AU2014262169B2 - 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
AU2014262169B2
AU2014262169B2 AU2014262169A AU2014262169A AU2014262169B2 AU 2014262169 B2 AU2014262169 B2 AU 2014262169B2 AU 2014262169 A AU2014262169 A AU 2014262169A AU 2014262169 A AU2014262169 A AU 2014262169A AU 2014262169 B2 AU2014262169 B2 AU 2014262169B2
Authority
AU
Australia
Prior art keywords
sequence
antibody
acid sequence
rsv
cell
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.)
Active
Application number
AU2014262169A
Other versions
AU2014262169A1 (en
Inventor
Tim Beaumont
Mark Jeroen KWAKKENBOS
Hergen Spits
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
Priority claimed from EP07109472A external-priority patent/EP1997830A1/en
Application filed by MedImmune Ltd filed Critical MedImmune Ltd
Priority to AU2014262169A priority Critical patent/AU2014262169B2/en
Publication of AU2014262169A1 publication Critical patent/AU2014262169A1/en
Application granted granted Critical
Publication of AU2014262169B2 publication Critical patent/AU2014262169B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Landscapes

  • Peptides Or Proteins (AREA)

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 2014262169 15 Jan 2015
The present application is a divisional application from Australian Patent Application No. 2008257801, which claims priority from EP Application No. 07109472.6, filed 1 June 2007, the entire disclosure of which is incorporated herein by reference. 5 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 10 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 15 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 20 alternative antibodies and therapies against RSV. A reference herein to a patent document or other matter which is given as prior art is not to be taken as an admission that that document or matter was known or that the information it contains was part of the common general knowledge as at the priority date of any of the claims. 25 Throughout the description and claims of the specification, the word "comprise" and variations of the word, such as "comprising" and "comprises", is not intended to exclude other additives, components, integers or steps.
It is an aspect of the present invention to provide means and methods for counteracting and/or preventing an RSV-related disease. It is a further aspect of the invention to provide 30 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. 1
It is a further aspect of the present invention to provide an isolated, synthetic or recombinant antibody or a functional part, derivative or analogue thereof which specifically binds Respiratory Syncytial Virus (RSV) and which comprises: 2014262169 07 Dec 2016 a variable heavy chain sequence comprising a heavy chain complementarity 5 determining region CDR1 comprising the amino acid sequence GFSFSHYA (SEQ ID NO:73), a heavy chain CDR2 sequence comprising the amino acid sequence ISYDGENT (SEQ ID NO:74), and a heavy chain CDR3 sequence comprising the amino acid sequence ARDRIVDDYYYYGMDV (SEQ ID NO:75), and a variable light chain sequence comprising a light chain CDR1 comprising the amino acid 10 sequence QDIKKY (SEQ ID NO:76), a light chain CDR2 sequence comprising the amino acid sequence DAS, and a light chain CDR3 sequence comprising the amino acid sequence QQYDNLPPLT (SEQ ID NO:77). It is still a further aspect of the present invention to provide an isolated antibody producing cell producing an antibody, functional part, derivative, or analogue thereof, wherein the antibody, functional part, derivative, or analogue thereof 15 specifically binds Respiratory Syncytial Virus (RSV), comprises a heavy chain comprising a heavy chain CDR1 sequence comprising the sequence GFSFSHYA (SEQ ID NO:73), a heavy chain CDR2 sequence comprising the sequence ISYDGENT (SEQ ID NO:74), and a heavy chain CDR3 sequence comprising the sequence ARDRIVDDYYYY GMDV (SEQ ID NO:75), and 20 comprises a light chain comprising a light chain CDR1 sequence comprising the sequence QDIKKY (SEQ ID NO:76), a light chain CDR2 sequence comprising the sequence DAS, and a light chain CDR3 sequence comprising the sequence QQYDNLPPLT (SEQ ID NO:77); and wherein the heavy chain and the light chain are encoded by exogenous nucleotide 25 sequences.
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. 30 Non-specific sticking is not la PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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 5 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 antigenbinding properties of D25, are depicted in Figure 11D. Antibody D25 appears to have superior characteristics as compared to the registered anti-RSV antibody 10 Palivizumab (Figure 8). For instance, D25 has an IC50 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 IC50 value of about 453 ng/ml. A functional equivalent of an antibody is defined herein as a functional 15 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 20 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 25 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. 30 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 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014
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 5 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 10 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 15 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. 20 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 25 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 30 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 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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 5 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% 10 identical to the sequence GIIPVLGTVHYAPKFQG, and/or - a heavy chain CDR3 sequence comprising a sequence which is at least 70% identical to the sequence ETALWSTTYLPHYFDN, and/or - a light chain CDR1 sequence comprising a sequence which is at least 85% identical to the sequence QASQDIVNYLN, and/or 15 - 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. 20 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 25 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 30 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 35 - a heavy chain CDR3 sequence comprising the sequence 4 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 ETALWSTTYLPHYFDN, 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 5 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 10 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 15 sequence comprising a sequence which is at least 70% identical to the sequence ETALWSTTYLPHYFDN 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 20 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 25 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 30 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 35 the sequence 5 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 QVQLVQSGAEVKKPGSSVMVSCQASGGPLRNYIINWLRQAPGQGPEWMGGII PVLGTVHYAPKFQGRVTITADESTDTAYIHLISLRSEDTAMYYCATETALWST TYLPHYFDNWGQGTLVTVSS. Moreover, variable light chain amino acid sequences which are at least 70% identical to the light chain sequence as 5 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 10 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 15 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 20 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 25 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 30 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 35 sequence as depicted in Figure 11 is therefore also provided. Said functional part 6 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014
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 5 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 10 depicted in Figure 11D
Another particularly preferred anti-RSV antibody according to the present invention is the antibody designated “AM 14”, which has a heavy chain region and 15 a light chain region as depicted in Figure 14A. The CDR sequences of AM 14, 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 20 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. 25 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 30 binding characteristics of AM14 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 35 thereof comprising an amino acid sequence which is at least 70% identical to a 7 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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 5 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 10 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% 15 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 20 - 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. 25 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 30 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 35 equivalent thereof which is capable of specifically binding Respiratory Syncytial 8 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014
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 5 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 10 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 15 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 20 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 25 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. 30
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 35 provided is therefore an antibody or a functional equivalent thereof, having a 9 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 heavy chain sequence comprising a sequence which is at least 70% identical to the sequence EVQLVESGGGWQPGRSLRLSCAASGFSFSHYAMHWVRQAPGKGLEWVAVIS YDGENTYYADSVKGRFSISRDNSKNTVSLQMNSLRPEDTALYYCARDRIVDD 5 YYYYGMDVWGQGATVTVSS. 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 10 DIQMTQSPSSLSASVGDRVTITCQASQDIKKYLNWYHQKPGKVPELLMHDASNLETGVPSRF SGRGSGTDFTLTISSLQPEDIGTYYCQQYDNLPPLTFGGGTKVEIKRTV 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%, 15 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 20 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. 25 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 30 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 35 functional equivalent of an antibody comprising at least a functional part of a 10 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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. 5
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 10 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 15 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, 20 with at least one altered property as compared to AM16. 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 25 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 30 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. 35 11 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014
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: 5 - a heavy chain CDRl 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% 10 identical to the sequence AREDYGPGNYYSPNWFDP, and/or - a light chain CDRl 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 15 - 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 20 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 25 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: 30 - a heavy chain CDRl 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 CDRl sequence comprising the sequence SSNIGAGYD, and/or 12 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 - a light chain CDR2 sequence comprising the sequence GNT, 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 5 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 10 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 15 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 20 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. 25
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 30 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 EVQLVETGGGLAQPGGSLRLSCAASGFTFSSYNMNWVRQAPGKGLEWVSHISAGSSYIYYSD SVKGRFTVSRDNVRNSVYLQMNS LRAADTAVYYCARE DYGPGNYYS PNWFDPWGQGTLVTVS 35 S. Moreover, light chain amino acid sequences which are at least 70% identical to 13 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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 5 QSWTQPPSVSGAPGQRVTISCTGSSSNIGAGYDVHWYQQLPGTAPKLLIYGNTNRPSGVSD RFSGSKSGTSASLAITGLQAEDEADYYCHSYDRSLSGSVFGGGTKLTV 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%, 10 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 15 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. 20 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 25 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 30 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. 35 14 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014
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 5 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 10 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, 15 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 20 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 25 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. 30
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: 35 - a heavy chain CDR1 sequence comprising a sequence which is at least 70% 15 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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% 5 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 10 - 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 15 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 20 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: 25 - a heavy chain CDR1 sequence comprising the sequence GFNFHNYG, and/or - a heavy chain CDR2 sequence comprising the sequence VWYDGSKK, and/or - a heavy chain CDR3 sequence comprising the sequence VRDKVGPTPYFDS, and/or - a light chain CDR1 sequence comprising the sequence NIGSET, and/or 30 - 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 35 thereto. Further provided is therefore an isolated, synthetic or recombinant 16 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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 5 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 10 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 15 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. 20 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 25 heavy chain sequence comprising a sequence which is at least 70% identical to the sequence EVQLVESGGNVVKPGTSLRLSCAATGFNFHNYGMNWVRQAPGKGLEWVAWWYDGSKKYYAD SVTGRFAISRDNSKNTLYLQMNSLRVEDTAVYYCVRDKVGPTPYFDSWGQGTLVTVSS. Moreover, light chain amino acid sequences which are at least 70% identical to a 30 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 SYVLTQPPSVSLAPGGTAAITCGRNNIGSETVHWYQQKPGQAPVLWYDDDDRPSGIPERFS 35 GSNSGNTATLTISRVEAGDEADYYCQVWDRSNYHQVFGGGTKLTV is therefore also 17 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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 5 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 10 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 15 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 20 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 14C is produced. A functional equivalent of an antibody comprising at least a functional part of a 25 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 30 equivalents thereof having improved properties as compared to prior art antibodies. The inventors have succeeded in generating RSV-specific antibodies with low IC50 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. 35 One embodiment provides an antibody which has an IC50 value of less than 10 18 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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 ICgQ value of less than 5 ng/ml, more preferably less than 2 ng/ml. The preferred antibody D25 has an IC5Q value of about 0.5-1.5 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-10 effects due to an immunological reaction in a human individual against nonhuman 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 15 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 20 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 25 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 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. 19 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014
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 5 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 10 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 15 therapeutically effective amount of an antibody or functional equivalent according to the invention. Said antibody preferably comprises antibody D25, AM14, AM16 and/or AM23, 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 20 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 25 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 30 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 35 acceptable carrier, adjuvant, diluent and/or excipient. Examples of suitable 20 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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 5 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 10 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 15 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 20 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 25 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 30 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 21 for at least nine weeks, preferably for at least three months, more preferably for at least six months is therefore also herewith provided. 2014262169 15 Jan 2015
The present inventors have used their insight that the stability of an RSV-specific 5 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 10 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 disclosed to differentiate upon stimulation into an antibody-secreting plasma cell via a stage which some researchers call a 15 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 20 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 25 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 BCL6 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 30 in which the cells continue to proliferate. With a method of the inventors it has therefore become possible to increase the replicative life span 22 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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. 5
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 10 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 15 comprises a B cell and/or 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 naive 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. 20
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 25 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 30 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 35 Prdml. Blimp-1 drives plasma cell differentiation. BCL6 and Blimp-1 repress 23 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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 naive or memory B cells involves downregulation of BCL6 and upregulation of Blimp-1. In germinal 5 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 10 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 15 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-1 20 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 25 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 30 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 35 sequence encoding BCL6 or a functional part, derivative and/or analogue thereof 24 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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 5 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. 10
Preferably, as outlined below in more detail, an RSV-specific antibody producing cell according to the invention comprises an exogenous nucleic acid sequence encoding Bcl-xL or a functional part, derivative and/or analogue thereof. If Bcl-xL or a functional part, derivative and/or analogue thereof is present, it is 15 possible to grow plasmablasts under conditions of low cell density. Expression of said nucleic acid sequence encoding Bcl-xL or a functional part, derivative and/or analogue thereof is preferably regulated by an exogenous inducer of repressor, so that the extent of Bcl-xL expression is regulated at will. A preferred embodiment therefore provides an RSV-specific antibody producing cell comprising: 20 - 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. Said RSV-specific antibody producing cell preferably comprises both an exogenous nucleic acid sequence encoding BCL6 - or 25 a functional part, derivative and/or analogue thereof - and an exogenous nucleic acid sequence encoding Bcl-xL - or a functional part, derivative and/or analogue thereof. Preferably, expression of said 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 30 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 35 antibody producing cell. An RSV-specific antibody producing cell is preferably 25 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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 5 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 coexpressing BCL6 as well as Blimp-1.
One embodiment involves first stabilizing total antibody-producing cells 10 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 15 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 20 Bcl-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. 25 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. 30 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 35 producing RSV-specific antibodies or functional equivalents thereof, the method 26 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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. 5
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 10 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 coexpression 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 15 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 20 the addition of the anti-apoptotic gene Bcl-xL. With the introduction of Bcl-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 Bcl-xL with any of the herein 25 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 30 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 35 part, derivative and/or analogue thereof, and/or a nucleic acid sequence coding 27 therefore. STAT5 is a signal transducer capable of enhancing BCL6 expression. There are two disclosed 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 5 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. 2014262169 15 Jan 2015 10 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. 15 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 20 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, 25 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, 30 an antibody producing cell is provided with a nucleic acid sequence encoding an estrogen receptor (ER) and STAT5 as a fusion protein 28 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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 5 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 10 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 15 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 20 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 25 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. 30
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 35 analogue thereof. As explained herein before, an exogenous nucleic acid encoding 29 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 5 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 10 encoding BCL6 or a functional part, derivative and/or analogue thereof, so that BCL6 2014262169 15 Jan 2015 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 15 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 20 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. E12 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 25 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 disclosed 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 30 (plasmacell). 30 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014
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 5 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 10 according to the invention comprising culturing said antibody producing cell in the presence of a compound capable of directly or indirectly influencing Blimp-1 expression. Preferably, a compound is used that is capable of enhancing Blimp-1 expression in order to counteract downregulation of Blimp-1 during expression of BCL6. Said compound most preferably comprises IL-21. 15
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. STAT3 20 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 25 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 30 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 35 possible to indirectly regulate Blimp-1 expression by administering a compound 31 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 5 producing cell is provided with a compound capable of directly or indirectly enhancing activity of STAT3. 2014262169 15 Jan 2015
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. 10 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 disclosed to activate STAT3. Moreover, Toll-like receptors (TLRs) which are involved in innate immunity 15 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 20 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 25 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 30 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. 32 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014
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 5 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 10 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 15 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 STAT5 via E47 and SOCS results in decreased BCL6 levels and subsequently in 20 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). 25
By at least a functional part of a STAT5 protein, a STAT3 protein, Bcl-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, Bcl-xL and/or 30 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, Bcl-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 35 essentially the same in kind, not necessarily in amount. A derivative is provided 33 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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, 5 such as a STAT5-ER or STAT3-ER fusion protein whose activity depends on the presence of 4 hydroxy-tamoxifen (4HT). An analogue of a STAT5 protein, a STAT3 protein, Bcl-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 STAT5 10 protein, STAT3 protein, Bcl-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 15 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. 20
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 Bcl-xL expression product within said antibody producing cell. This is for example 25 accomplished by providing said antibody producing cell with a nucleic acid sequence encoding Bcl-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 Bcl-2. In yet another embodiment this is accomplished by providing said antibody producing cell with a compound capable of directly or indirectly 30 enhancing Bcl-xL expression, preferably said compound comprises APRIL, BAFF, CD40, BCR stimulation, cytokines, growth factors or downstream effectors like JNK and AKT (PKB).
Bcl-xL is a member of the anti-apoptotic Bcl-2 family, Bcl2-proteins interact with and counteract so-called Bcl-2 homology domain 3 (BH3)-only 35 family members such as Bax, Bak, Bim, and Bad, which induce cytochome c 34 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 release following intrinsic death stimuli (Boise, L. H., 1993). Thus, protection of mitochondrial membrane integrity through proteins like Bcl-xL is critical for cell survival. STAT5 activation has been shown to protect cells from cell death. STAT5 5 has been shown to regulate the expression of Bcl-xL, supporting an anti-apoptotic role for STAT5. STAT5 positively regulates the Bcl-xL expression through STAT binding elements within the Bcl-xL promoter. In vivo, Bcl-xL expression is absent in bone marrow of STAT5A/B-doubly deficient mice. Furthermore, STAT5-mediated erythroblast survival is dependent upon upregulation of Bcl-xL. 10 Recently, it has been shown that transgenic overexpression of Bcl-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 15 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 20 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 25 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 30 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 Bcl-xL. An RSV-specific 35 antibody producing cell according to the present invention provides the 35 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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 5 according to the invention is preferably cultured in the presence of CD40L since replication of most B cells is favoured by CD40L.
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 10 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 Bcl-xL expression, resulting in even more preferred proliferation and antibody production properties and/or stability. 15 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; 20 - 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 25 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 Bcl-xL. Further provided is therefore a method for producing an RSV-specific antibody producing cell which is stable for at least three months, 30 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 Bcl-xL or a functional part, derivative and/or analogue thereof; and 35 - culturing said B cell. 36 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014
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 Bcl-xL or a functional part, derivative and/or analogue thereof.
Said B cell is preferably cultured in the presence of a compound capable of 5 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 10 cultured in the presence of IL-21 before said B cell is transduced with a nucleic acid sequence encoding BCL6 and/or Bcl-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 15 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 20 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 25 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 Bcl-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 30 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 Bcl-xL. Afterwards, the B cells are again cultured in the 35 absence of 11-21 and in the presence of 11-4 and L-cells during 3-5 days in order to 37 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 allow BCL6 expression. Subsequently, cells expressing BCL6 and Bcl-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 Bcl-6,
Blimp 1 and Bcl-XL expressing cells in the culture supernatant are preferably 5 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 10 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 15 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 20 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 25 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 microorganisms. 30 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 35 produced. This is preferably performed by using memory B cells that have for 38 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 instance been isolated from a sample by selection for CD 19 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 5 component derived from RSV, such as for instance the RSV F protein, G protein and/or SH 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 Bcl-xL 10 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 15 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 20 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 25 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 30 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 39 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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 5 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 14B and/or Figure 14C is provided. Said functional part preferably comprises at least 10 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 15 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, 20 GGGATCATTCCTGTCTTGGGTACAGTACACTACGCACCGAAGTTCCAGGGC,
AGAGTCACGATTACCGCGGACGAATCCACAGACACAGCCTACATCCATCTGATCAGCCTGAG ATCTGAGGACACGGCCATGTATTACTGTGCGACG, GAAACAGCTCTGGTTGTATCTACTACCTACCTACCACACTACTTTGACAAC, TGGGGCCAGGGAACCCTGGTCACCGTCTCCTCAG, and/or 25 CAGGTGCAGCTGGTACAGTCTGGGGCTGAAGTGAAGAAGCCTGGGTCCTCGGTGATGGTCTC CTGCCAGGCCTCTGGAGGCCCCCTCAGAAACTATATTATCAACTGGCTACGACAGGCCCCTG GACAAGGCCCTGAGTGGATGGGAGGGATCATTCCTGTCTTGGGTACAGTACACTACGCACCG AAGTTCCAGGGCAGAGTCACGATTACCGCGGACGAATCCACAGACACAGCCTACATCCATCT GATCAGCCTGAGATCTGAGGACACGGCCATGTATTACTGTGCGACGGAAACAGCTCTGGTTG 30 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 35 isolated, synthetic or recombinant nucleic acid sequence comprising a heavy 40 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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 5 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, 10 GTTGCATCCAATTTGGAGACA, GGGGTCCCATCAAGGTTCAGTGGAAGTGGATCTGGGACAGATTTTAGTCTCACCATCAGCAG CCTGCAGCCTGAAGATGTTGCAACATATTATTGT, CAACAATATGATAATCTCCCA, CTCACATTCGGCGGAGGGACCAAGGTTGAGATCAAAAGA and/or GACATCCAGATGACCCAGTCTCCATCCTCCCTGTCTGCAGCTGTAGGAGACAGAGTCACCAT 15 CACTTGCCAGGCGAGTCAGGACATTGTCAACTATTTAAATTGGTATCAACAGAAACCAGGGA AAGCCCCTAAGCTCCTGATCTACGTTGCATCCAATTTGGAGACAGGGGTCCCATCAAGGTTC AGTGGAAGTGGATCTGGGACAGATTTTAGTCTCACCATCAGCAGCCTGCAGCCTGAAGATGT TGCAACATATTATTGTCAACAATATGATAATCTCCCACTCACATTCGGCGGAGGGACCAAGG T T GAG AT C AAAAG A, said part having at least 15 nucleotides, is also provided. Said 20 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 25 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 30 sequence GAGGTGCAGCTGGTGGAGTCTGGGGGAGGCGTGGTCCAGCCTGGGAGGTCCCTGAGACTCTC CTGTGCGGCCTCT, GGATTCAGCTTCAGTCACTATGCC, ATGCACTGGGTCCGCCAGGCTCCAGGCAAGGGACTGGAGTGGGTGGCAGTT, ATAT C T TAT GAT G GAGAAAATACA, 35 TATTACGCAGACTCCGTGAAGGGCCGATTCTCCATCTCCAGAGACAATTCCAAGAACACAGT GTCTCTGCAAATGAACAGCCTGAGACCTGAGGACACGGCTCTATATTACTGT, 41 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 GCGAGAGACCGCATAGTGGACGACTACTACTACTACGGTATGGACGTC, TGGGGCCAAGGGGCCACGGTCACCGTCTCCTCAG and/or GAGGTGCAGCTGGTGGAGTCTGGGGGAGGCGTGGTCCAGCCTGGGAGGTCCCTGAGACTCTC CTGTGCGGCCTCTGGATTCAGCTTCAGTCACTATGCCATGCACTGGGTCCGCCAGGCTCCAG 5 GCAAGGGACTGGAGTGGGTGGCAGTTATATCTTATGATGGAGAAAATACATATTACGCAGAC TCCGTGAAGGGCCGATTCTCCATCTCCAGAGACAATTCCAAGAACACAGTGTCTCTGCAAAT GAACAGCCTGAGACCTGAGGACACGGCTCTATATTACTGTGCGAGAGACCGCATAGTGGACG ACTACTACTACTACGGTATGGACGTCTGGGGCCAAGGGGCCACGGTCACCGTCTCCTCA, said part having at least 15 nucleotides. Said heavy chain sequence is preferably 10 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 15 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, 20 AATTTGGAAACAGGGGTCCCATCAAGGTTCAGTGGCAGGGGATCTGGGACAGATTTTACTCT CACCATTAGCAGCCTGCAGCCTGAAGATATTGGAACATATTACTGT, CAACAGTATGATAATCTGCCTCCGCTCACT, TTCGGCGGAGGGACCAAGGTGGAGATCAAAC and/or GACATCCAGATGACCCAGTCTCCATCTTCCCTGTCTGCATCTGTAGGAGACAGAGTCACCAT 25 CACTTGCCAGGCGAGTCAGGACATTAAGAAGTATTTAAATTGGTATCATCAGAAACCAGGGA AAGTCCCTGAGCTCCTGATGCACGATGCATCCAATTTGGAAACAGGGGTCCCATCAAGGTTC AGTGGCAGGGGATCTGGGACAGATTTTACTCTCACCATTAGCAGCCTGCAGCCTGAAGATAT TGGAACATATTACTGTCAACAGTATGATAATCTGCCTCCGCTCACTTTCGGCGGAGGGACCA AGG T G GAG AT CAAAC GAAC T GT G, said part having at least 15 nucleotides, is also 30 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. 35 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%, 42 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 more preferably at least 90% homologous to at least part of the sequence GAGGTGCAGCTGGTGGAGACCGGGGGAGGCCTGGCCCAGCCTGGGGGGTCCCTGAGACTCTC CTGTGCAGCCTCT, GGAT T CACAT T CAGTAGT TATAAC, ATGAACTGGGTCCGCCAGGCTCCAGGGAAGGGGCTGGAGTGGGTCTCACAC, 5 ATTAGTGCGGGTAGTAGTTACATA, TACTACTCAGACTCAGTGAAGGGCCGATTCACCGTCTCCAGAGACAACGTCAGGAACTCAGT ATATCTGCAAATGAACAGCCTGAGAGCCGCTGACACGGCTGTGTATTACTGT, GCGAGAGAGGATTATGGTCCGGGAAATTATTATAGTCCTAACTGGTTCGACCCC, TGGGGCCAGGGAACCCTGGTCACCGTCTCCTCAG and/or 10 GAGGTGCAGCTGGTGGAGACCGGGGGAGGCCTGGCCCAGCCTGGGGGGTCCCTGAGACTCTC CTGTGCAGCCTCTGGATTCACATTCAGTAGTTATAACATGAACTGGGTCCGCCAGGCTCCAG GGAAGGGGCTGGAGTGGGTCTCACACATTAGTGCGGGTAGTAGTTACATATACTACTCAGAC TCAGTGAAGGGCCGATTCACCGTCTCCAGAGACAACGTCAGGAACTCAGTATATCTGCAAAT GAACAGCCTGAGAGCCGCTGACACGGCTGTGTATTACTGTGCGAGAGAGGATTATGGTCCGG 15 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. 20
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 25 CTGCACTGGGAGC, AGCTCCAACATCGGGGCAGGTTATGAT, GTACACTGGTACCAGCAGCTTCCAGGAACAGCCCCCAAACTCCTCATCTAT, GGCAACACT, AATCGGCCCTCAGGGGTCTCCGACCGATTCTCTGGCTCCAAGTCTGGCACCTCAGCCTCCCT GGCCATCACTGGACTCCAGGCTGAGGATGAGGCTGATTATTACTGC, CACTCCTATGACAGAAGCCTGAGTGGT, 30 TCAGTATTCGGCGGAGGGACCAAGCTGACCGTCCTAG and/or CAGTCTGTCGTGACGCAGCCGCCCTCAGTGTCTGGGGCCCCAGGGCAGAGAGTCACCATCTC CTGCACTGGGAGCAGCTCCAACATCGGGGCAGGTTATGATGTACACTGGTACCAGCAGCTTC CAGGAACAGCCCCCAAACTCCTCATCTATGGCAACACTAATCGGCCCTCAGGGGTCTCCGAC CGATTCTCTGGCTCCAAGTCTGGCACCTCAGCCTCCCTGGCCATCACTGGACTCCAGGCTGA 35 GGATGAGGCTGATTATTACTGCCACTCCTATGACAGAAGCCTGAGTGGTTCAGTATTCGGCG GAGGGACCAAGCTGACCGTC, said part having at least 15 nucleotides, is also 43 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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. 5
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 10 CAGGTGCAACTGGTGGAGTCTGGGGGAAATGTGGTCAAGCCTGGGACGTCCCTGAGACTGTC CTGTGCAGCGACT, GGAT T CAACTTCCATAACTACGGC, ATGAACTGGGTCCGCCAGGCTCCAGGCAAGGGGCTGGAGTGGGTGGCGGTT, GTTTGGTATGATGGAAGTAAGAAA, TACTATGCAGACTCCGTGACGGGCCGATTCGCCATCTCCAGAGACAATTCCAAGAACACTCT 15 GTATCTGCAAATGAACAGCCTGAGAGTCGAGGACACGGCTGTTTATTATTGT, GTGAGAGATAAAGTGGGACCGACTCCCTACTTTGACTCC, TGGGGCCAGGGAACCCTGGTCACCGTATCCTCAG and/or GAGGTGCAGCTGGTGGAGTCTGGGGGAAATGTGGTCAAGCCTGGGACGTCCCTGAGACTGTC CTGTGCAGCGACTGGATTCAACTTCCATAACTACGGCATGAACTGGGTCCGCCAGGCTCCAG 20 GCAAGGGGCTGGAGTGGGTGGCGGTTGTTTGGTATGATGGAAGTAAGAAATACTATGCAGAC TCCGTGACGGGCCGATTCGCCATCTCCAGAGACAATTCCAAGAACACTCTGTATCTGCAAAT GAACAGCCTGAGAGTCGAGGACACGGCTGTTTATTATTGTGTGAGAGATAAAGTGGGACCGA CTCCCTACTTTGACTCCTGGGGCCAGGGAACCCTGGTCACCGTCTCGAGT, said part having at least 15 nucleotides. Said heavy chain sequence is preferably derived 25 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 30 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, 35 GACCGGCCCTCAGGGATCCCTGAGCGATTCTCTGGCTCCAACTCTGGGAACACGGCCACCCT GACCATCAGCAGGGTCGAGGCCGGGGATGAGGCCGACTATTACTGT, 44 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 CAGGTGTGGGATAGGAGTAATTATCATCAGGTA, TTCGGCGGAGGGACCAAGTTGACCGTCCTAG and/or TCCTATGTGCTGACTCAGCCCCCCTCGGTGTCACTGGCCCCAGGAGGGACGGCCGCGATCAC CTGTGGAAGAAACAACATTGGAAGTGAAACTGTGCACTGGTACCAGCAGAAGCCAGGCCAGG 5 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, 10 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 15 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 20 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% 25 identical to a heavy chain sequence depicted in Figure 11 A, 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 30 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 35 preferably encodes an amino acid sequence which is at least 80% identical to 45 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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, 5 and/or at least 75% identical to the sequence GIIPVLGTVHYAPKFQG, and/or at least 70% identical to the sequence ETALWSTTYLPHYFDN, 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 10 PVLGTVHYAPKFQGRVTITADESTDTAYIHLISLRSEDTAMYYCATETALWST TYLPHYFDN WGQGTLVTVSS, and/or at least 70% identical to the sequence DIQMTQSPSSLSAAVGDRVTITCQASQDIVNYLNWYQQKPGKAPKLLIYVASN LETGVPSRFSGSGSGTDFSLTISSLQPEDVATYYCQQYDNLPLTFGGGTKVEIK RTV. 15 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 20 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 25 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, EVQLVESGGGWQPGRSLRLSCAASGFSFSHYAMHWVRQAPGKGLEWVAVIS 30 YDGENTYYADSVKGRFSISRDNSKNTVSLQMNSLRPEDTALYYCARDRIVDD YYYY GMD VWGQGATVTV S S, DIQMTQSPSSLSASVGDRVTITCQASQDIKKYLNWYHQKPGKVPELLMHDASNLETGVPSRF SGRGSGTDFTLTISSLQPEDIGTYYCQQYDNLPPLTFGGGTKVEIKRTV, GFTFSSYN, ISAGSSYI, AREDYGPGNYYSPNWFDP, SSNIGAGYD, GNT, HSYDRSLSG,
35 EVQLVETGGGLAQPGGSLRLSCAASGFTFSSYNMNWVRQAPGKGLEWVSHISAGSSYIYYSD 46 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014
SVKGRFTVSRDNVRNSVYLQMNSLRAADTAVYYCAREDYGPGNYYSPNWFDPWGQGTLVTVS S, QSVVTQPPSVSGAPGQRVTISCTGSSSNIGAGYDVHWYQQLPGTAPKLLIYGNTNRPSGVSD RFSGSKSGTSASLAITGLQAE DEADYYCHSYDRSLSGSVFGGGTKLTV, GFNFHNYG, 5 VWYDGSKK, VRDKVGPTPYFDS, NIGSET, DDD, QVWDRSNYHQV, EVQLVESGGNWKPGTSLRLSCAATGFNFHNYGMNWVRQAPGKGLEWVAWWYDGSKKYYAD SVTGRFAISRDNSKNTLYLQMNSLRVEDTAVYYCVRDKVGPTPYFDSWGQGTLVTVSS, and SYVLTQPPSVSLAPGGTAAITCGRNNIGSETVHWYQQKPGQAPVLWYDDDDRPSGIPERFS GSNSGNTATLTISRVEAGDEADYYCQVWDRSNYHQVFGGGTKLTV. 10 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 15 the present invention are particularly suitable for expressing an antibody or a functional part, derivative or analogue thereof according to the invention, preferably D25, AM 14, AM 16, 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 20 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 25 invention. 47 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014
Examples MATERIALS AND METHODS Maintenance and isolation of human B cells 5 Using standard procedures, CD 19 positive human B cells were isolated from bloodbank derived huffy coat (other sources can be fresh blood with an anticoagulation 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 10 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 CD 19, 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 15 and IgD were then sorted using the FACSAria (BD) (Figure 1). Besides memory B cells, other B cells subsets, like naive, naive, 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. 20
Cell Culture
Sorted cells were washed and cultured in 24 well plates (1.5 to 2xl05cells/ml) on 80 Gray, irradiated CD40L-expressing L-cells (5xl04 cells/ml; provided by DR. J. 25 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 CD40L-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 36 30 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. 35 48 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014
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). Bcl-6 was identified in a senescence rescue 5 screen in murine fibroblasts as an inhibitor of anti-proliferative pl9ARF-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 10 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-15 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 20 (Roche Diagnostics Netherlands, Almere, Netherlands) according to manufacturers protocols. Two days later selection of transfected cells started by the addition of 2 pg/ml puromycin (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 25 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. 30
Retroviral Transduction
The recombinant human fibronectin fragments CH-296 transduction procedure (RetroNectin™; Takara, Otsu, Japan) was performed as described previously (Heemskerk et al., 1997; Heemskerk et al., 1999). Non-tissue culture-treated 24 35 wells plates (Costar, Badhoevedorp, Netherlands) were coated with 0.3 ml of 30 49 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 μg/ml 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 5 buffered saline (PBS) for 30 min at room temperature, followed by washing once with PBS. 5xl05 B cells, which were prepared for retroviral transduction were plated in 0.25 ml RPMI without FCS and L-cells and mixed with 0.25 ml of thawed retroviral supernatant. For the Bcl-6 Bcl-XL double transduction 125 μΐ of Bcl-6-IRES-NGFR (or IRES-YFP) (Shvarts A. et al. Genes Dev., 2002) and 125 10 μΐ 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 15 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, 20 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. 25 Flowcytometry
Antibodies against the human molecules IgD, IgG, CD3, CD 19, CD20, CD27, CD38, CD40, CD45, CD56, CD70, CD80, CD86, HLA-DR (BD) directly labeled with FITC, PE, PERCP, PE-Cy5, APC or APC-Cy7 and IgM, kappa light chain, lambda light chain, CD 138, directly labeled with PE (DAKO) were used for 30 flowcytometry analysis. Stained cells were analyzed using a LSRII (BD) and FACS data was processed with Flow Jo computer software (Tree Star, Inc).
Proliferation experiment
Naive and memory B cells were isolated from fresh PBMC on the FACSAria: 35 Naive B cells: CD19-Pe-Cy7 pos, CD27-APC neg, IgD-ΡΕ pos 50
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 2μΜ 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 5 washing the cell with cold FCS. Cells were resuspended in 500μ1 IMDM-8% FCS and 2014262169 15 Jan 2015 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 Bcl-6-IRES-NGFR, cultured for 3 10 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 15 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. 20
Determining the B cell receptor (BCR) expression alter long term culture of Bcl-6 and Bcl-XL transduced cells
It is disclosed 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 25 with Bcl-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 FACS Aria (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 30 the FACS Canto (BD). Therefore cells were harvested and stained in 96-well plates with GFP, NGFR, CD 19 and TT-PE. 51 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014
Development of Bcl-6 and Bcl-XL double positive B cell lines that secrete antibodies B cell lines were created that produces monoclonal antibodies and are 100% Bcl-6 and Bcl-XL double positive. First this was achieved by inducing proliferation and 5 differentiation using IL-21. Meanwhile these cells are transduced with the Bcl-6-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). 10 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 15 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 20 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 (FACS Aria). 25 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 30 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 35 supernatant was carefully removed and 15 ml medium containing 1% FCS was 52 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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 μιη) and spin at 1450 rpm at RT before samples were snap frozen and stored in liquid nitrogen. A 5 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 10 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 15 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/ml (50 μΐ) in ELISAs.
Determining TCID50 and PFU of RSV stocks
To determine the TCID50, 104 HEp2 were seeded in 96 well plates and infected 20 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 25 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 TCID50. 30 To determine the amount of plaque forming units (PFU) of the virus, lxl05/ml 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 μΐ) 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 35 cells through the culture medium. Thereby the virus can infect only neighboring 53 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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 5 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 10 (donor B62 and B63). Before sorting CD19poTgMnesIgDnesIgAnesCD27pos 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 Bcl-6-IRES-NGFR only. After 12h cells were harvested and cultured for 3 15 days in the presence of IL-4 before NGFR expressing cells were sorted using MACS beads (Miltenyi) and immediately transduced with Bcl-XL-IRES-GFP. The B cells that did not bind to the MACS beads were washed and transduced with Bcl-6 and Bcl-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 20 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 Bcl-6 and Bcl-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, 104 HEp2 cells are seeded 25 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 μΐ and the RSV end concentration is 0.1 MOI. After lh the virus supernatant mixture is 9 times diluted with PBS and replaced with 100 μΐ 30 IMDM/5%FCS. After 2 days cells are fixed with 80% acetone and stained with polyclonal anti-RSV-HRP (Biodesign). Using H2O2 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. 35 54 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 RT-PCR and cloning of VH and VL regions
Total RNA was isolated from ~5xl05 B cells with the RNeasy® mini kit (Qiagen, Venlo, The Netherlands). 250 ng of total RNA was reverse transcribed in a 5 volume of 20 μΐ containing IX first strand buffer, 500 μΜ 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 μΐ of cDNA was subjected to PCR in a 50 μΐ solution containing 20 mM Tris-HCL, 50 mM KCL, 2.5 mM MgC12, 250 μΜ dNTP, 1 U AmpliTaq Gold DNA polymerase 10 (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 15 was performed using BigDye Terminator chemistry (Applied Biosystems Inc.) and Vector-NTI software (Invitrogen).
To rule out reverse transcriptase and/or DNA polymerase induced mutations, several independent cDNA conversions and PCR reactions were performed and individually cloned and sequence analyzed. Consensus sequences were 20 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 25 chain leader sequence and VH region of clone D25 introducing a 5’-NheI site and a 3’-XhoI site. The IgGl constant region (CHl-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 Nhel/Notl digested pCDNA3.1(+)Zeo. The full length light chain expression 30 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 Nhel/Notl digested pCDNA3.1(+)Zeo to obtain a full length light chain expression vector.
Sequence analysis was performed to confirm correctness of the expression 35 constructs. 55 WO 2008/147196 PCT/NL2008/050333 2014262169 11 Nov 2014 10
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: VHl-For 5'-AAATCGATACCACCATGGACTGGACCTGGAGG-3' VHIB-For 5'-AAATCGATACCACCATGGACTGGACCTGGAGM-3' -AAATCGATACCACCATGGACACACTTTGCTMCAC-3' -AAATCGATACCACCATGGACATACTTTGTTCCAAC-3 -AAATCGATACCACCATGGAGTTTGGGCTGAGC-3' -AAATCGATACCACCATGGARYTKKGRCTBHGC-3' -AAATCGATACCACCATGAAACACCTGTGGTTCTT-3' -AAATCGATACCACCATGGGGTCAACCGCCATC-3' -AAATCGATACCACCATGTCTGTCTCCTTCCTC-3' VH2A-For 5' VH2B-For 5' VH3-For 5' VH3B-For 5' 15 VH4-For 5' VH5-For 5' VH6- For 5' Cgamma-Rev 5 ' 20 Vkaooa reaions: Vkl-For 5' VklB-For 5' Vk2-For 5' Vk3-For 5' 25 Vk4-For 5' Ck-Rev 5' Vlambda reaions: 30 Vllaecb 5' Vllg 5' V12/10 5' V13jpah 5' V15/7 5' 35 V16/9 5' V13rm 5' V131 5' -AAATCGATACCACCATGGACATGAGGGTCCCY-3' -AAATCGATACCACCATGGCCTGGGCTCCTCTCCTTC-3' -AAATCGATACCACCATGGCCTGGATCCCTCTCCTCC-3' -AAATCGATACCACCATGGCCTGGACCCCTCTCTGGC-3' 56 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 V13e 5'-AAATCGATACCACCATGGCCTGGGCCACACTCCTGC-3' VI4c 5'-AAATCGATACCACCATGGCCTGGGTCTCCTTCTACC-3' VI8a 5'-AAATCGATACCACCATGGCCTGGATGATGCTTCTCC-3' C12/7 5'-GATCGCGGCCGCTTATCAWGARCATTCTGYAGGGGCCACTG-3' 5
The oligonucleotides used for expression vector constructions were:
Heavy chain expression vector: VHl-L-Nhel: 5'-GCGGCTAGCCACCATGGACTGGACCTGGAGG-3' 10 JH4/5-XhoI: 5'-GCGCTCGAGACGGTGACCAGGGTTCCCTG-3' CHfw-XhoI: 5'-CGCGCTCGAGTGCCTCCACCAAGGGCCCATCGGTC-3' CHrev-Notl: 5'-GATCGCGGCCGCTTATCATTTACCCGGRGACAGGGAGAGGC- 3' 15 Light chain expression vector: VKl-L-Nhel: 5'-GCGGCTAGCCACCATGGACATGAGGGTCCCY-3' CK-Notl: 5'-GATCGCGGCCGCTTATCAACACTCTCCCCTGTTGAAGCTCTT- 3' 20
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 25 cycles of 30s at 94°C, 30 s at 62°C (HPRT1), 52°C (LMP-1) and 58°C (EBNAl/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’-GACACAAACATGATTCAAATCC CTGA- 3'); LMP-1 forward: (5'-30 GCGACTCTGCTGGAAATGAT-3') and LMP-1 reverse (5'-GACATGGTAATGCCTAGAAG-3'); EBNAl/2 forward (5'-AGCAAGAAGAGGAGGTGGTAAG-3') and EBNAl/2 reverse (5'-GGCTCAAAGTGGTCTCTAATGC-3').
In addition to the RT-PCR we performed a PCR directly on cell pellet and 35 supernatant DNA that was isolated using the QIAmp isolation kit (Qiagen). 57 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 EXAMPLE 1 RESULTS B cell phenotype 5 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 10 on genetic modifications of human B cells. We studied downstream targets of STAT5. One target besides others is Bcl-6. Bcl-6 inhibits differentiation of B cells to plasma cells that are arrested in proliferation. Overexpression of Bcl-6 keeps BLIMP1 in balance, a transcription factor which expression is strongly enhanced by stimulating B cells with IL-21 (works via STAT3). BLIMP1 is necessary to 15 induce the development of Ig producing cells (CD20-CD38+) whereas Bcl-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 20 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 Bcl-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 25 identical to memory B cells after another 3 days of culture (data not shown).
Next we show that Bcl-6, together with Bcl-XL (anti-apoptotic downstream target of STAT5), CD40L signaling and in the presence of IL-21, maintain human IgG memory B cells in the CD20+CD38dull phenotype for long periods of time (>3 30 months) (Figure 3). In addition, the Bcl-6 Bcl-XL B cells have a phenotype corresponding to activated B cells (see Table 1, exemplified by FACS staining of 3 TT+ B cell clones), since these cells have high expression of CD80, CD86 and HLA-DR. 58 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 determined on three different Bcl-6 Bcl-XL B cell clones cultured with IL-21 and CD40L 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 CD 154 (CD40L) 8%pos CD28 neg ICOSL pos CD30 pos(56-74°/o) IgM neg CD38 pos/ i ntermediate IgG pos CD40 pos HLA-DR pos(high) CD44 pos Kappa pos/neg CD45 pos Lambda pos/neg CD45RA pos/high IL21-R pos
Antibody membrane expression 5 The Bcl-6 Bcl-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. 10 This was confirmed by single cell sorting of Bcl-6 and Bcl-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 15 instance in screening assays, the B cells are preferably transduced with Bcl-6 and Bcl-XL and not infected with EBV.
Cell division and growth curves
Bcl-6 Bcl-XL transduced B cells divide on average 0.6 times per day. Division rate 20 varies between donors and cell density of the cultures (Figure 5a). The anti-RSV 59 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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 Bcl-6 Bcl-XL B cells 5 The Bcl-6 Bcl-XL transduced B cells secrete on average one pg/ml 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. 10 Determine EBV content EBV RT-PCR on mRNA of Bcl-6 Bcl-XL cell lines that were cultured with IL-21 and CD40L signaling. In the cell lines obtained with this immortalization technique no EBV gene transcript have ever been detected (data not shown). 15 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 Bcl-6 and Bcl-XL by FACS or Magnetic Bead 20 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 Bcl-6 Bcl-XL transduced memory (or any other) B cells at low cell densities (for example 100 cells/well). Supernatants from these lOOc/w cultures can be collected and tested for their specificity. 100 cell/well cultures that are 25 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 BCR on the FACSAria or they were selected by ELISA screening of series of cultures till the single anti-TT monoclonal cell line 30 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 35 anti-RSV antibody which we cloned by limiting dilution culture. One of the 60 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 monoclonal antibodies, D25 was used to continue studies. D25, a monoclonal antibody with an IgGl 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 IC50 value of between 0.5 and 1.5 ng/ml 5 (± ΙΟρΜ) whereas the IC50 of the standard anti-RSV antibody used in the clinic (palivizumab developed by Medlmmune) is 0.453 pg/ml (3.02nM) (H. Wu et al. 2005 J.Mol.Biol. and A. Mejias et al. 2005 Antimicrob. Agents Chemother.) (Figure 8). 10 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 μΐ culture supernatant. Cells were washed and stained with 15 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. 20 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 25 versus palivizumab, VSV-F protein infected EL-4 cells were incubated with increasing amounts of D25 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, D25 binding was not affected (data not shown). 30
Figure 9c shows the binding of Palivizumab (Synagis) and D25 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. 35 61 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014
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 5 different then the epitope recognized by palivizumab. This is illustrated in figure 10 in which D25 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 μg/ml, therefore we should be able to obtain enough purified antibody to perform pre-clinical (animal) studies. The purification is performed 15 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 lipofectamine LTX (Invitrogen). The amount of IgG that were present in the supernatant was 20 approximately 22 pg/ml (total volume 50 ml). This antibody derived from the cloned nucleotide sequence of the antibody expressed by the D25 B cell line did also recognized infected HEp2 cells (data not shown).
Antibody sequence 25 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 (Vhl-69) and light (Vkl 08/018) chain sequences were determined. The whole antibody sequence was cloned by using TOPO vectors and after sequence control, subcloned into the pCDNA3.1 mammalian protein 30 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 Vhl-69 that must have occurred during affinity maturation and further B cell selection. 35 62 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014
To summarize, we here show the isolation, characterization and long-term culture of human memory B cells using the transgenes Bcl-6 and Bcl-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 5 origin, they can readily be deployed as a therapeutic medicine. 63 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 EXAMPLE 2
The D25 heavy and light chain were cloned into standard expression vectors as described before (p44 ‘antibody sequence’). To create an expression construct that 5 allows for maximum protein expression the D25 heavy and light chain sequences were codon optimized by GENE ART (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 10 sequence was optimized but the amino acid sequence remained unchanged.
Shown in EXAMPLE 4 is the neutralizing capacity of purified B cell supernatant derived D25, recombinant D25 and GENEART optimized D25. All efficiently neutralize RSV.
The GENEART modifications compared to the original D25 sequence are 15 depicted in Figure 12. 64 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 EXAMPLE 3
Next to the in vitro RSV neutralization experiments we tested the D25 5 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;pl811, Johnson S et al., JID 1997;pl215 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 10 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 ΙΟΟμΙ antibodies 15 Day 0.1.N infection lxlO7 pfu RSV A2 in 50μ1
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 2.0ml BAL via trachea canule 20 Collect lungs
Immediately start TCID50 on BAL material (1 ml)
Freeze 1ml BAL material (ELISA cytokine/RT-PCR) -80C Perform TCID50 on prepared long material (1ml)
25 Freeze 1 ml long material (ELISA cytokine/RT-PCR) -80C Collect/spin blood for hlgG ELISA on serum en store at -80C
The results are shown in Figure 13: 30 (A) One day before RSV challenge (lxlO7 RSV-A2 particles) by nasal spray, animals were IP injected with different amounts of Synagis (Medlmmune), purified D25 or an IgGl 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 35 13D depicts virus titers found in lung lavages (BAL) at day 5 in treated and 65 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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. 5
Conclusion/Result:
An estimate of the D25 half-live is 5 to 9 days based on the (linear) calculation that 60 and 30μg of antibody was injected on day 0 (2 and 1 mg/kg respectively) and at day 5 33 or 16pg was detected (total volume of mice 1,5). When we started 10 with 0,5mg/kg injection per animal on dO then Ig levels drop from 15pg to llgg on day 5, which would indicate a 9 day half life (Table 4).
Table 4 mg/kg total administered dO ^g) detected on d5 ^g) half-life (days) 2,0 60 33 5,6 1,0 30 16 5,4 0,5 15 11 9,4 15
Virus titer as determined TCID50 assay shows that in control animals lxlO4 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 20 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. 25 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 D25 treated animals seem to have less cellular infiltration into the lungs and around the bronchi. 66 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014
To test D25 in the Cottonrats, experiments are set up to compare animals pretreated with Synagis and D25 before challenge with the RSV-X virus at the NVI (Bilthoven, Netherlands). 5 EXAMPLE 4 10 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 15 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 20 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 IgGl isotype. AM14 has a Kappa light 25 chain, while AM 16 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. 30 The results are shown in Figures 15-1 and 15-11: 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 15-1) Vero and 35 (Figure 15-11) Hep2 cells with different antibodies: A2 (A), X (B) and 2006/1 (C) 67 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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 lOOul Vero or HEp2 cells (lxl06/ml) were added. Virus antibody mixture was not washed away. After three days 5 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, 10 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 15 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, 20 although still comparable to Synagis. Table 5 IC50 values (ng/ml) Cell line used r «η* subtype rD25 rD25 GA AM14 AM16 AM23 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 IC50 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 D25 rD25 GA = supernatant of 293T cells with GENEART codon optimized recombinnat D25 25 68 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 EXAMPLE 5
Synergistic and blocking effects of anti-RSV antibodies. 5 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 10 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 15 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 AM14. That indicates that 20 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. 25 69 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014
Brief description of the drawings
Figure 1.
Isolation of human, IgG positive, memory B cells. PBMC isolated from huffy coat 5 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 CD 19, 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). 10
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 Bcl-6-IRES-NGFR. Cells were kept an additional 3 days on IL-21 before CFSE content was 15 determined. The CFSE dye is diluted with every cell division.
Figure 3
An example of human B cells transduced with Bcl-6 and Bcl-XL or Bcl-XL only. Cells were maintained on irradiated L cells expressing CD40L and the cytokine 20 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 CD38dullCD20+ staining indicates memory or germinal center B cells; the CD38+CD20' staining indicate plasmablasts. 25
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 Bcl-6-IRES-NGFR and Bcl-XL-IRES-GFP. Cells expressing NGFR, GFP and were 30 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). 35 70 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014
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. 5
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 10 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 15 To determine the light chain phenotype of the D25 anti-RSV clone, the D25 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. 20 Figure 8
From donor B63, 100 cell/well cultures were grown using Bcl-6 Bcl-XL positive human memory B cells. One of those cultures, D10 showed strong neutralization. LD derived monoclonal cell lines were made, one D25 neutralized the RSV A-2 virus efficiently. Shown here is D25 compared to palivizumab (synagis) and a 25 polyclonal goat ant-RSV. Not shown are irrelevant culture supernatants of Bcl6 Bcl-XL transduced B cell clones cultured with IL-21 and CD40L signaling that produce high levels of antibodies but did not block RSV infection The D25 clone was used for further characterization. 30 Figure 9
In figure 9a: HEp2 cell were seeded at 10-12e6 cells per T175 flask (Nunc) in IMDM/5%FCS. The next day the medium was replaced with 5ml 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 35 replaced with IMDM/1%FCS and cultured o/n with a closed lid at 37°C. The next 71 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 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 5 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 10 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 (MFI). 15 Figure 10
Binding of polyclonal goat anti-RSV (pos Ctrl), palivizumab (synagis) and D25 to coated HEp2 infected cell lysate.
Figure 11 20 Sequence analysis of the D25 clone. 11a shows nucleotide and predicted amino acid sequence of the variable heavy and light chain domains, llb/c show the D25 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. 25
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 30 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 35 Synagis. (A) One day before RSV challenge (lxlO7 RSV-A2 particles) by nasal 72 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014 spray, animals were IP injected with different amounts of Synagis (Medlmmune), purified D25 or an IgGl 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 5 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. 10 Figure 14
Nucleotide and amino acid sequences of three new potent RSV neutralizing antibodies (A) AM14, (B) AM16 and (C) AM23.
Figure 15
15 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 15-1) Vero and (figure 15-11) Hep2 cells with different antibodies A2 (A), X 20 (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 lOOul Vero or HEp2 cells (lxl06/ml) were added. 25 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. 73 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014
References
Banchereau, J. , de Paoli, P. , Valle, A. , Garcia, E. , Rousset, F., (1991). Long 5 term human B cell lines dependent on interleukin-4 and antibody to CD40, Science 251, 70-2.
Boise, L. Η., 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 bcl-2-related gene 10 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, 15 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 20
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 25 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, 30 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;pl811, Johnson S 35 et al., JID 1997;pl215 74 PCT/NL2008/050333 WO 2008/147196 2014262169 11 Nov 2014
Wu H et al., JMB 2007:p652
Shvarts A. et al, 2002: Genes Dev. Vol 16, page 681-686 5
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, 10 871-875.
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 protooncogene controls germinal-centre formation and Th2-type inflammation. Nat 15 Genet 16, 161-170. 75

Claims (32)

  1. The claims defining the invention are as follows:
    1. An isolated, synthetic or recombinant antibody or a functional part, derivative or analogue thereof which specifically binds Respiratory Syncytial Vims (RSV) and which comprises: a variable heavy chain sequence comprising a heavy chain complementarity determining region CDR1 comprising the amino acid sequence GFSFSHYA (SEQ ID NO:73), a heavy chain CDR2 sequence comprising the amino acid sequence ISYDGENT (SEQ ID NO:74), and a heavy chain CDR3 sequence comprising the amino acid sequence ARDRIVDDYYYYGMDV (SEQ ID NO:75), and a variable light chain sequence comprising a light chain CDR1 comprising the amino acid sequence QDIKKY (SEQ ID NO:76), a light chain CDR2 sequence comprising the amino acid sequence DAS, and a light chain CDR3 sequence comprising the amino acid sequence QQYDNLPPLT (SEQ ID NO:77).
  2. 2. An antibody, functional part, derivative or analogue thereof according to claim 1, having a heavy chain sequence comprising a sequence which is at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% identical to the sequence E V QLVES GGG V V QPGRS LRLS C A AS GFS FS Η Y AMHW VRQ APGKGLEW V AVIS YDGE NT Y Y ADS VKGRFS IS RDN S KNT V S LQMN S LRPEDT ALY Y C ARDRIVDDYYYY GMDV WGQGATVTVSS (SEQ ID NO: 78) and/or having a light chain sequence which is at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% identical to the sequence DIQMTQSPSSLSASVGDRVTITCQASQDIKKYLNWYHQKPGKVPELLMHDASNLETG VPSRFSGRGSGTDFTLTISSLQPEDIGTYYCQQYDNLPPLTFGGGTKVEIKRTV (SEQ ID NO: 79).
  3. 3. The antibody, functional part, derivative, or analogue thereof of claim 1 or claim 2, wherein the antibody, functional part, derivative, or analogue thereof comprises a variable heavy chain sequence comprising the amino acid sequence EVQLVESGGGVVQPGRSLRLSCAASGFSFSHYAMHWVRQAPGKGLEWVA VIS YDGE NT YY ADS VKGRFSISRDN S KNTV S LQMN S LRPEDT ALYY C ARDRIVDDYYYY GMDV WGQGATVTVSS (SEQ ID NO:78).
  4. 4. The antibody, functional part, derivative, or analogue thereof of any one of claims 1 to 3, wherein the antibody or functional part thereof comprises a variable light chain sequence comprising the amino acid sequence DIQMTQSPSSLSASVGDRVTITCQASQDIKKYLNWYHQKPGKVPELLMHDASNLETG VPS RF SGRGSGTDFTLTISSLQPEDIGTYYCQQYDNLPPLTFGGGTKVEIKRTV (SEQ ID NO:79).
  5. 5. The antibody, functional part, derivative, or analogue thereof of claim 1, wherein the antibody, functional part, derivative, or analogue thereof comprises a variable heavy chain sequence comprising the amino acid sequence E V QFVES GGG V V QPGRS FRFS C A AS GFS FS Η Y AMHW VRQ APGKGFEW V A VIS YD GENT Y Y ADS VKGRFS IS RDN S KNT V S LQMN S LRPEDT ALY Y CARD RIVDDYYYYGMDVWGQGATVTVSS (SEQ ID NO:78) and a variable light chain sequence comprising the amino acid sequence DIQMTQSPSSLSASVGDRVTITCQASQDIKKYLNWYHQKPGKVPELLMHDASNLETG VPSRFS GRGS GTDFTLTIS S LQPEDIGT YYCQQ YD NLPP LTFGGGTKVEIKRTV (SEQ ID NO:79).
  6. 6. An antibody, functional part, derivative, or analogue thereof of any one of claims 1 to 5 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.
  7. 7. An antibody, functional part, derivative, or analogue thereof of any one of claims 1 to 6 comprising an IgM heavy chain constant region or an IgG heavy chain constant region.
  8. 8. An antibody, functional part, derivative, or analogue thereof of any one of claims 1 to 6 wherein the heavy chain comprises the IgGl isotype and/or the light chain comprises the kappa isotype.
  9. 9. The antibody, functional part, derivative or analogue thereof according to any one of claims 1 to 8, which is a human antibody or a chimeric antibody.
  10. 10. The antibody, functional part, derivative, or analogue thereof of any one of claims 1 to 9, wherein the functional part thereof is a single domain antibody, a single chain antibody, a single chain variable fragment (scFv), a Fab fragment, or a F(ab') fragment.
  11. 11. The antibody, functional part, derivative, or analogue thereof of any one of claims 1 to 10, wherein the antibody is a monoclonal antibody.
  12. 12. An isolated, synthetic or recombinant nucleic acid sequence encoding an antibody or, functional part thereof according to any one of claims 1 to 11.
  13. 13. A nucleic acid sequence according to claim 12, wherein: the nucleic acid sequence encoding the heavy chain CDR1 amino acid sequence GFSFSHYA (SEQ ID NO:73) comprises SEQ ID NO:95, the nucleic acid sequence encoding the heavy chain CDR2 amino acid sequence ISYDGENT (SEQ ID NO:74) comprises SEQ ID NO:97, the nucleic acid sequence encoding the heavy chain CDR3 amino acid sequence ARDRIVDDYYYYGMDV (SEQ ID NO:75) comprises SEQ ID NO:99, the nucleic acid sequence encoding the light chain CDR1 amino acid sequence QDIKKY (SEQ ID NO:76) comprises SEQ ID NO: 103, the nucleic acid sequence encoding the light chain CDR2 amino acid sequence DAS comprises the nucleotide sequence GATGCATCC, and/or the nucleic acid sequence encoding the light chain CDR3 amino acid sequence QQYDNLPPLT (SEQ ID NO:77) comprises SEQ ID NO: 106
  14. 14. The isolated, synthetic or recombinant nucleic acid sequence of claim 12, wherein the nucleic acid sequence comprises a. a variable heavy chain nucleotide sequence comprising the nucleotide sequence of SEQ ID NO: 101; b. a variable light chain nucleotide sequence comprising the nucleotide sequence of SEQ ID NO: 108; or c. a variable heavy chain nucleotide sequence comprising the nucleotide sequence of SEQ ID NO: 101 and a variable light chain nucleotide sequence comprising the nucleotide sequence of SEQ ID NO: 108.
  15. 15. The nucleic acid sequence of any one of claims 12 to 14, wherein the nucleic acid sequence comprises a cDNA sequence.
  16. 16. The nucleic acid sequence of any one of claims 12 to 15, wherein the nucleotide sequence has been codon optimized to maximize translation into protein.
  17. 17. A vector comprising the nucleic acid sequence of any one of claims 12 to 16.
  18. 18. A vector comprising an isolated, synthetic or recombinant nucleic acid sequence encoding an antibody, functional part, analogue, or derivative thereof according to any one of claims 1 to 11.
  19. 19. An isolated mammalian cell comprising the nucleic acid sequence of any one of claims 12 to 16 or the vector of claim 17 or 18.
  20. 20. A method for producing an antibody or a functional part thereof, the method comprising culturing the cell of claim 19 in vitro, and obtaining said antibodies or functional parts thereof produced by the cell.
  21. 21. Use of a nucleic acid sequence according to any one of claims 12 to 16 or the vector of claim 17 or 18 for the preparation of a medicament for treating and/or preventing a RSV-related disorder, or preventing or counter-acting the adverse effects of an RSV infection in a human subject.
  22. 22. An isolated antibody producing cell capable of producing an antibody, functional part, derivative or analogue thereof according to any one of claims 1 to 11 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.
  23. 23. 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 wherein BCL6 and Blimp-1 are co-expressed; and/or 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.
  24. 24. 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, functional part, derivative or analogue thereof according to any one of claims 1 to 11.
  25. 25. The method of claim 24, wherein the medicament is formulated for administration at a dosage of 0.1 to 10 mg/kg of the body weight of the human subject.
  26. 26. The method of claim 24 or claim 25, wherein the human is a subject having a chronic lung disease, congenital heart disease or compromised immunity, or the human is a child younger than 6 weeks old or an elderly subject.
  27. 27. A composition for use in treating or preventing a RSV-related disorder, or preventing or counteracting the adverse effects of an RSV infection in a human subject, wherein the composition comprises a therapeutically effective amount of the antibody, functional part, derivative, or analogue thereof of any one of claims 1 to 11, and a pharmaceutically acceptable carrier, diluent or excipient.
  28. 28. The composition of claim 27 further comprising a second antibody or functional part thereof which is capable of specifically binding respiratory syncytial vims (RSV) F antigen and wherein the second antibody comprises: a variable heavy chain sequence comprising a heavy chain complementarity determining region CDR 1 comprising the amino acid sequence NYIIN (SEQ ID NO: 1), a heavy chain CDR2 comprising the ammo acid sequence GIIPVLGTVHYAPKFQG (SEQ ID NO: 2), a heavy chain CDR3 comprising the amino acid sequence ET-ALVVSTTYLPHYFDN (SEQ ID NO: 3), and a variable light chain sequence comprising a light chain CDR1 comprising the amino acid sequence QASQDIVNYLN (SEQ ID NO: 4), a light chain CDR2 comprising the amino acid sequence VASNLET (SEQ ID NO: 5), and a light chain CDR3 comprising the amino acid sequence QQYDNLP (SEQ ID NO: 6).
  29. 29. 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 a composition of claim 27 or claim 28.
  30. 30. The method of claim 29, wherein the human is a subject having a chronic lung disease, congenital heart disease or compromised immunity, or the human is a child younger than 6 weeks old or an elderly subject.
  31. 31. An isolated antibody producing cell producing an antibody, functional part, derivative, or analogue thereof, wherein the antibody, functional part, derivative, or analogue thereof specifically binds Respiratory Syncytial Vims (RSV), comprises a heavy chain comprising a heavy chain CDR1 sequence comprising the sequence GFSFSHYA (SEQ ID NO:73), a heavy chain CDR2 sequence comprising the sequence ISYDGENT (SEQ ID NO:74), and a heavy chain CDR3 sequence comprising the sequence ARDRIVDDYYYYGMDV (SEQ ID NO:75), and comprises a light chain comprising a light chain CDR1 sequence comprising the sequence QDIKKY (SEQ ID NO:76), a light chain CDR2 sequence comprising the sequence DAS, and a light chain CDR3 sequence comprising the sequence QQYDNLPPLT (SEQ ID NO:77); and wherein the heavy chain and the light chain are encoded by exogenous nucleotide sequences.
  32. 32. A method for producing an antibody, functional part, derivative or analogue thereof, the method comprising culturing the isolated antibody producing cell of claim 31 in vitro and obtaining said antibodies, functional part, derivative or analogue thereof produced by said cell.
AU2014262169A 2007-06-01 2014-11-11 Rsv-specific binding molecules and means for producing them Active AU2014262169B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2014262169A AU2014262169B2 (en) 2007-06-01 2014-11-11 Rsv-specific binding molecules and means for producing them

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP07109472.6 2007-06-01
EP07109472A EP1997830A1 (en) 2007-06-01 2007-06-01 RSV specific binding molecules and means for producing them
AU2008257801A AU2008257801B8 (en) 2007-06-01 2008-05-30 RSV-specific binding molecules and means for producing them
AU2014262169A AU2014262169B2 (en) 2007-06-01 2014-11-11 Rsv-specific binding molecules and means for producing them

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2008257801A Division AU2008257801B8 (en) 2007-06-01 2008-05-30 RSV-specific binding molecules and means for producing them

Publications (2)

Publication Number Publication Date
AU2014262169A1 AU2014262169A1 (en) 2014-11-27
AU2014262169B2 true AU2014262169B2 (en) 2017-02-16

Family

ID=51946419

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2014262169A Active AU2014262169B2 (en) 2007-06-01 2014-11-11 Rsv-specific binding molecules and means for producing them

Country Status (1)

Country Link
AU (1) AU2014262169B2 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005079479A2 (en) * 2004-02-17 2005-09-01 Absalus, Inc. Super-humanized antibodies against respiratory syncytial virus

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005079479A2 (en) * 2004-02-17 2005-09-01 Absalus, Inc. Super-humanized antibodies against respiratory syncytial virus

Also Published As

Publication number Publication date
AU2014262169A1 (en) 2014-11-27

Similar Documents

Publication Publication Date Title
US10730931B2 (en) RSV-specific binding molecules and means for producing them
US10273454B2 (en) Means and methods for influencing the stability of antibody producing cells
AU2014262169B2 (en) Rsv-specific binding molecules and means for producing them

Legal Events

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