu Trp Met Gly Gly Ile Ile Pro Val Leu Gly Thr Val His Tyr Ala Pro Lys Phe Gin Gly Arg Val Thr Ile Thr Ala Asp Glu Ser Thr Asp Thr Ala Tyr Ile His Leu Ile Ser Leu Arg Ser Glu Asp Thr Ala Met Tyr Tyr Cys Ala Thr Glu Thr Ala Leu Val Val Ser Thr Thr Tyr Leu Pro His Tyr Phe Asp Asn Trp Gly Gin Gly Thr Leu Val Thr Val Ser Ser <210> 57 <211> 95 <212> PRT
<213> artificial sequence <220>
<223> VkI 08/018 <400> 57 Asp Ile Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser Val Gly Asp Arg Val Thr Ile Thr Cys Gin Ala Ser Gin Asp Ile Ser Asn Tyr Leu Asn Trp Tyr Gin Gin Lys Pro Gly Lys Ala Pro Lys Leu Leu Ile Tyr Asp Ala Ser Asn Leu Glu Thr Gly Val Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Phe Thr Ile Ser Ser Leu Gin Pro Glu Asp lie Ala Thr Tyr Tyr Cys Gin Gin Tyr Asp Asn Leu Pro <210> 58 <211> 110 <212> PRT
<213> artificial sequence <220>
<223> B63D10-D25 <400> 58 Asp Ile Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ala Val Gly Asp Arg Val Thr Ile Thr Cys Gin Ala Ser Gin Asp Ile Val Asn Tyr Leu Asn Trp Tyr Gin Gln Lys Pro Gly Lys Ala Pro Lys Leu Leu Ile Tyr Val Ala Ser Asn Leu Glu Thr Gly Val Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Ser Leu Thr Ile Ser Ser Leu Gin Pro Glu Asp Val Ala Thr Tyr Tyr Cys Gin Gin Tyr Asp Asn Leu Pro Leu Thr Phe Gly Gly Gly Thr Lys Val Glu Ile Lys Arg Thr Val <210> 59 <211> 91 <212> DNA
<213> artificial sequence <220>
<223> FR1 VHeavy region 025 <400> 59 caggtgcagc tggtacagtc tggggctgaa gtgaagaagc ctgggtcctc ggtgatggtc 60 tcctgccagg cctctggagg ccccctcaga a 91 <210> 60 <211> 14 <212> DNA
<213> artificial sequence <220>
<223> CDR1 VHeavy region D25 <400> 60 actatattat caac 14 <210> 61 <211> 42 <212> DNA
<213> artificial sequence <220>
<223> FR2 VHeavy region D25 <400> 61 tggctacgac aggcccctgg acaaggccct gagtggatgg ga 42 <210> 62 <211> 51 <212> DNA
<213> artificial sequence <220>
<223> CDR2 VHeavy region D25 <400> 62 gggatcattc ctgtcttggg tacagtacac tacgcaccga agttccaggg c 51 <210> 63 <211> 96 <212> DNA
<213> artificial sequence <220>
<223> FR3 VHeavy region D25 <400> 63 agagtcacga ttaccgcgga cgaatccaca gacacagcct acatccatct gatcagcctg 60 agatctgagg acacggccat gtattactgt gcgacg 96 <210> 64 <211> 51 <212> DNA
<213> artificial sequence <220>
<223> CDR3 VHeavy region D25 <400> 64 gaaacagctc tggttgtatc tactacctac ctaccacact actttgacaa c 51 <210> 65 <211> 34 <212> DNA
<213> artificial sequence <220>
<223> FR4 VHeavy region D25 <400> 65 tggqgccagg gaaccctggt caccgtctcc tcag 34 <210> 66 <211> 69 <212> DNA
<213> artificial sequence <220>
<223> FR1 VLight region D25 <400> 66 gacatccaga tgacccagtc tccatcctcc ctgtctgcag ctgtaggaga cagagtcacc 60 atcacttgc 69 <210> 67 <211> 33 <212> DNA
<213> artificial sequence <220>
<223> CDR1 VLight region D25 <400> 67 caggcgagtc aggacattgt caactattta aat 33 <210> 68 <211> 45 <212> DNA
<213> artificial sequence <220>
<223> FR2 VLight region D25 <400> 68 tggtatcaac agaaaccagg gaaagcccct aagctcctga tctac 45 <210> 69 <211> 21 <212> DNA
<213> artificial sequence <220>
<223> CRD2 VLight region D25 <400> 69 gttgcatcca atttggagac a 21 <210> 70 <211> 96 <212> DNA
<213> artificial sequence <220>
<223> FR3 VLight region D25 <400> 70 ggggtcccat caaggttcag tggaagtgga tctgggacag attttagtct caccatcagc 60 agcctgcagc ctgaagatgt tgcaacatat tattgt 96 <210> 71 <211> 21 <212> DNA
<213> artificial sequence <220>
<223> CDR3 VLight region D25 <400> 71 caacaatatg ataatctccc a 21 <210> 72 <211> 39 <212> DNA
<213> artificial sequence <220>
<223> FR4 VLight region D25 <400> 72 ctcacattcg gcggagggac caaggttgag atcaaaaga 39 <210> 73 <211> 8 <212> PRT
<213> Homo sapiens <400> 73 Gly Phe Ser Phe Ser His Tyr Ala <210> 74 <211> 8 <212> PRT
<213> Homo sapiens <400> 74 Ile Ser Tyr Asp Gly Glu Asn Thr <210> 75 <211> 16 <212> PRT
<213> Homo sapiens <400> 75 Ala Arg Asp Arg Ile Val Asp Asp Tyr Tyr Tyr Tyr Gly Met Asp Val <210> 76 <211> 6 <212> PRT
<213> Homo sapiens <400> 76 Gin Asp Ile Lys Lys Tyr <210> 77 <211> 10 <212> PRT
<213> Homo sapiens <400> 77 Gin Gin Tyr Asp Asn Leu Pro Pro Leu Thr <210> 78 <211> 123 <212> PRT
<213> Homo sapiens <400> 78 Glu Val Gin Leu Val Glu Ser Gly Gly Gly Val Val Gin Pro Gly Arg Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Ser Phe Ser His Tyr Ala Met His Trp Val Arg Gin Ala Pro Gly Lys Gly Leu Glu Trp Val Ala Val Ile Ser Tyr Asp Gly Glu Asn Thr Tyr Tyr Ala Asp Ser Val Lys Gly Arg Phe Ser Ile Ser Arg Asp Asn Ser Lys Asn Thr Val Ser Leu Gin Met Asn Ser Leu Arg Pro Glu Asp Thr Ala Leu Tyr Tyr Cys Ala Arg Asp Arg Ile Val Asp Asp Tyr Tyr Tyr Tyr Gly Met Asp Val Trp Gly Gin Gly Ala Thr Val Thr Val Ser Ser <210> 79 <211> 111 <212> PRT
<213> Homo sapiens <400> 79 Asp Ile Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser Val Gly Asp Arg Val Thr Ile Thr Cys Gin Ala Ser Gin Asp Ile Lys Lys Tyr Leu Asn Trp Tyr His Gin Lys Pro Gly Lys Val Pro Glu Leu Leu Met His Asp Ala Ser Asn Leu Glu Thr Gly Val Pro Ser Arg Phe Ser Gly Arg Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Gin Pro Glu Asp Ile Gly Thr Tyr Tyr Cys Gin Gin Tyr Asp Asn Leu Pro Pro Leu Thr Phe Gly Gly Gly Thr Lys Val Glu Ile Lys Arg Thr Val <210> 80 <211> 8 <212> PRT
<213> Homo sapiens <400> 80 Gly Phe Thr Phe Ser Ser Tyr Asn <210> 81 <211> 8 <212> PRT
<213> Homo sapiens <400> 81 Ile Ser Ala Gly Ser Ser Tyr Ile <210> 82 <211> 18 <212> PRT
<213> Homo sapiens <400> 82 Ala Arg Glu Asp Tyr Gly Pro Gly Asn Tyr Tyr Ser Pro Asn Trp Phe Asp Pro <210> 83 <211> 9 <212> PRT
<213> Homo sapiens <400> 83 Ser Ser Asn Ile Gly Ala Gly Tyr Asp <210> 84 <211> 9 <212> PRT
<213> Homo sapiens <400> 84 His Ser Tyr Asp Arg Ser Leu Ser Gly <210> 85 <211> 125 <212> PRT
<213> Homo sapiens <400> 85 Glu Val Gin Leu Val Glu Thr Gly Gly Gly Leu Ala Gin Pro Gly Gly Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser Ser Tyr Asn Met Asn Trp Val Arg Gin Ala Pro Giy Lys Gly Leu Glu Trp Val Ser His Ile Ser Ala Gly Ser Ser Tyr Ile Tyr Tyr Ser Asp Ser Val Lys Gly Arg Phe Thr Val Ser Arg Asp Asn Val Arg Asn Ser Val Tyr Leu Gin Met Asn Ser Leu Arg Ala Ala Asp Thr Ala Val Tyr Tyr Cys Ala Arg Glu Asp Tyr Gly Pro Gly Asn Tyr Tyr Ser Pro Asn Trp Phe Asp Pro Trp Gly Gin Gly Thr Leu Val Thr Val Ser Ser <210> 86 <211> 110 <212> PRT
<213> Homo sapiens <400> 86 Gin Ser Val Val Thr Gin Pro Pro Ser Val Ser Gly Ala Pro Gly Gin Arg Val Thr Ile Ser Cys Thr Gly Ser Ser Ser Asn Ile Gly Ala Gly Tyr Asp Val His Trp Tyr Gin Gin Leu Pro Gly Thr Ala Pro Lys Leu Leu Ile Tyr Gly Asn Thr Asn Arg Pro Ser Gly Val Ser Asp Arg Phe Ser Gly Ser Lys Ser Gly Thr Ser Ala Ser Leu Ala Ile Thr Gly Leu Gin Ala Glu Asp Glu Ala Asp Tyr Tyr Cys His Ser Tyr Asp Arg Ser Leu Ser Gly Ser Val Phe Gly Gly Gly Thr Lys Leu Thr Val <210> 87 <211> 8 <212> PRT
<213> Homo sapiens <400> 87 Gly Phe Asn Phe His Asn Tyr Gly <210> 88 <211> 8 <212> PRT
<213> Homo sapiens <400> 88 Val Trp Tyr Asp Gly Ser Lys Lys <210> 89 <211> 13 <212> PRT
<213> Homo sapiens <400> 89 Val Arg Asp Lys Val Gly Pro Thr Pro Tyr Phe Asp Ser <210> 90 <211> 6 <212> PRT
<213> Homo sapiens <400> 90 Asn Ile Gly Ser Glu Thr <210> 91 <211> 11 <212> PRT
<213> Homo sapiens <400> 91 Gin Val Trp Asp Arg Ser Asn Tyr His Gin Val <210> 92 <211> 120 <212> PRT
<213> Homo sapiens <400> 92 Glu Val Gin Leu Val Glu Ser Gly Gly Asn Val Val Lys Pro Gly Thr Ser Leu Arg Leu Ser Cys Ala Ala Thr Gly Phe Asn Phe His Asn Tyr Gly Met Asn Trp Val Arg Gin Ala Pro Gly Lys Gly Leu Glu Trp Val Ala Val Val Trp Tyr Asp Gly Ser Lys Lys Tyr Tyr Ala Asp Ser Val Thr Gly Arg Phe Ala Ile Ser Arg Asp Asn Ser Lys Asn Thr Leu Tyr Leu Gin Met Asn Ser Leu Arg Val Glu Asp Thr Ala Val Tyr Tyr Cys Val Arg Asp Lys Val Gly Pro Thr Pro Tyr Phe Asp Ser Trp Gly Gin Gly Thr Leu Val Thr Val Ser Ser <210> 93 <211> 107 <212> PRT
<213> Homo sapiens <400> 93 Ser Tyr Val Leu Thr Gin Pro Pro Ser Val Ser Leu Ala Pro Gly Gly Thr Ala Ala Ile Thr Cys Gly Arg Asn Asn Ile Gly Ser Glu Thr Val His Trp Tyr Gin Gin Lys Pro Gly Gin Ala Pro Val Leu Val Val Tyr Asp Asp Asp Asp Arg Pro Ser Gly Ile Pro Glu Arg Phe Ser Gly Ser Asn Ser Gly Asn Thr Ala Thr Leu Thr Ile Ser Arg Val Glu Ala Gly Asp Glu Ala Asp Tyr Tyr Cys Gin Val Trp Asp Arg Ser Asn Tyr His Gin Val Phe Gly Gly Gly Thr Lys Leu Thr Val <210> 94 <211> 75 <212> DNA
<213> Homo sapiens <400> 94 gaggtgcagc tggtggagtc tgggggaggc gtggtccagc ctgggaggtc cctgagactc 60 tcctgtgcgg cctct 75 <210> 95 <211> 24 <212> DNA
<213> Homo sapiens <400> 95 ggattcagct tcagtcacta tgcc 24 <210> 96 <211> 51 <212> DNA
<213> Homo sapiens <400> 96 atgcactggg tccgccaggc tccaggcaag ggactggagt gggtggcagt t 51 <210> 97 <211> 24 <212> DNA
<213> Homo sapiens <400> 97 atatcttatg atggagaaaa taca 24 <210> 98 <211> 114 <212> DNA
<213> Homo sapiens <400> 98 tattacgcag actccgtgaa gggccgattc tccatctcca gagacaattc caagaacaca 60 gtgtctctgc aaatgaacag cctgagacct gaggacacgg ctctatatta ctgt 114 <210> 99 <211> 48 <212> DNA
<213> Homo sapiens <400> 99 gcgagagacc gcatagtgga cgactactac tactacggta tggacgtc 48 <210> 100 <211> 34 <212> DNA
<213> Homo sapiens <400> 100 tggggccaag gggccacggt caccgtctcc tcag 34 <210> 101 <211> 369 <212> DNA
<213> Homo sapiens <400> 101 gaggtgcagc tggtggagtc tgggggaggc gtggtccagc ctgggaggtc cctgagactc 60 tcctgtgcgg cctctggatt cagcttcagt cactatgcca tqcactgggt ccgccaggct 120 ccaggcaagg gactggagtg ggtggcagtt atatcttatg atggagaaaa tacatattac 180 gcagactccg tgaagggccg attctccatc tccagagaca attccaagaa cacagtgtct 240 ctgcaaatga acagcctqag acctgaggac acggctctat attactgtgc gagagaccgc 300 atagtggacg actactacta ctacggtatg gacgtctggg gccaaggggc cacggtcacc 360 gtctcctca 369 <210> 102 <211> 78 <212> DNA
<213> Homo sapiens <400> 102 gacatccaga tgacccagtc tccatcttcc ctgtctgcat ctgtaggaga cagagtcacc 60 atcacttgcc aggcgagt 76 <210> 103 <211> 18 <212> DNA
<213> Homo sapiens <400> 103 caggacatta agaagtat 18 <210> 104 <211> 51 <212> DNA
<213> Homo sapiens <400> 104 ttaaattggt atcatcagaa accagggaaa gtccctgagc tcctgatgca c 51 <210> 105 <211> 108 <212> DNA
<213> Homo sapiens <400> 105 aatttggaaa caggggtccc atcaaggttc agtggcaggg gatctgggac agattttact 60 ctcaccatta gcagcctgca gcctgaagat attggaacat attactgt 108 <210> 106 <211> 30 <212> DNA
<213> Homo sapiens <400> 106 caacagtatg ataatctgcc tccgctcact 30 <210> 107 <211> 31 <212> DNA
<213> Homo sapiens <400> 107 ttcggcggag ggaccaaggt ggagatcaaa c 31 <210> 108 <211> 333 <212> DNA
<213> Homo sapiens <400> 108 gacatccaga tgacccagtc tccatcttcc ctgtctgcat ctgtaggaga cagagtcacc 60 atcacttgcc aggcgagtca ggacattaag aagtatttaa attggtatca tcagaaacca 120 gggaaagtcc ctgagctcct gatgcacgat gcatccaatt tggaaacagg ggtcccatca 180 aggttcagtg gcaggggatc tgggacagat tttactctca ccattagcag cctgcagcct 240 gaagatattg gaacatatta ctgtcaacag tatgataatc tgcctccgct cactttcggc 300 ggagggacca aggtggagat caaacgaact gtg 333 <210> 109 <211> 75 <212> DNA
<213> Homo sapiens <400> 109 gaggtgcagc tggtggagac cgggggaggc ctggcccagc ctggggggtc cctgagactc 60 tcctgtgcag cctct 75 <210> 110 <211> 24 <212> DNA
<213> Homo sapiens <400> 110 ggattcacat tcagtagtta taac 24 <210> 111 <211> 51 <212> DNA
<213> Homo sapiens <400> 111 atgaactggg tccgccaggc tccagggaag gggctggagt gggtctcaca c 51 <210> 112 <211> 24 <212> DNA
<213> Homo sapiens <400> 112 attagtgcgg gtagtagtta cata 24 <210> 113 <211> 114 <212> DNA
<213> Homo sapiens <400> 113 tactactcag actcagtgaa gggccgattc accgtctcca gagacaacgt caggaactca 60 gtatatctgc aaatgaacag cctgagagcc gctgacacgg ctgtgtatta ctgt 114 <210> 114 <211> 54 <212> DNA
<213> Homo sapiens <400> 114 gcgagagagg attatggtcc gggaaattat tatagtccta actggttcga cccc 54 <210> 115 <211> 34 <212> DNA
<213> Homo sapiens <400> 115 tggggccagg gaaccctggt caccgtctcc tcag 34 <210> 116 <211> 375 <212> DNA
<213> Homo sapiens <400> 116 gaggtgcagc tggtggagac cgggggaggc ctggcccagc ctggggggtc cctgagactc 60 tcctgtgcag cctctggatt cacattcagt agttataaca tgaactgggt ccgccaggct 120 ccagggaagg ggctggagtg ggtctcacac attagtgcgg gtagtagtta catatactac 180 tcagactcag tgaagggccg attcaccgtc tccagagaca acgtcaggaa ctcagtatat 240 ctgcaaatga acagcctgag agccgctgac acggctgtgt attactgtgc gagagaggat 300 tatggtccgg gaaattatta tagtcctaac tggttcgacc cctgqggcca gggaaccctg 360 gtcaccgtct cctca 375 <210> 117 <211> 75 <212> DNA
<213> Homo sapiens <400> 117 cagtctgtcg tgacgcagcc gccctcagtg tctggggccc cagggcagag agtcaccatc 60 tcctgcactg ggagc 75 <210> 118 <211> 27 <212> DNA
<213> Homo sapiens <400> 118 agctccaaca tcggggcagg ttatgat 27 <210> 119 <211> 51 <212> DNA
<213> Homo sapiens <400> 119 gtacactggt accagcagct tccaggaaca gcccccaaac tcctcatcta t 51 <210> 120 <211> 108 <212> DNA
<213> Homo sapiens <400> 120 aatcggccct caggggtctc cgaccgattc tctggctcca agtctggcac ctcagcctcc 60 ctggccatca ctggactcca ggctgaggat gaggctgatt attactgc 108 <210> 121 <211> 27 <212> DNA
<213> Homo sapiens <400> 121 cactcctatg acagaagcct gagtggt 27 <210> 122 <211> 37 <212> DNA
<213> Homo sapiens <400> 122 tcagtattcg gcggagggac caagctgacc gtcctag 37 <210> 123 <211> 330 <212> DNA
<213> Homo sapiens <400> 123 cagtctgtcg tgacgcagcc gccctcagtg tctggggccc cagggcagag agtcaccatc 60 tcctgcactg ggagcagctc caacatcggg gcaggttatg atgtacactg gtaccagcag 120 cttccaggaa cagcccccaa actcctcatc tatggcaaca ctaatcggcc ctcaggggtc 180 tccgaccgat tctctggctc caagtctggc acctcagcct ccctggccat cactggactc 240 caggctgagg atgaggctga ttattactgc cactcctatg acagaagcct gagtggttca 300 gtattcggcg gagggaccaa gctgaccgtc 330 <210> 124 <211> 75 <212> DNA
<213> Homo sapiens <400> 124 caggtgcaac tggtggagtc tgggggaaat gtggtcaagc ctgggacgtc cctgagactg 60 tcctgtgcag cgact 75 <210> 125 <211> 24 <212> DNA
<213> Homo sapiens <400> 125 ggattcaact tccataacta cggc 24 <210> 126 <211> 51 <212> DNA
<213> Homo sapiens <400> 126 atgaactggg tccgccaggc tccaggcaag gggctggagt gggtggcggt t 51 <210> 127 <211> 24 <212> DNA
<213> Homo sapiens <400> 127 gtttggtatg atggaagtaa gaaa 24 <210> 128 <211> 114 <212> DNA
<213> Homo sapiens <400> 128 tactatgcag actccgtgac gggccgattc gccatctcca gagacaattc caagaacact 60 ctgtatctgc aaatgaacag cctgagagtc gaggacacgg ctgtttatta ttgt 114 <210> 129 <211> 39 <212> DNA
<213> Homo sapiens <400> 129 gtgagagata aagtgggacc gactccctac tttgactcc 39 <210> 130 <211> 34 <212> DNA
<213> Homo sapiens <400> 130 tggggccagg gaaccctggt caccgtatcc tcag 34 <210> 131 <211> 360 <212> DNA
<213> Homo sapiens <400> 131 gaggtgcagc tggtggagtc tgggggaaat gtggtcaagc ctgggacgtc cctgagactg 60 tcctgtgcag cgactggatt caacttccat aactacggca tgaactgggt ccgccaggct 120 ccaggcaagg ggctggagtg ggtggcggtt gtttggtatg atggaagtaa gaaatactat 180 gcagactccg tgacgggccg attcgccatc tccagagaca attccaagaa cactctgtat 240 ctgcaaatga acagcctgag agtcgaggac acggctgttt attattgtgt gagagataaa 300 gtgggaccga ctccctactt tgactcctgg ggccagggaa ccctggtcac cgtctcgagt 360 <210> 132 <211> 75 <212> DNA
<213> Homo sapiens <400> 132 tcctatgtgc tgactcagcc accctcggtg tcactggccc caggagggac ggccgcgatc 60 acctgtggaa gaaac 75 <210> 133 <211> 18 <212> DNA
<213> Homo sapiens <400> 133 aacattggaa gtgaaact 18 <210> 134 <211> 51 <212> DNA
<213> Homo sapiens <400> 134 gtgcactggt accagcagaa gccaggccag gcccctgtgc tggtcgtcta t 51 <210> 135 <211> 108 <212> DNA
<213> Homo sapiens <400> 135 gaccggccct cagggatccc tgagcgattc tctggctcca actctgggaa cacggccacc 60 ctgaccatca gcagggtcga ggccggggat gaggccgact attactgt 108 <210> 136 <211> 33 <212> DNA
<213> Homo sapiens <400> 136 caggtgtggg ataggagtaa ttatcatcag gta 33 <210> 137 <211> 31 <212> DNA
<213> Homo sapiens <400> 137 ttcggcggag ggaccaagtt gaccgtccta g 31 <210> 138 <211> 321 <212> DNA
<213> Homo sapiens <400> 138 tcctatgtgc tgactcagcc cccctcggtg tcactggccc caggagggac ggccgcgatc 60 acctgtggaa gaaacaacat tggaagtgaa actgtgcact ggtaccagca gaagccaggc 120 caggcccctg tgctggtcgt ctatgatgat gacgaccggc cctcagggat ccctgagcga 180 ttctctggct ccaactctgg gaacacggcc accctgacca tcagcagggt cgaggccggg 240 gatgaggccg actattactg tcaggtgtgg gataggagta attatcatca ggtattcggc 300 ggagggacca agctgaccgt c 321 <210> 139 <211> 378 <212> DNA
<213> Homo sapiens <400> 139 caggtgcagc tggtacagtc tggggctgaa gtgaagaagc ctgggtcctc ggtgatggtc 60 tcctgccagg cctctggagg ccccctcaga aactatatta tcaactggct acgacaggcc 120 cctggacaag gccctgagtg gatgggaggg atcattcctg tcttgggtac agtacactac 180 gcaccgaagt tccagggcag agtcacgatt accgcggacg aatccacgga cacagcctac 240 atccatctga tcagcctgag atctgaggac acggccatgt attactgtgc gacggaaaca 300 gctctggttg tatctactac ctacctacca cactactttg acaactgggg ccagggaacc 360 ctggtcaccg tctcctca 378 BOT
(69E)-(1) <ZZZ>
SO3 <TZZ>
<OZZ>
suaTdes owoH <ETZ>
VNO <ZTZ>
69E <ITZ>
ED'T <OTZ>
9ZE pe560b PeDqefrebbq bbeepouebb obbobbqqqo opbqoppobq oppuppboeg bpobupobqo pqougpoupp bbgboPbb-eb poo5pobqop beo5poqppo e6qopbpoqq. Dubopeobbo oqobbobpob bpbPqqqbbo obeopobqbp bbopeeebbq opeepbpoob b45opqoqpb gobqobpepo opobbeeob5 o opob?ebpob uoqe4664pe ubqopeqopp bqbogpoub5 poobeop5be pobqopeoqu opabqbbboo Pbobbbgboo boobqoqbqo obPo5Ppoop bebeopoeb4 ubepogeoeb ZD'T <OOP>
pozTmTqdo uopo3 qA SN#ASE <EZZ>
<OZZ>
epuenbos TPT0T-F4-le <ETZ>
VNO <ZTZ>
9E <TTZ>
ZVI <OTZ>
9ZE oeebee eeoqubpbqg bfreup3ebb5 2bbobbaii.4E. opoqouppog ogpeqebqpi. eeoepoqbqq. eqqegpopeo bqqbqpbpeb goo5Pobqop buo5Pogpoo eogoqbeTT4 qbpopbbbq pgebblbeeb bgbuoqqbbp epqeopoq.E.6 bbepubpbbq 44epDo4e3b qgboegogeb qopqobp4o poobeeebbb OZT
POOPPPbPOP poge46bqq-e ppqqq?goee oqbqqeoebb eo4bebobbu pobqqoeoge 002ogb2b20 252bbuqbqo beobqoqbqo op4DoTeDoq oqbuoD3pbq ubuoDqeoeb <0017>
suoTdes owoH <ETZ>
VNG <ZTZ>
9E <TTZ>
-CPT <OTZ>

gfrebogog bpoeblbbqo oppobbEceop 55554op2o2 boqqopqopo poobqDopqo peoouooqbq bbqbbqoop5 opub2BooPo obobgoegou gbgeopboou DebbPbobeb bobqopfreog 2b4DD-eo3Te OD'Z
oPqopboopo pboopobebe boubooboop oqppoeb4bb boobbbeopq 46Pupopoob 0eq0P061.50 opobbbgabg boopo4poge obbobbbgeb bgbpbqopob bbe3obEcepo OZT
pobbpobbob 4obbqoPPoq pogeopqoPe Mobqopooe bbobbpbpoo boobooq bqbb42b450 bpobeobboo oeeebe-ebqb beboobpbbo bubeobgb64 obPobqbbuo 017I <00>
pezTwTi.do uopo3 HA gZO#ASE <EZZ>
<OZZ>
aDuarlbs T2T0TJT43-e <ETZ>
VNO <zTZ>
BLE <FEZ>
OT <OTZ>

<400> 143 gag gtg cag ctg gtg gag tct ggg gga ggc gtg gtc cag cct ggg agg 48 Glu Val Gin Leu Val Glu Ser Gly Gly Gly Val Val Gin Pro Gly Arg tcc ctg aga ctc tcc tgt gcg gcc tct gga ttc agc ttc agt cac tat 96 Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Ser Phe Ser His Tyr gcc atg cac tgg gtc cgc cag gct cca ggc aag gga ctg gag tgg gtg 144 Ala Met His Trp Val Arg Gin Ala Pro Gly Lys Gly Leu Glu Trp Val gca gtt ata tct tat gat gga gaa aat aca tat tac gca gac tcc gtg 192 Ala Val Ile Ser Tyr Asp Gly Glu Asn Thr Tyr Tyr Ala Asp Ser Val aag ggc cga ttc tcc atc tcc aga gac aat tcc aag aac aca gtg tct 240 Lys Gly Arg Phe Ser Ile Ser Arg Asp Asn Ser Lys Asn Thr Val Ser ctg caa atg aac ago ctg aga cct gag gac acg got cta tat tac tgt 288 Leu Gin Met Asn Ser Leu Arg Pro Glu Asp Thr Ala Leu Tyr Tyr Cys gcg aga gac cgc ata gtg gac gac tac tac tac tac ggt atg gac gtc 336 Ala Arg Asp Arg Ile Val Asp Asp Tyr Tyr Tyr Tyr Gly Met Asp Val tgg ggc caa ggg gcc acg gtc acc gtc tcc tca 369 Trp Gly Gin Gly Ala Thr Val Thr Val Ser Ser <210> 144 <211> 123 <212> PRT
<213> Homo sapiens <400> 144 Glu Val Gin Leu Val Glu Ser Gly Gly Gly Val Val Gin Pre Gly Arg Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Ser Phe Ser His Tyr Ala Met His Trp Val Arg Gin Ala Pro Gly Lys Gly Leu Glu Trp Val Ala Val Ile Ser Tyr Asp Gly Glu Asn Thr Tyr Tyr Ala Asp Ser Val Lys Gly Arg Phe Ser Ile Ser Arg Asp Asn Ser Lys Asn Thr Val Ser Leu Gin Met Asn Ser Leu Arg Pro Glu Asp Thr Ala Lou Tyr Tyr Cys Ala Arg Asp Arg Ile Val Asp Asp Tyr Tyr Tyr Tyr Gly Met Asp Val Trp Gly Gin Gly Ala Thr Val Thr Val Ser Ser <210> 145 <211> 333 <212> DNA
<213> Homo sapiens <220>
<221> CDS
<222> (1)..(333) <400> 145 gac atc cag atg acc cag tct cca tct tcc ctg tct gca tct gta gga 48 Asp Ile Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser Val Gly gac aga gtc acc atc act tgc cag gcg agt cag gac att aag aag tat 96 Asp Arg Val Thr Ile Thr Cys Gin Ala Ser Gin Asp Ile Lys Lys Tyr tta aat tgg tat cat cag aaa cca ggg aaa gtc cct gag ctc ctg atg 144 Leu Asn Trp Tyr His Gin Lys Pro Gly Lys Val Pro Glu Leu Leu Met cac gat gca tcc aat ttg gaa aca ggg gtc cca tca agg ttc agt ggc 192 His Asp Ala Ser Asn Leu Glu Thr Gly Val Pro Ser Arg Phe Ser Gly agg gga tct ggg aca gat ttt act ctc acc att agc agc ctg cag cct 240 Arg Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Gin Pro gaa gat att gga aca tat tac tgt caa cag tat gat aat ctg cct ccg 288 Glu Asp Ile Gly Thr Tyr Tyr Cys Gin Gin Tyr Asp Asn Leu Pro Pro ctc act ttc ggc gga ggg acc aag gtg gag atc aaa cga act gtg 333 Leu Thr Phe Gly Gly Gly Thr Lys Val Glu Ile Lys Arg Thr Val <210> 146 <211> 111 <212> PRT
<213> Homo sapiens <400> 146 Asp Ile Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser Val Gly Asp Arg Val Thr Ile Thr Cys Gin Ala Ser Gin Asp Ile Lys Lys Tyr Leu Asn Trp Tyr His Gin Lys Pro Gly Lys Val Pro Glu Leu Leu Met His Asp Ala Ser Asn Leu Glu Thr Gly Val Pro Ser Arg Phe Ser Gly Arg Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Gin Pro Glu Asp Ile Gly Thr Tyr Tyr Cys Gin Gin Tyr Asp Asn Leu Pro Pro Leu Thr Phe Gly Gly Gly Thr Lys Val Glu Ile Lys Arg Thr Val <210> 147 <211> 98 <212> PRT
<213> artificial sequence <220>
<223> IGHV#-30 germl.
<400> 147 Gin Val Gin Leu Val Glu Ser Gly Gly Gly Val Val Gin Pro Gly Arg Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser Ser Tyr Gly Met His Trp Val Arg Gin Ala Pro Gly Lys Gly Leu Glu Trp Val Ala Val Ile Ser Tyr Asp Gly Ser Asn Lys Tyr Tyr Ala Asp Ser Val Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys Asn Thr Leu Tyr Leu Gin Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val Tyr Tyr Cys Ala Arg <210> 148 <211> 98 <212> PRT
<213> artificial sequence <220>
<223> IGKV1-33 germl.
<400> 148 Asp Ile Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser Val Gly Asp Arg Val Thr Ile Thr Cys Gln Ala Ser Gin Asp Ile Ser Asn Tyr Leu Asn Trp Tyr Gin Gin Lys Pro Gly Lys Ala Pro Lys Leu Leu Ile Tyr Asp Ala Ser Asn Leu Glu Thr Gly Val Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Phe Thr Ile Ser Ser Leu Gin Pro Glu Asp Ile Ala Thr Tyr Tyr Cys Gin Gin Tyr Asp Asn Leu Pro Pro Leu Thr <210> 149 <211> 375 <212> DNA
<213> Homo sapiens <220>
<221> CDS
<222> (1)..(375) <400> 149 gag gtg cag ctg gtg gag ace ggg gga ggc ctg gcc cag cct ggg ggg 48 Glu Val Gin Leu Val Glu Thr Gly Gly Gly Leu Ala Gin Pro Gly Gly tcc ctg aga ctc tcc tgt gca gcc tct gga ttc aca ttc agt agt tat 96 Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser Ser Tyr aac atg aac tgg gtc cgc cag gct cca ggg aag ggg ctg gag tgg gtc 144 Asn Met Asn Trp Val Arg Gin Ala Pro Gly Lys Gly Leu Glu Trp Val tca cac att agt gcg ggt agt agt tac ata tac tac tca gac tca gtg 192 Ser His Ile Ser Ala Gly Ser Ser Tyr Ile Tyr Tyr Ser Asp Ser Val aag ggc cga ttc acc gtc tcc aga gac aac gtc agg aac tca gta tat 240 Lys Gly Arg Phe Thr Val Ser Arg Asp Asn Val Arg Asn Ser Val Tyr ctg caa atg aac agc ctg aga gcc gct gac acg gct gtg tat tat tgt 288 Leu Gin Met Asn Ser Leu Arg Ala Ala Asp Thr Ala Val Tyr Tyr Cys gcg aga gag gat tat ggt ccg gga aat tat tat agt cct aac tgg ttc 336 Ala Arg Glu Asp Tyr Gly Pro Gly Asn Tyr Tyr Ser Pro Asn Trp Phe gac ccc tgg ggc cag gga acc ctg gtc acc gtc tcc tca 375 Asp Pro Trp Gly Gin Gly Thr Leu Val Thr Val Ser Ser <210> 150 <211> 125 <212> PRT
<213> Homo sapiens <400> 150 Glu Val Gin Leu Val Glu Thr Gly Gly Gly Leu Ala Gin Pro Gly Gly Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser Ser Tyr Asn Met Asn Trp Val Arg Gin Ala Pro Gly Lys Gly Leu Glu Trp Val Ser His Ile Ser Ala Gly Ser Ser Tyr Ile Tyr Tyr Ser Asp Ser Val Lys Gly Arg Phe Thr Val Ser Arg Asp Asn Val Arg Asn Ser Val Tyr Leu Gin Met Asn Ser Leu Arg Ala Ala Asp Thr Ala Val Tyr Tyr Cys Ala Arg Glu Asp Tyr Gly Pro Gly Asn Tyr Tyr Ser Pro Asn Trp Phe Asp Pro Trp Gly Gin Gly Thr Leu Val Thr Val Ser Ser <210> 151 <211> 330 <212> DNA
<213> Homo sapiens <220>
<221> CDS
<222> (1)..(330) <400> 151 cag tct gtc gtg acg cag ccg ccc tca gtg tct ggg gcc cca ggg cag 48 Gin Ser Val Val Thr Gin Pro Pro Ser Val Ser Gly Ala Pro Gly Gin aga gtc acc atc tcc tgc act ggg agc agc tcc aac atc ggg gca ggt 96 Arg Val Thr Ile Ser Cys Thr Gly Ser Ser Ser Asn Ile Gly Ala Gly tat gat gta cac tgg tac cag cag ctt cca gga aca gcc ccc aaa ctc 144 Tyr Asp Val His Trp Tyr Gin Gin Leu Pro Gly Thr Ala Pro Lys Leu ctc atc tat ggc aac act aat cgg ccc tca ggg gtc tcc gac cga ttc 192 Leu Ile Tyr Gly Asn Thr Asn Arg Pro Ser Gly Val Ser Asp Arg Phe tct ggc tcc aag tct ggc acc tca gcc tcc ctg gcc atc act gga ctc 240 Ser Gly Ser Lys Ser Gly Thr Ser Ala Ser Leu Ala Ile Thr Gly Leu cag gct gag gat gag gct gat tat tac tgc cac tcc tat gac aga agc 288 Gin Ala Glu Asp Glu Ala Asp Tyr Tyr Cys His Ser Tyr Asp Arg Her ctg agt ggt tca gta ttc ggc gga ggg acc aag ctg acc gtc 330 Lou Ser Gly Ser Val Phe Gly Gly Gly Thr Lys Leu Thr Val <210> 152 <211> 110 <212> PRT
<213> Homo sapiens <400> 152 Gin Ser Val Val Thr Gin Pro Pro Ser Val Ser Gly Ala Pro Gly Gln Arg Val Thr Ile Ser Cys Thr Gly Ser Ser Her Asn Ile Gly Ala Gly Tyr Asp Val His Trp Tyr Gin Gin Leu Pro Gly Thr Ala Pro Lys Leu Leu Ile Tyr Gly Asn Thr Asn Arg Pro Ser Gly Val Ser Asp Arg Phe Ser Gly Ser Lys Ser Gly Thr Ser Ala Ser Leu Ala Ile Thr Gly Leu Gln Ala Glu Asp Glu Ala Asp Tyr Tyr Cys His Ser Tyr Asp Arg Ser Leu Ser Gly Ser Val Phe Gly Gly Gly Thr Lys Leu Thr Val <210> 153 <211> 73 <212> PRT
<213> artificial sequence <220>
<223> IGHV3-21 germl.
<400> 153 Gly Phe Thr Phe Ser Ser Tyr Ser Met Asn Trp Val Arg Gin Ala Pro Gly Lys Gly Leu Glu Trp Val Ser Ser Ile Ser Ser Ser Ser Ser Tyr Ile Tyr Tyr Ala Asp Ser Val Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ala Lys Asn Ser Leu Tyr Leu Gin Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val Tyr Tyr Cys Ala Arg <210> 154 <211> 99 <212> PRT
<213> artificial sequence <220>
<223> IGLV1-40 germl.
<400> 154 Gin Ser Val Val Thr Gin Pro Pro Ser Val Ser Gly Ala Pro Gly Gin Arg Val Thr Ile Ser Cys Thr Gly Ser Ser Ser Asn Ile Gly Ala Gly Tyr Asp Val His Trp Tyr Gin Gin Leu Pro Gly Thr Ala Pro Lys Leu Leu Ile Tyr Gly Asn Ser Asn Arg Pro Ser Gly Val Pro Asp Arg Phe Ser Gly Ser Lys Ser Gly Thr Ser Ala Ser Leu Ala Ile Thr Gly Leu Gin Ala Glu Asp Glu Ala Asp Tyr Tyr Cys Gin Ser Tyr Asp Ser Ser Leu Ser Gly <210> 155 <211> 360 <212> DNA
<213> Homo sapiens <220>
<221> CDS
<222> (1)..(360) <400> 155 gag gtg cag ctg gtg gag tot ggg gga aat gtg gtc aag cot ggg acg 48 Glu Val Gin Leu Val Glu Ser Gly Gly Asn Val Val Lys Pro Gly Thr tcc ctg aga ctg tcc tgt gca gcg act gga ttc aac ttc cat aac tac 96 Ser Leu Arg Leu Ser Cys Ala Ala Thr Gly Phe Asn Phe His Asn Tyr ggc atg aac tgg gtc cgc cag got cca ggc aag ggg ctg gag tgg gtg 144 Gly Met Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val gcg gtt gtt tgg tat gat gga agt aag aaa tac tat gca gac tcc gtg 192 Ala Vol Val Trp Tyr Asp Gly Ser Lys Lys Tyr Tyr Ala Asp Ser Val acg ggc cga ttc gcc atc tcc aga gac aat tcc aag aac act ctg tat 240 Thr Gly Arg Phe Ala Ile Ser Arg Asp Asn Ser Lys Asn Thr Leu Tyr ctg caa atg aac ago ctg aga gtc gag gac acg got gtt tat tat tgt 288 Leu Gin Met Asn Ser Leu Arg Val Glu Asp Thr Ala Vol Tyr Tyr Cys gtg aga gat aaa gtg gga ccg act ccc tac ttt gac tcc tgg ggc cag 336 Vol Arg Asp Lys Vol Gly Pro Thr Pro Tyr Phe Asp Ser Trp Gly Gin gga acc ctg gtc acc gtc tog agt 360 Gly Thr Leu Val Thr Val Ser Ser <210> 156 <211> 120 <212> PRT
<213> Homo sapiens <400> 156 Glu Vol Gin Leu Val Glu Ser Gly Gly Asn Val Val Lys Pro Gly Thr Ser Leu Arg Leu Ser Cys Ala Ala Thr Gly Phe Asn Phe His Asn Tyr Gly Met Asn Trp Val Arg Gin Ala Pro Gly Lys Gly Leu Glu Trp Val Ala Vol Val Trp Tyr Asp Gly Ser Lys Lys Tyr Tyr Ala Asp Ser Vol Thr Gly Arg Phe Ala Ile Ser Arg Asp Asn Ser Lys Asn Thr Leu Tyr Leu Gin Met Asn Ser Leu Arg Val Glu Asp Thr Ala Val Tyr Tyr Cys Val Arg Asp Lys Val Gly Pro Thr Pro Tyr Phe Asp Ser Trp Gly Gin Gly Thr Leu Val Thr Val Ser Ser <210> 157 <211> 321 <212> DNA
<213> Homo sapiens <220>
<221> CDS
<222> (1)..(321) <400> 157 too tat gtg ctg act cag ccc ccc tcg gtg tca ctg gcc cca gga ggg 48 Ser Tyr Val Leu Thr Gln Pro Pro Ser Val Ser Leu Ala Pro Gly Gly acg gcc gcg atc acc tgt gga aga aac aac att gga agt gaa act gtg 96 Thr Ala Ala Ile Thr Cys Gly Arg Asn Asn Ile Gly Ser Glu Thr Val cac tgg tac cag cag aag cca ggc cag gcc cct gtg ctg gtc gtc tat 144 His Trp Tyr Gin Gin Lys Pro Gly Gin Ala Pro Val Leu Val Val Tyr gat gat gac gac cgg ccc tca ggg atc cct gag cga ttc tct ggc tcc 192 Asp Asp Asp Asp Arg Pro Ser Gly Ile Pro Glu Arg Phe Ser Gly Ser aac tct ggg aac acg gcc acc ctg acc atc agc agg gtc gag gcc ggg 240 Asn Ser Gly Asn Thr Ala Thr Leu Thr Ile Ser Arg Val Glu Ala Gly gat gag gcc gac tat tac tgt cag gtg tgg gat agg agt aat tat cat 288 Asp Glu Ala Asp Tyr Tyr Cys Gin Val Trp Asp Arg Ser Asn Tyr His cag gta ttc ggc gga ggg acc aag ctg acc gtc 321 Gin Val Phe Gly Gly Gly Thr Lys Leu Thr Val <210> 158 <211> 107 <212> PRT
<213> Homo sapiens <400> 158 Ser Tyr Val Leu Thr Gin Pro Pro Ser Val Ser Leu Ala Pro Gly Gly Thr Ala Ala Ile Thr Cys Gly Arg Asn Asn Ile Gly Ser Glu Thr Val His Trp Tyr Gin Gin Lys Pro Gly Gin Ala Pro Val Leu Val Val Tyr Asp Asp Asp Asp Arg Pro Ser Gly Ile Pro Glu Arg Phe Ser Gly Ser Asn Ser Gly Asn Thr Ala Thr Leu Thr Ile Ser Arg Val Glu Ala Gly Asp Glu Ala Asp Tyr Tyr Cys Gin Val Trp Asp Arg Ser Asn Tyr His Gin Val Phe Gly Gly Gly Thr Lys Leu Thr Val <210> 159 <211> 98 <212> PRT
<213> artificial sequence <220>
<223> IGHV3-33 germl.
<400> 159 Gin Vol Gin Leu Vol Glu Ser Gly Gly Gly Val Vol Gin Pro Gly Arg Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser Ser Tyr Gly Met His Trp Vol Arg Gin Ala Pro Gly Lys Gly Leu Glu Trp Val Ala Val Ile Trp Tyr Asp Gly Ser Asn Lys Tyr Tyr Ala Asp Ser Vol Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys Asn Thr Leu Tyr Leu Gin Met Asn Ser Leu Arg Ala Giu Asp Thr Ala Val Tyr Tyr Cys Ala Arg <210> 160 <211> 98 <212> PRT
<213> artificial sequence <220>
<223> IGLV3-21 germl.
<400> 160 Ser Tyr Vol Leu Thr Gin Pro Pro Ser Vol Ser Vol Ala Pro Gly Gln Thr Ala Arg Ile Thr Cys Gly Gly Asn Asn Ile Gly Ser Lys Ser Vol His Trp Tyr Gin Gin Lys Pro Gly Gin Ala Pro Val Leu Val Val Tyr , Asp Asp Ser Asp Arg Pro Ser Gly Ile Pro Glu Arg Phe Ser Gly Ser Asn Ser Gly Asn Thr Ala Thr Leu Thr Ile Ser Arg Val Glu Ala Gly Asp Glu Ala Asp Tyr Tyr Cys Gln Val Trp Asp Ser Ser Ser Asp His Gin Val

Claims (35)

Claims
1. An isolated, synthetic or recombinant antibody or a functional part, derivative and/or analogue thereof which is capable of specifically binding Respiratory Syncytial Virus and which comprises.
- a heavy chain CDR1 sequence comprising a sequence which is at least 70%
identical to the sequence NYIIN, and/or - a heavy chain CDR2 sequence comprising a sequence which is at least 75%
identical to the sequence GIIPVLGTVHYAPKFQG, and/or - a heavy chain CDR3 sequence comprising a sequence which is at least 70%
identical to the sequence ETALVVSTTYLPHYFDN, and/or - a light chain CDR1 sequence comprising a sequence which is at least 85%
identical to the sequence QASQDIVNYLN, and/or - a light chain CDR2 sequence comprising a sequence which is at least 70%
identical to the sequence VASNLET.
2. An antibody, functional part, derivative or analogue according to claim 1, having a heavy chain sequence comprising a sequence which is at least 70%
identical to the sequence QVQLVQSGAEVKKPGSSVMVSCQASGGPLRNYIINWLRQAPGQGPEWMG
GIIPVLGTVHYAPKFQGRVTITADESTDTAYIHLISLRSEDTAMYYCATETA
LVVSTTYLPHYFDN WGQGTLVTVSS and/or having a light chain sequence which is at least 70% identical to the sequence DIQMTQSPSSLSAAVGDRVTITCQASQDIVNYLNWYQQKPGKAPKLLIYVA
SNLETGVPSRFSGSGSGTDFSLTISSLQPEDVATYYCQQYDNLPLTFGGGT
KVEIKRTV.
3. An antibody which has an IC50 value of less than 10 ng/ml, preferably less than 2 ng/ml, in an in vitro neutralization assay wherein HEp-2 cells are infected with RSV, or a functional part, derivative and/or analogue of said antibody.
4. An isolated, synthetic or recombinant antibody or a functional part, derivative and/or analogue thereof which is capable of specifically binding Respiratory Syncytial Virus and which comprises:
- a heavy chain CDR1 sequence comprising a sequence which is at least 80%
identical to the sequence GFSFSHYA, and/or - a heavy chain CDR2 sequence comprising a sequence which is at least 80%
identical to the sequence ISYDGENT, and/or - a heavy chain CDR3 sequence comprising a sequence which is at least 80%
identical to the sequence ARDRIVDDYYYYGMDV, and/or - a light chain CDR1 sequence comprising a sequence which is at least 80%
identical to the sequence QDIKKY, and/or - a light chain CDR2 sequence comprising a sequence which is at least 80%
identical to the sequence DAS, and/or - a light chain CDR3 sequence comprising a sequence which is at least 80%
identical to the sequence QQYDNLPPLT.
5. An antibody, functional part, derivative or analogue according to claim 4, having a heavy chain sequence comprising a sequence which is at least 70%
identical to the sequence EVQLVESGGGVVQPGRSLRLSCAASGFSFSHYAMHWVRQAPGKGLEWVA
VISYDGENTYYADSVKGRFSISRDNSKNTVSLQMNSLRPEDTALYYCARD
RIVDDYYYYGMDVWGQGATVTVSS and/or having a light chain sequence which is at least 70% identical to the sequence DIQMTQSPSSLSASVGDRVTITCQASQDIKKYLNWYHQKPGKVPELLMHDASNLETGVPS
RFSGRGSGTDFTLTISSLQPEDIGTYYCQQYDNLPPLTFGGGTKVEIKRTV.
6. An isolated, synthetic or recombinant antibody or a functional part, derivative and/or analogue thereof which is capable of specifically binding Respiratory Syncytial Virus and which comprises:
- a heavy chain CDR1 sequence comprising a sequence which is at least 80%
identical to the sequence GYITSSYN, and/or - a heavy chain CDR2 sequence comprising a sequence which is at least 80%
identical to the sequence ISAGSSYI, and/or - a heavy chain CDR3 sequence comprising a sequence which is at least 80%
identical to the sequence AREDYGPGNYYSPNWFDP, and/or - a light chain CDR1 sequence comprising a sequence which is at least 80% =
identical to the sequence SSNIGAGYD, and/or - a light chain CDR2 sequence comprising a sequence which is at least 80%
identical to the sequence GNT, and/or - a light chain CDR3 sequence comprising a sequence which is at least 80%
identical to the sequence HSYDRSLSG.
7. An antibody, functional part, derivative or analogue according to claim 6, having a heavy chain sequence comprising a sequence which is at least 70%
identical to the sequence EVQLVETGGGLAQPGGSLRLSCAASGFTFSSYNMNWVRQAPGKGLEWVSHISAGSSYIYY
SDSVKGRFTVSRDNVRNSVYLQMNSLRAADTAVYYCAREDYGPGNYYSPNWFDPWGQGTL
VTVSS and/or having a light chain sequence which is at least 70% identical to the sequence QSVVTQPPSVSGAPGQRVTISCTGSSSNIGAGYDVHWYQQLPGTAPKLLIYGNTNRPSGV
SDRFSGSKSGTSASLAITGLQAEDEADYYCHSYDRSLSGSVFGGGTKLTV.
8. An isolated, synthetic or recombinant antibody or a functional part, derivative and/or analogue thereof which is capable of specifically binding Respiratory Syncytial Virus and which comprises:
- a heavy chain CDR1 sequence comprising a sequence which is at least 80%
identical to the sequence GFNFHNYG, and/or - a heavy chain CDR2 sequence comprising a sequence which is at least 80%
identical to the sequence VWYDGSKK, and/or - a heavy chain CDR3 sequence comprising a sequence which is at least 80%
identical to the sequence VRDKVGPTPYFDS, and/or - a light chain CDR1 sequence comprising a sequence which is at least 80%
identical to the sequence NIGSET, and/or - a light chain CDR2 sequence comprising a sequence which is at least 80%
identical to the sequence DDD, and/or - a light chain CDR3 sequence comprising a sequence which is at least 80%
identical to the sequence QVWDRSNYHQV.
9. An antibody, functional part, derivative or analogue according to claim 6, having a heavy chain sequence comprising a sequence which is at least 70%
identical to the sequence EVQLVESGGNVVKPGTSLRLSCAATGFNFHNYGMNWVRQAPGKGLEWVAVVWYDGSKKYY
ADSVTGRFAISRDNSKNTLYLQMNSLRVEDTAVYYCVRDKVGPTPYFDSWGQGTLVTVSS
and/or having a light chain sequence which is at least 70% identical to the sequence SYVLTQPPSVSLAPGGTAAITCGRNNIGSETVHWYQQKPGQAPVLVVYDDDDRPSGIPER
FSGSNSGNTATLTISRVEAGDEADYYCQVWDRSNYHQVFGGGTKLTV.
10. An antibody, functional part, derivative or analogue according to any one of claims 1-9, which is a human antibody and/or a chimeric antibody.
11 An isolated, synthetic or recombinant nucleic acid sequence, or a functional part, derivative or analogue thereof, encoding an antibody, functional part, derivative or analogue according to any one of claims 1-10
12 An antibody, functional part, derivative or analogue according to any one of claims 1-10, for use as a medicament and/or prophylactic agent.
13 Use of an antibody, functional part, derivative or analogue according to any one of claims 1-10 for the preparation of a medicament and/or prophylactic agent for at least in part treating and/or preventing a RSV-related disorder
14 A nucleic acid sequence, functional part, derivative and/or analogue according to claim 11 for use as a medicament and/or prophylactic agent.
15 Use of a nucleic acid sequence, functional part, derivative and/or analogue according to claim 11 for the preparation of a medicament and/or prophylactic agent for at least in part treating and/or preventing a RSV-related disorder
16 An isolated antibody producing cell capable of producing an antibody, functional part, derivative or analogue according to any one of claims 1-10
17. An antibody producing cell according to claim 16, which is stable for at least nine weeks, preferably for at least three months, more preferably for at least six months.
18 An antibody producing cell according to claim 16 or 17, wherein BCL6 and Blimp-1 are co-expressed.
19 An antibody producing cell according to any one of claims 16-18, comprising:
- an exogenous nucleic acid sequence encoding BCL6 or a functional part, derivative and/or analogue thereof, and/or - an exogenous nucleic acid sequence encoding Bcl-xL or a functional part, derivative and/or analogue thereof.
20 An antibody producing cell according to any one of claims 16-19, wherein expression of a nucleic acid sequence encoding BCL6, Bcl-xL or a functional part, derivative and/or analogue of BCL6 or Bcl-xL, is regulated by an activator and/or repressor that is inducible by an exogenous compound.
21. A method for producing an antibody producing cell, which is stable for at least three months and which is capable of producing RSV-specific antibodies, the method comprising:
- increasing an expression level of Blimp-1 in a B cell which is capable of producing RSV-specific antibodies; and - increasing and/or maintaining a BCL6 expression level in said B cell.
22 A method according to claim 21, wherein the BCL6 expression level in said B cell is brought to, and/or maintained at, essentially the same level or at a higher level as compared to a plasmablast.
23 A method according to claim 21 or 22, comprising providing said B cell with a nucleic acid sequence encoding BCL6 or a functional part, derivative and/or analogue thereof
24 A method according to any one of claims 21-23, further comprising increasing an expression level of Bcl-xL in said cell.
25. A method according to claim 24, comprising providing said cell with a nucleic acid encoding Bcl-xL or a functional part, derivative and/or analogue thereof.
26. A method according to any one of claims 21-25, comprising.
- providing a B cell capable of producing RSV-specific antibodies with BCL6, or a functional part, derivative and/or analogue thereof;
- providing said B cell with Bcl-xL or a functional part, derivative and/or analogue thereof; and - culturing said B cell
27. A method according to any one of claims 21-26, wherein said B cell is cultured in the presence of IL-21
28. A method according to any one of claims 21-27, wherein said B cell has been obtained from an individual, which individual had been previously exposed to Respiratory Syncytial Virus
29 A method according to any one of claims 21-28, further comprising expressing a gene of said B cell encoding the Ig heavy chain and/or Ig light chain in a second cell.
30. A method for producing antibodies which are capable of specifically binding Respiratory Syncytial Virus, the method comprising:
- producing an antibody producing cell capable of producing RSV-specific antibodies with a method according to any one of claims 21-29; and - obtaining antibodies produced by said antibody producing cell.
31 An isolated antibody obtainable by a method according to claim 30, or a functional part, derivative and/or analogue of said antibody.
32 . An isolated, synthetic or recombinant nucleic acid sequence comprising a sequence which is at least 70% homologous to a least part of a nucleotide sequence as depicted in Figure 11, Figure 12, Figure 14A, Figure 14B or Figure 14C, said part having at least 15 nucleotides
33. A nucleic acid sequence according to claim 32, wherein said nucleic acid sequence comprises a sequence which is at least 70% homologous to the sequenceACTATATTATCAAC, GGGATCATTCCTGTCTTGGGTACAGTACACTACGCACCGAAGTTCCAGGGC, GAAACAGCTCTGGTTGTATCTACTACCTACCTACCACACTACTTTGACAAC, CAGGCGAGTCAGGACATTGTCAACTATTTAAAT, GTTGCATCCAATTTGGAGACA, CAACAATATGATAATCTCCCA, GGATTCAGCTTCAGTCACTATGCC, ATATCTTATGATGGAGAAAATACA, GCGAGAGACCGCATAGTGGACGACTACTACTACTACGGTATGGACGTC, CAGGACATTAAGAAGTAT, GATGCATCC, CAACAGTATGATAATCTGCCTCCGCTCACT, GGATTCACATTCAGTAGTTATAAC, ATTAGTGCGGGTAGTAGTTACATA, GCGAGAGAGGATTATGGTCCGGGAAATTATTATAGTCCTAACTGGTTCGACCCC, AGCTCCAACATCGGGGCAGGTTATGAT, GGCAACACT, CACTCCTATGACAGAAGCCTGAGTGGT, GGATTCAACTTCCATAACTACGGC, GTTTGGTATGATGGAAGTAAGAAA, GTGAGAGATAAAGTGGGACCGACTCCCTACTTTGACTCC, AACATTGGAAGTGAAACT, GATGATGAC and/or CAGGTGTGGGATAGGAGTAATTATCATCAGGTA.
34. An isolated, synthetic or recombinant nucleic acid sequence comprising a sequence encoding an amino acid sequence which is at least 70% identical to the sequence NYIIN, and/or at least 75% identical to the sequence GIIPVLGTVHYAPKFQG, andlor at least 70% identical to the sequence ETALVVSTTYLPHYFDN, and/or at least 85% identical to the sequence QASQDIVNYLN, and/or at least 70% identical to the sequence VASNLET, and/or at least 70% identical to the sequence QVQLVQSGAEVKKPGSSVMVSCQASGGPLRNYIINWLRQAPGQGPEWMG
GIIPVLGTVHYAPKFQGRVTITADESTDTAYIHLISLRSEDTAMYYCATETA
LVVSTTYLPHYFDN WGQGTLVTVSS, and/or at least 70% identical to the sequence DIQMTQSPSSLSAAVGDRVTITCQASQDIVNYLNWYQQKPGKAPKLLIYVA
SNLETGVPSRFSGSGSGTDFSLTISSLQPEDVATYYCQQYDNLPLTFGGGT
KVEIKRTV and/or at least 70% identical to a sequence selected from the group consisting of: GFSFSHYA, ISYDGENT, ARDRIVDDYYYYGMDV, QDIKKY, DAS, QQYDNLPPLT, EVQLVESGGGVVQPGRSLRLSCAASGFSFSHYAMHWVRQAPGKGLEWVA
VISYDGENTYYADSVKGRFSISRDNSKNTVSLQMNSLRPEDTALYYCARD

RIVDDYYYYGMDVWGQGATVTVSS, DIQMTQSPSSLSASVGDRVTITCQASQDIKKYLNWYHQKPGKVPELLMHDASNLETGVPS
RFSGRGSGTDFTLTISSLQFEDIGTYYCQQYDNLPPLTFGGGTKVEIKRTV, GFTFSSYN,ISAGSSYI, AREDYGPGNYYSPNWFDP, SSNIGAGYD, GNT, HSYDRSLSG, EVQLVETGGGLAQPGGSLRLSCAASGFTFSSYNMNWVRQAPGKGLEWVSHISAGSSYIYY
SDSVKGRFTVSRDNVRNSVYLQMNSLRAADTAVYYCAREDYGPGNYYSPNWFDPWGQGTL
VTVSS , QSVVTQPPSVSGAPGQRVTISCTGSSSNIGAGYDVHWYQQLPGTAPKLLIYGNTNRPSQV
SDRFSGSKSGTSASLAITGLQAEDEADYYCHSYDRSLSGSVFGGGTKLTV, GFNFHNYG, VWYDGSKK, VRDKVGPTPYFDS, NIGSET, DDD, QVWDRSNYHQV, EVQLVESGGNVVKPGTSLRLSCAATGFNFHNYGMNWVRQAPGKGLEWVAVVWYDGSKKYY
ADSVTGRFAISRDNSKNTLYLQMNSLRVEDTAVYYCVRDKVGPTPYFDSWGQGTLVTVSS
, and SYVLTQPPSVSLAPGGTAAITCGRNNIGSETVHWYQQKPGQAPVLWYDDDDRPSGIPER
FSGSNSGNTATLTISRVEAGDEADYYCQVWDRSNYHQVFGGGTKLTV
35. A method for at least in part treating or preventing an RSV-related disorder, the method comprising administering to an individual in need thereof a therapeutically effective amount of an antibody or functional part, derivative or analogue according to any one of claims 1-10 or 31.
CA2950515A 2007-06-01 2008-05-30 Rsv specific binding molecules and means for producing them Abandoned CA2950515A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP07109472A EP1997830A1 (en) 2007-06-01 2007-06-01 RSV specific binding molecules and means for producing them
EP07109472.6 2007-06-01
CA2689290A CA2689290C (en) 2007-06-01 2008-05-30 Rsv-specific binding molecules and means for producing them

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CA2689290A Division CA2689290C (en) 2007-06-01 2008-05-30 Rsv-specific binding molecules and means for producing them

Publications (1)

Publication Number Publication Date
CA2950515A1 true CA2950515A1 (en) 2008-12-04

Family

ID=38578633

Family Applications (2)

Application Number Title Priority Date Filing Date
CA2689290A Active CA2689290C (en) 2007-06-01 2008-05-30 Rsv-specific binding molecules and means for producing them
CA2950515A Abandoned CA2950515A1 (en) 2007-06-01 2008-05-30 Rsv specific binding molecules and means for producing them

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CA2689290A Active CA2689290C (en) 2007-06-01 2008-05-30 Rsv-specific binding molecules and means for producing them

Country Status (23)

Country Link
US (5) US8562996B2 (en)
EP (3) EP1997830A1 (en)
JP (4) JP5450395B2 (en)
KR (2) KR101599556B1 (en)
CN (2) CN101778866B (en)
AU (1) AU2008257801B8 (en)
BR (1) BRPI0812878C1 (en)
CA (2) CA2689290C (en)
CY (1) CY1117767T1 (en)
DK (1) DK2170952T3 (en)
ES (1) ES2575129T3 (en)
HK (1) HK1146073A1 (en)
HR (1) HRP20160632T1 (en)
HU (1) HUE029225T2 (en)
IL (1) IL202288A (en)
MX (2) MX350375B (en)
NZ (3) NZ581512A (en)
PL (1) PL2170952T3 (en)
PT (1) PT2170952E (en)
RU (2) RU2527067C2 (en)
SI (1) SI2170952T1 (en)
WO (1) WO2008147196A2 (en)
ZA (1) ZA200908320B (en)

Families Citing this family (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2357187A1 (en) 2000-12-12 2011-08-17 MedImmune, LLC Molecules with extended half-lives, compositions and uses thereof
US9127251B2 (en) 2005-12-09 2015-09-08 Academisch Medisch Centrum Bij De Universiteit Van Amsterdam Means and methods for influencing the stability of antibody producing cells
US9005974B2 (en) 2005-12-09 2015-04-14 Academish Medisch Centrum Bij De Universiteit Van Amsterdam Means and methods for influencing the stability of cells
EP1997830A1 (en) 2007-06-01 2008-12-03 AIMM Therapeutics B.V. RSV specific binding molecules and means for producing them
BRPI0817257A2 (en) 2007-09-24 2015-06-16 Univ Vanderbilt Monoclonal antibodies to respiratory syncytial virus and their uses
WO2010068722A1 (en) 2008-12-12 2010-06-17 Medimmune, Llc Crystals and structure of a human igg fc variant with enhanced fcrn binding
AU2015200208B2 (en) * 2009-07-15 2016-10-20 Kling Biotherapeutics B.V. Means and methods for producing high affinity antibodies
JP5781508B2 (en) * 2009-07-15 2015-09-24 アイム・セラピューティクス・べー・フェー Means and methods for producing high affinity antibodies
CN104628850B (en) * 2009-10-06 2020-07-28 医学免疫有限公司 RSV-specific binding molecules
US8568726B2 (en) 2009-10-06 2013-10-29 Medimmune Limited RSV specific binding molecule
ES2621458T3 (en) * 2009-10-06 2017-07-04 Medimmune Limited RSV specific binding molecule
NZ603488A (en) * 2010-07-09 2015-02-27 Crucell Holland Bv Anti-human respiratory syncytial virus (rsv) antibodies and methods of use
AU2011334867B2 (en) 2010-12-02 2016-10-13 Kling Biotherapeutics B.V. Means and methods for producing high affinity antibodies
EP3628689A1 (en) 2010-12-17 2020-04-01 Neurimmune Holding AG Human anti-sod1 antibodies
CN104080805B (en) * 2011-11-07 2017-02-22 美国政府健康及人类服务部 Neutralizing gp41 antibodies and their use
WO2013095091A2 (en) 2011-11-17 2013-06-27 Aimm Therapeutics B.V. Rsv g protein specific antibodies
CA2865856C (en) * 2012-03-20 2023-03-14 Humabs Biomed Sa Antibodies that neutralize rsv, mpv and pvm and uses thereof
US10017739B2 (en) 2012-09-06 2018-07-10 Duke University Methods of expanding and assessing B cells and using expanded B cells to treat disease
DK2950886T3 (en) * 2013-02-01 2020-03-23 Medimmune Llc RESPIRATORY SYNCYTIAL VIRUS-F-PROTEIN EPITOPER
CN112851766A (en) 2013-03-13 2021-05-28 美国政府(由卫生和人类服务部的部长所代表) Pre-fusion RSV F proteins and uses thereof
US9738689B2 (en) 2013-03-13 2017-08-22 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Prefusion RSV F proteins and their use
TWI659968B (en) 2013-03-14 2019-05-21 再生元醫藥公司 Human antibodies to respiratory syncytial virus f protein and methods of use thereof
US9060975B2 (en) 2013-03-14 2015-06-23 Mucosis Bv Heat-stable respiratory syncytial virus F protein oligomers and their use in immunological compositions
CN105722856B (en) * 2013-03-15 2019-04-16 厦门大学 The epitope of RSV fusion protein and the antibody for identifying it
US10047145B2 (en) * 2013-07-24 2018-08-14 Humabs Biomed Sa Antibodies that potently neutralize RSV and uses thereof
WO2015037992A1 (en) 2013-09-11 2015-03-19 Aimm Therapeutics B.V. Parechovirus specific antibodies
EA201691130A8 (en) 2013-12-17 2018-08-31 Аимм Терапьютикс Б.В. MEANS AND METHODS OF COUNTERACTION BY MYELOPROLIFERATIVE OR LYMPHOPROLIFERATIVE DISORDERS
LT3094353T (en) * 2014-01-15 2020-08-10 Medimmune, Llc Rsv-specific antibodies and functional parts thereof
CA2938193C (en) 2014-01-31 2023-05-02 Aimm Therapeutics B.V. Means and methods for producing stable antibodies
EP2974739A1 (en) 2014-07-15 2016-01-20 Novartis AG RSVF trimerization domains
US9340604B1 (en) * 2014-10-29 2016-05-17 Aridis Pharmaceuticals, Inc. Human monoclonal antibody specific for the F protein of respiratory syncytial virus (RSV)
JP5996707B2 (en) * 2015-04-15 2016-09-21 メディミューン リミテド RSV specific binding molecule
EP3124042A1 (en) 2015-07-28 2017-02-01 VIB, vzw Immunoglobulin single variable domain antibody against rsv prefusion f protein
MX2017016590A (en) 2015-06-18 2018-11-09 Vib Vzw Immunoglobulin single variable domain antibody against rsv prefusion f protein.
CA2989551A1 (en) 2015-06-24 2016-12-29 Aimm Therapeutics B.V. Aml antigens and uses thereof
KR102068915B1 (en) 2015-08-19 2020-01-22 화이자 인코포레이티드 Tissue Factor Pathway Inhibitor Antibodies and Uses thereof
JO3555B1 (en) 2015-10-29 2020-07-05 Merck Sharp & Dohme Antibody neutralizing human respiratory syncytial virus
RU2723039C2 (en) 2015-12-23 2020-06-08 Пфайзер Инк. Protein f rsv mutants
AU2017242020B2 (en) 2016-03-29 2021-08-05 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Substitutions-modified prefusion RSV F proteins and their use
CN110035771B (en) 2016-10-21 2024-03-08 阿迪马布有限责任公司 Anti-respiratory syncytial virus antibodies and methods of making and using the same
JP7265984B2 (en) 2016-10-21 2023-04-27 アディマブ, エルエルシー Anti-respiratory syncytial virus antibodies and methods of their production and use
AU2017345786A1 (en) 2016-10-21 2019-05-23 Adimab, Llc Anti-respiratory syncytial virus antibodies, and methods of their generation and use
US11084850B2 (en) 2016-12-16 2021-08-10 The Pirbright Institute Recombinant prefusion RSV F proteins and uses thereof
TWI761453B (en) 2017-03-01 2022-04-21 英商梅迪繆思有限公司 Anti-rsv monoclonal antibody formulation
KR20200115567A (en) 2018-01-29 2020-10-07 머크 샤프 앤드 돔 코포레이션 Stabilized RSV F protein and uses thereof
US11623004B2 (en) 2018-03-16 2023-04-11 University Of Pittsburgh Of The Commonwealth System Of Higher Education Compositions and methods for vaccination against respiratory syncytial virus infection
EP3773698A1 (en) 2018-04-03 2021-02-17 Sanofi Ferritin proteins
CN112512566A (en) 2018-04-03 2021-03-16 赛诺菲 Antigenic epstein-barr virus polypeptides
MX2020010199A (en) 2018-04-03 2021-01-08 Sanofi Sa Antigenic respiratory syncytial virus polypeptides.
WO2020212576A1 (en) * 2019-04-18 2020-10-22 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Biological and synthetic molecules inhibiting respiratory syncytial virus infection
JP2022060169A (en) 2020-10-02 2022-04-14 ファイザー・インク Cell culture process for rsv f protein production

Family Cites Families (161)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3164143D1 (en) 1980-07-01 1984-07-19 Nat Res Dev Production of viral antigens
JPS58500366A (en) 1981-03-06 1983-03-10 セルテツク リミテツド single clone antibody
US4526934A (en) * 1982-03-19 1985-07-02 Bridgestone Tire Company Limited Branched styrene-butadiene copolymers and pneumatic tires using the same
DE3378250D1 (en) 1982-04-22 1988-11-24 Ici Plc Continuous release formulations
US5340926A (en) 1983-03-25 1994-08-23 Celltech, Limited Process for the recovery of recombinantly produced protein from insoluble aggregate
JPS60100516A (en) 1983-11-04 1985-06-04 Takeda Chem Ind Ltd Preparation of sustained release microcapsule
US5332805A (en) 1984-02-03 1994-07-26 Celltech Limited Process for the recovery of recombinantly produced chymosin from insoluble aggregate
US5128326A (en) 1984-12-06 1992-07-07 Biomatrix, Inc. Drug delivery systems based on hyaluronans derivatives thereof and their salts and methods of producing same
FR2590674B1 (en) 1985-11-25 1989-03-03 Inst Nat Sante Rech Med NEW DIAGNOSTIC REAGENTS
US5149650A (en) 1986-01-14 1992-09-22 University Of North Carolina At Chapel Hill Vaccines for human respiratory virus
US4717766A (en) 1986-01-27 1988-01-05 Miles Laboratories, Inc. Method of preparing high titer anti-respiratory syncytial virus intravenous immune globulin
US4659563A (en) 1986-01-27 1987-04-21 Miles Laboratories, Inc. High titer anti-respiratory syncytial virus intravenous immune globulin
US5271927A (en) 1986-02-13 1993-12-21 Celltech Limited Antibody conjugates with macrocyclic ligands
US5468606A (en) 1989-09-18 1995-11-21 Biostar, Inc. Devices for detection of an analyte based upon light interference
DE3883899T3 (en) 1987-03-18 1999-04-22 Sb2 Inc CHANGED ANTIBODIES.
US4800078A (en) 1987-05-28 1989-01-24 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Immunotherapeutic method of treating respiratory disease by intranasal administration of Igb
GB8719041D0 (en) 1987-08-12 1987-09-16 Parker D Conjugate compounds
GB8720833D0 (en) 1987-09-04 1987-10-14 Celltech Ltd Recombinant dna product
US5223254A (en) 1987-09-29 1993-06-29 Praxis Biologics, Inc. Respiratory syncytial virus: vaccines
DE3878468T2 (en) 1987-12-23 1993-06-09 Upjohn Co CHIMARENE GLYCOPROTEINS, CONTAINING IMMUNOGENIC SEGMENTS OF THE HUMAN RESPIRATORY SYNCYTIAL VIRUS.
JP3095168B2 (en) 1988-02-05 2000-10-03 エル. モリソン,シェリー Antibodies with domain-denaturing constants
US5183657A (en) 1988-03-11 1993-02-02 Celltech Limited Antibodies for use in antilymphocyte antibody therapy
JPH01268646A (en) 1988-04-20 1989-10-26 Meiji Milk Prod Co Ltd Antitumor agent
US5137804A (en) 1988-05-10 1992-08-11 E. I. Du Pont De Nemours And Company Assay device and immunoassay
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
EP0368684B2 (en) 1988-11-11 2004-09-29 Medical Research Council Cloning immunoglobulin variable domain sequences.
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
IL162181A (en) 1988-12-28 2006-04-10 Pdl Biopharma Inc A method of producing humanized immunoglubulin, and polynucleotides encoding the same
US5354554A (en) 1989-02-10 1994-10-11 Celltech Limited Crosslinked antibodies and processes for their preparation
US6093872A (en) 1989-05-05 2000-07-25 Systemix, Inc. Extended human hematopoiesis in a heterologous host
FR2650598B1 (en) 1989-08-03 1994-06-03 Rhone Poulenc Sante DERIVATIVES OF ALBUMIN WITH THERAPEUTIC FUNCTION
US5332567A (en) 1989-08-24 1994-07-26 Immunomedics Detection and treatment of infections with immunoconjugates
US5518725A (en) 1989-09-25 1996-05-21 University Of Utah Research Foundation Vaccine compositions and method for induction of mucosal immune response via systemic vaccination
WO1991005548A1 (en) 1989-10-10 1991-05-02 Pitman-Moore, Inc. Sustained release composition for macromolecular proteins
AU642932B2 (en) 1989-11-06 1993-11-04 Alkermes Controlled Therapeutics, Inc. Protein microspheres and methods of using them
GB8928874D0 (en) 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
US5279935A (en) 1990-03-01 1994-01-18 Becton, Dickinson And Company Method of immunossay including deactivation of endogenous alkaline phosphatase
DE69120890T2 (en) 1990-05-03 1997-02-20 Systemix Inc HUMAN LYMPH TISSUE IN A HOST WITH ATTACKED IMMUNE SYSTEM
GB9020282D0 (en) 1990-09-17 1990-10-31 Gorman Scott D Altered antibodies and their preparation
JPH06509866A (en) 1991-04-22 1994-11-02 マサチューセッツ ヘルス リサーチ インスティチュート,インコーポレーテッド Method of screening plasma samples for effective antibody titers against respiratory viruses
DE69232706T2 (en) 1991-05-01 2002-11-28 Jackson H M Found Military Med METHOD FOR TREATING INFECTIOUS RESPIRATORY DISEASES
CA2044940A1 (en) 1991-06-10 1992-12-11 Inder M. Verma Transdominant negative proto-oncogene
WO1993005796A1 (en) 1991-09-19 1993-04-01 The Scripps Research Institute Method for producing human antibodies in a non-human animal, and animals therefor
US5240694A (en) 1991-09-23 1993-08-31 University Of Virginia Combined antiviral and antimediator treatment of common colds
US5418136A (en) 1991-10-01 1995-05-23 Biostar, Inc. Devices for detection of an analyte based upon light interference
EP0614530B1 (en) 1991-11-15 1998-03-04 Cornell Research Foundation, Inc. Indirect immunoassay for dioxinlike compounds
US20020102257A1 (en) 1998-09-21 2002-08-01 Leslie Sid Johnson Human-murine chimeric antibodies against respiratory syncytial virus
US5824307A (en) 1991-12-23 1998-10-20 Medimmune, Inc. Human-murine chimeric antibodies against respiratory syncytial virus
US5667988A (en) 1992-01-27 1997-09-16 The Scripps Research Institute Methods for producing antibody libraries using universal or randomized immunoglobulin light chains
FR2686901A1 (en) 1992-01-31 1993-08-06 Rhone Poulenc Rorer Sa NOVEL ANTITHROMBOTIC POLYPEPTIDES, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
FR2686899B1 (en) 1992-01-31 1995-09-01 Rhone Poulenc Rorer Sa NOVEL BIOLOGICALLY ACTIVE POLYPEPTIDES, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
US5912015A (en) 1992-03-12 1999-06-15 Alkermes Controlled Therapeutics, Inc. Modulated release from biocompatible polymers
JPH07507923A (en) 1992-03-20 1995-09-07 イムネット Human monoclonal antibodies and human monoclonal antibody production methods
GB9207479D0 (en) 1992-04-06 1992-05-20 Scotgen Ltd Novel antibodies for treatment and prevention of respiratory syncytial virus infection in animals and man
US6685942B1 (en) 1993-12-10 2004-02-03 The Scripps Research Institute Human neutralizing monoclonal antibodies to respiratory syncytial virus
ATE231002T1 (en) 1992-09-16 2003-02-15 Scripps Research Inst HUMAN NEUTRALIZING MONOCLONAL ANTIBODIES AGAINST RESPIRATORY SYNZYTIAL VIRUS
JPH08508240A (en) 1993-01-12 1996-09-03 ジョージ グリスティーナ,アンソニー Methods and compositions for direct transfer of passive immunity
US5934272A (en) 1993-01-29 1999-08-10 Aradigm Corporation Device and method of creating aerosolized mist of respiratory drug
US5424189A (en) 1993-03-05 1995-06-13 Kansas State University Research Foundation Bovine respiratory syncytial virus detection and primers
DE69426077T3 (en) 1993-05-25 2004-09-02 Wyeth Holdings Corp. ADJUVANTS FOR VACCINE AGAINST THE RESPIRATORY SYNCITIAL VIRUS
JPH08511420A (en) 1993-06-16 1996-12-03 セルテック・セラピューテイクス・リミテッド Body
AU689489B2 (en) 1993-07-30 1998-04-02 Oravax, Inc Monoclonal IgA antibody against respiratory syncytial virus
US5538952A (en) 1994-05-26 1996-07-23 Abbott Laboratories Inhibition of infection of mammalian cells by respiratory syncytial virus
US5506209A (en) 1994-05-26 1996-04-09 Abbott Laboratories Product for inhibition of infection of mammalian cells by respiratory syncytial virus
US5538733A (en) 1994-07-07 1996-07-23 Willmar Poultry Company, Inc. Method of priming an immune response in a one-day old animal
US5792456A (en) 1994-08-04 1998-08-11 Bristol-Myers Squibb Company Mutant BR96 antibodies reactive with human carcinomas
US5786464C1 (en) 1994-09-19 2012-04-24 Gen Hospital Corp Overexpression of mammalian and viral proteins
AU4755696A (en) 1995-01-05 1996-07-24 Board Of Regents Acting For And On Behalf Of The University Of Michigan, The Surface-modified nanoparticles and method of making and using same
US6019968A (en) 1995-04-14 2000-02-01 Inhale Therapeutic Systems, Inc. Dispersible antibody compositions and methods for their preparation and use
US6121022A (en) 1995-04-14 2000-09-19 Genentech, Inc. Altered polypeptides with increased half-life
US5811524A (en) 1995-06-07 1998-09-22 Idec Pharmaceuticals Corporation Neutralizing high affinity human monoclonal antibodies specific to RSV F-protein and methods for their manufacture and therapeutic use thereof
JP2000507912A (en) 1995-08-31 2000-06-27 アルカームズ コントロールド セラピューティックス,インコーポレイテッド Sustained release composition of active agent
AU7075496A (en) 1995-09-18 1997-04-09 Intracel Corporation Neutralizing monoclonal antibodies to respiratory syncytial virus
DK0885002T3 (en) 1996-03-04 2011-08-22 Penn State Res Found Materials and methods for enhancing cellular internalization
AU728657B2 (en) 1996-03-18 2001-01-18 Board Of Regents, The University Of Texas System Immunoglobulin-like domains with increased half-lives
US5874064A (en) 1996-05-24 1999-02-23 Massachusetts Institute Of Technology Aerodynamically light particles for pulmonary drug delivery
US5985309A (en) 1996-05-24 1999-11-16 Massachusetts Institute Of Technology Preparation of particles for inhalation
US5855913A (en) 1997-01-16 1999-01-05 Massachusetts Instite Of Technology Particles incorporating surfactants for pulmonary drug delivery
US6114148C1 (en) 1996-09-20 2012-05-01 Gen Hospital Corp High level expression of proteins
WO1998023289A1 (en) 1996-11-27 1998-06-04 The General Hospital Corporation MODULATION OF IgG BINDING TO FcRn
FR2758331B1 (en) 1997-01-14 1999-03-05 Univ Bourgogne NEW MEANS FOR DIAGNOSIS, PREVENTION AND TREATMENT FOR CONTAMINATION OR INFECTIONS WITH MUCOUS TROPISM VIRUSES
PT954282E (en) 1997-01-16 2005-06-30 Massachusetts Inst Technology PREPARATION OF PARTICLES FOR INHALATION
US6590079B2 (en) 1997-01-30 2003-07-08 Ixsys, Incorporated Anti-αvβ3 recombinant human antibodies, nucleic acids encoding same
JP2002515056A (en) 1997-02-06 2002-05-21 メルク エンド カンパニー インコーポレーテッド Thimerosal-free preservative for vaccines
US6117980A (en) 1997-02-21 2000-09-12 Genentech, Inc. Humanized anti-IL-8 monoclonal antibodies
US6277375B1 (en) 1997-03-03 2001-08-21 Board Of Regents, The University Of Texas System Immunoglobulin-like domains with increased half-lives
US5989463A (en) 1997-09-24 1999-11-23 Alkermes Controlled Therapeutics, Inc. Methods for fabricating polymer-based controlled release devices
SE512663C2 (en) 1997-10-23 2000-04-17 Biogram Ab Active substance encapsulation process in a biodegradable polymer
ATE317437T1 (en) 1997-12-01 2006-02-15 Us Gov Health & Human Serv ANTIBODIES, AS WELL AS FV MOLECULES, AND IMMUNOCONJUGATES WITH HIGH BINDING AFFINITY FOR MESOTHELIN AND METHODS FOR THEIR USE
DK1068241T3 (en) 1998-04-02 2008-02-04 Genentech Inc Antibody variants and fragments thereof
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
US6528624B1 (en) 1998-04-02 2003-03-04 Genentech, Inc. Polypeptide variants
WO1999066903A2 (en) 1998-06-24 1999-12-29 Advanced Inhalation Research, Inc. Large porous particles emitted from an inhaler
US6572856B1 (en) 1998-09-10 2003-06-03 The University Of Virginia Patent Foundation Methods for the prevention and treatment of cancer using anti-C3b(i) antibodies
DK1135498T3 (en) 1998-11-18 2008-05-26 Genentech Inc Antibody variants with higher binding affinity than parental antibodies
US7553487B2 (en) * 1998-12-14 2009-06-30 Genetics Institute, Llc Method and compositions for treating asthma
NZ539776A (en) 1999-01-15 2006-12-22 Genentech Inc Polypeptide variants with altered effector function
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
AR022952A1 (en) 1999-03-19 2002-09-04 Smithkline Beecham Corp MONOCLONAL ANTIBODY OF ROEDOR SPECIFICALLY NEUTRALIZING FOR HUMAN INTERLEUQUINE-18, A FAB NEUTRALIZING FRAGMENT OR FRAGMENT F (AB ') 2, A COMPLEMENTARITY REGION OF IMMONOGLOBULIN LIGHT CHAIN (CDR), WHICH MAKES IT COMPRESSED , THE
US7090973B1 (en) * 1999-04-09 2006-08-15 Oscient Pharmaceuticals Corporation Nucleic acid sequences relating to Bacteroides fragilis for diagnostics and therapeutics
CO5280147A1 (en) * 1999-05-18 2003-05-30 Smithkline Beecham Corp MONOCLONAL HUMAN ANTIBODY
WO2000073346A1 (en) 1999-05-27 2000-12-07 The Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services Immunoconjugates having high binding affinity
JP4992068B2 (en) 2000-01-27 2012-08-08 メディミューン,エルエルシー Ultra high affinity neutralizing antibody
US7229619B1 (en) 2000-11-28 2007-06-12 Medimmune, Inc. Methods of administering/dosing anti-RSV antibodies for prophylaxis and treatment
US20010034062A1 (en) 2000-02-09 2001-10-25 Scott Koenig Antibody gene therapy with adeno-associated viral vectors
MXPA02007733A (en) 2000-02-11 2004-09-10 Merck Patent Gmbh Enhancing the circulating half life of antibody based fusion proteins.
CA2401652A1 (en) 2000-03-01 2001-09-07 Medimmune, Inc. High potency recombinant antibodies and method for producing them
WO2001064248A1 (en) 2000-03-02 2001-09-07 Medimmune, Inc. Methods of enhancing activity of vaccines and vaccine compositions
ES2529300T3 (en) 2000-04-12 2015-02-18 Novozymes Biopharma Dk A/S Albumin fusion proteins
WO2001079299A1 (en) 2000-04-13 2001-10-25 The Rockefeller University Enhancement of antibody-mediated immune responses
US20020004046A1 (en) 2000-05-03 2002-01-10 Johnson Leslie S. Combination therapy of respiratory diseases using antibodies
EP1152199B1 (en) 2000-05-03 2005-07-27 IPV Inheidener Produktions- und Vertriebsgesellschaft mbH Thermal container
ATE457177T1 (en) 2000-05-03 2010-02-15 Medimmune Llc COMBINATION THERAPY FOR THE TREATMENT OF RESPIRATORY DISEASES USING ANTIBODIES AND ANTI-INFLAMMATORY ACTIVES
AU2001263443A1 (en) 2000-05-25 2001-12-03 Med Immune, Inc. F-protein epitope-based vaccine for respiratory syncytial virus infection
EP1336410A4 (en) 2000-08-04 2005-10-12 Chugai Pharmaceutical Co Ltd Protein injection preparations
US6565888B1 (en) 2000-08-23 2003-05-20 Alkermes Controlled Therapeutics, Inc. Methods and compositions for the targeted delivery of biologically active agents
US6699473B2 (en) * 2000-10-13 2004-03-02 Uab Research Foundation Human anti-epidermal growth factor receptor single-chain antibodies
US6855493B2 (en) 2000-11-28 2005-02-15 Medimmune, Inc. Methods of administering/dosing anti-RSV antibodies for prophylaxis and treatment
TWI327600B (en) 2000-11-28 2010-07-21 Medimmune Llc Methods of administering/dosing anti-rsv antibodies for prophylaxis and treatment
US6818216B2 (en) * 2000-11-28 2004-11-16 Medimmune, Inc. Anti-RSV antibodies
US7179900B2 (en) 2000-11-28 2007-02-20 Medimmune, Inc. Methods of administering/dosing anti-RSV antibodies for prophylaxis and treatment
US7658921B2 (en) 2000-12-12 2010-02-09 Medimmune, Llc Molecules with extended half-lives, compositions and uses thereof
EP2357187A1 (en) 2000-12-12 2011-08-17 MedImmune, LLC Molecules with extended half-lives, compositions and uses thereof
US20040096451A1 (en) 2002-07-25 2004-05-20 Young James F. Methods of treating and preventing RSV, hMPV, and PIV using anti-RSV, anti-hMPV, and anti-PIV antibodies
US7091323B2 (en) 2001-04-24 2006-08-15 Bayer Corporation Human TIMP-1 antibodies
AR039067A1 (en) * 2001-11-09 2005-02-09 Pfizer Prod Inc ANTIBODIES FOR CD40
EP2141228A1 (en) * 2001-12-18 2010-01-06 Cancer Research Technology Limited Method for the generation of proliferating and differentiating B cell lines
US6911321B2 (en) 2001-12-19 2005-06-28 Genentech, Inc. Non-human primate Fc receptors and methods of use
US20040002587A1 (en) 2002-02-20 2004-01-01 Watkins Jeffry D. Fc region variants
DE60328195D1 (en) 2002-03-29 2009-08-13 Schering Corp HUMAN MONOCLONAL ANTIBODIES TO INTERLEUKIN-5 AND THESE COMPREHENSIVE METHODS AND COMPOSITIONS
WO2003102136A2 (en) * 2002-05-30 2003-12-11 Human Genome Sciences, Inc. Antibodies that specifically bind to neurokinin b
US7132100B2 (en) 2002-06-14 2006-11-07 Medimmune, Inc. Stabilized liquid anti-RSV antibody formulations
US7425618B2 (en) 2002-06-14 2008-09-16 Medimmune, Inc. Stabilized anti-respiratory syncytial virus (RSV) antibody formulations
EP1539236A4 (en) 2002-07-03 2007-07-18 Univ Pennsylvania Compositions, methods and kits relating to anti-platelet autoantibodies and inhibitors thereof
PT1534335E (en) 2002-08-14 2012-02-28 Macrogenics Inc Fcgammariib-specific antibodies and methods of use thereof
EP2042517B1 (en) 2002-09-27 2012-11-14 Xencor, Inc. Optimized FC variants and methods for their generation
CA2499891A1 (en) 2002-10-03 2004-04-15 Large Scale Biology Corporation Multimeric protein engineering
DE60334141D1 (en) 2002-10-15 2010-10-21 Facet Biotech Corp CHANGE OF FcRn BINDING SAFFINITIES OR SERUM HALF TIMES OF ANTIBODIES BY MUTAGENESIS
WO2004043989A2 (en) 2002-11-07 2004-05-27 Medarex, Inc. Human monoclonal antibodies to heparanase
US7696334B1 (en) * 2002-12-05 2010-04-13 Rosetta Genomics, Ltd. Bioinformatically detectable human herpesvirus 5 regulatory gene
GB0306618D0 (en) 2003-03-22 2003-04-30 Univ Newcastle Antibody
AU2005214988A1 (en) 2004-02-17 2005-09-01 Absalus, Inc. Super-humanized antibodies against respiratory syncytial virus
JP2008513540A (en) 2004-09-21 2008-05-01 メディミューン,インコーポレーテッド Antibody to respiratory syncytial virus and method for producing vaccine for the virus
US20060083741A1 (en) 2004-10-08 2006-04-20 Hoffman Rebecca S Treatment of respiratory syncytial virus (RSV) infection
US20060115485A1 (en) 2004-10-29 2006-06-01 Medimmune, Inc. Methods of preventing and treating RSV infections and related conditions
US7423128B2 (en) * 2004-11-03 2008-09-09 Amgen Fremont Inc. Anti-properdin antibodies, and methods for making and using same
WO2006055638A2 (en) 2004-11-17 2006-05-26 Abgenix, Inc. Fully human monoclonal antibodies to il-13
CA2591665C (en) 2004-12-20 2015-05-05 Crucell Holland B.V. Binding molecules capable of neutralizing west nile virus and uses thereof
ATE521638T1 (en) 2004-12-21 2011-09-15 Medimmune Ltd ANTIBODIES DIRECTED AGAINST ANGIOPOIETIN-2 AND USES THEREOF
ZA200709291B (en) 2005-04-25 2009-01-28 Pfizer Antibodies to myostatin
KR20080025174A (en) 2005-06-23 2008-03-19 메디뮨 인코포레이티드 Antibody formulations having optimized aggregation and fragmentation profiles
US9127251B2 (en) * 2005-12-09 2015-09-08 Academisch Medisch Centrum Bij De Universiteit Van Amsterdam Means and methods for influencing the stability of antibody producing cells
AR056857A1 (en) * 2005-12-30 2007-10-24 U3 Pharma Ag DIRECTED ANTIBODIES TO HER-3 (RECEIVER OF THE HUMAN EPIDERMAL GROWTH FACTOR-3) AND ITS USES
EP1997830A1 (en) 2007-06-01 2008-12-03 AIMM Therapeutics B.V. RSV specific binding molecules and means for producing them
JP2010531890A (en) 2007-06-26 2010-09-30 メディミューン,エルエルシー Method of treating RSV infection and related symptoms
EP2185590A2 (en) 2007-09-07 2010-05-19 Symphogen A/S Methods for recombinant manufacturing of anti-rsv antibodies
WO2009114815A1 (en) 2008-03-13 2009-09-17 Dyax Corp Libraries of genetic packages comprising novel hc cdr3 designs
US8568726B2 (en) 2009-10-06 2013-10-29 Medimmune Limited RSV specific binding molecule
ES2621458T3 (en) 2009-10-06 2017-07-04 Medimmune Limited RSV specific binding molecule
DK2950886T3 (en) 2013-02-01 2020-03-23 Medimmune Llc RESPIRATORY SYNCYTIAL VIRUS-F-PROTEIN EPITOPER
LT3094353T (en) 2014-01-15 2020-08-10 Medimmune, Llc Rsv-specific antibodies and functional parts thereof

Also Published As

Publication number Publication date
CY1117767T1 (en) 2017-05-17
NZ621824A (en) 2016-02-26
KR20150036796A (en) 2015-04-07
IL202288A (en) 2016-10-31
AU2008257801B8 (en) 2014-09-11
JP2010528601A (en) 2010-08-26
EP2170952B1 (en) 2016-03-09
HK1146073A1 (en) 2011-05-13
BRPI0812878C1 (en) 2021-05-25
BRPI0812878A2 (en) 2015-03-31
DK2170952T3 (en) 2016-06-20
HUE029225T2 (en) 2017-02-28
NZ581512A (en) 2012-10-26
AU2008257801B2 (en) 2014-08-14
BRPI0812878B8 (en) 2020-04-22
MX350375B (en) 2017-09-05
RU2014125126A (en) 2015-09-20
JP2018196380A (en) 2018-12-13
KR101599556B1 (en) 2016-03-04
MX2009012843A (en) 2010-04-21
CN104961825A (en) 2015-10-07
PL2170952T3 (en) 2016-10-31
SI2170952T1 (en) 2016-08-31
RU2527067C2 (en) 2014-08-27
ZA200908320B (en) 2010-08-25
US9321831B2 (en) 2016-04-26
JP5450395B2 (en) 2014-03-26
RU2628095C2 (en) 2017-08-14
BRPI0812878B1 (en) 2020-01-21
EP2170952A2 (en) 2010-04-07
IL202288A0 (en) 2010-06-16
RU2009148886A (en) 2011-07-20
US20100239593A1 (en) 2010-09-23
KR101652652B1 (en) 2016-08-30
NZ712667A (en) 2017-01-27
JP2017019836A (en) 2017-01-26
EP1997830A1 (en) 2008-12-03
US20180327484A1 (en) 2018-11-15
AU2008257801A1 (en) 2008-12-04
US10730931B2 (en) 2020-08-04
US20160244509A1 (en) 2016-08-25
PT2170952E (en) 2016-06-16
US20200325213A1 (en) 2020-10-15
CA2689290A1 (en) 2008-12-04
US20140072575A1 (en) 2014-03-13
KR20100028592A (en) 2010-03-12
EP2578600A2 (en) 2013-04-10
US10059757B2 (en) 2018-08-28
WO2008147196A2 (en) 2008-12-04
JP2014079251A (en) 2014-05-08
WO2008147196A3 (en) 2009-02-05
CN101778866A (en) 2010-07-14
CN101778866B (en) 2014-11-26
HRP20160632T1 (en) 2016-07-15
US20140377279A9 (en) 2014-12-25
US8562996B2 (en) 2013-10-22
CA2689290C (en) 2017-01-17
EP2578600A3 (en) 2013-07-31
AU2008257801A8 (en) 2014-09-11
ES2575129T3 (en) 2016-06-24

Similar Documents

Publication Publication Date Title
CA2689290C (en) Rsv-specific binding molecules and means for producing them
ES2367322T3 (en) METHODS TO OBTAIN IMMORTALIZED CELLS THAT SECRET ANTIBODIES.
CN102083858B (en) Anti-IL 17A/IL-17F cross-reactive antibodies and methods of use thereof
AU2011329647B2 (en) Neutralizing anti-CCL20 antibodies
KR20140014384A (en) Anti-il-17f antibodies and methods of use thereof
AU2014262169B2 (en) Rsv-specific binding molecules and means for producing them

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20170503

EEER Examination request

Effective date: 20170503

FZDE Discontinued

Effective date: 20230616