OA19273A - Stabilized pre-fusion RSV F proteins. - Google Patents

Stabilized pre-fusion RSV F proteins. Download PDF

Info

Publication number
OA19273A
OA19273A OA1201800460 OA19273A OA 19273 A OA19273 A OA 19273A OA 1201800460 OA1201800460 OA 1201800460 OA 19273 A OA19273 A OA 19273A
Authority
OA
OAPI
Prior art keywords
rsv
protein
fusion
seq
amino acid
Prior art date
Application number
OA1201800460
Inventor
Petrus Maria Langedijk Johannes
Original Assignee
Janssen Vaccines & Prevention B.V.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Janssen Vaccines & Prevention B.V. filed Critical Janssen Vaccines & Prevention B.V.
Publication of OA19273A publication Critical patent/OA19273A/en

Links

Abstract

The present invention provides stable pre-fusion respiratory syncitial virus (RSV) F proteins, immunogenic compositions comprising said proteins and uses thereof for the prevention and/or treatment of RSV infection.

Description

Janssen Vaccines & Prévention B.V.
Stabilized pre-fusion RSV F proteins
The présent invention relates to the field of medicine. The invention in particular relates to recombinant pre-fusion RSV F proteins, to nucleic acid molécules encoding the
RSV F proteins, and uses thereof, e.g. in vaccines.
Background of the invention ! ! I l ( 10 After discovery of the respiratory syncytial virus (RSV) in the 1950s, the virus soon became a recognized pathogen associated with lower and upper respiratory tract infections in humans. Worldwide, it is estimated that 64 million RSV infections occur each year resulting f in 160.000 deaths (WHO Acute Respiratory Infections Update September 2009). Tlje most severe disease occurs particularly in prématuré infants, the elderly and immunocompromised individuals. In children younger than 2 years, RSV is the most common respiratory tract pathogen, accounting fofapproximately 50% of the hospitalizations due to respiratory infections, and the peak of hospitalization occurs at 2-4 months of âge. It has been reported that almost ail children hâve been infected by RSV by the âge of two. Repeated infection during lifetime is attributed to ineffective natural immunity. In the elderly, the RSV disease burden is similar to those caused by non-pandemic influenza A infections.
RSV is a paramyxovirus, belonging to the subfamily of pneumovirinae. Its genome encodes for various proteins, including the membrane proteins known as RSV Glycoprotein (G) and RSV fusion (F) protein which are the major antigenic targets for neutralizing antibodies. Antibodies against the fusion-mediating part of the Fl protein can prevent virus uptake in the cell and thus hâve a neutralizing effect.
RSV F fuses the viral and host-cell membranes by irréversible protein refolding from the labile pre-fusion conformation to the stable post-fusion conformation. Structures of both conformations hâve been determined for RSV F (McLellan JS, et al. Science 342, 592-598 (2013); McLellan JS, et al. Nat Struct Mol Biol 17, 248-250 (2010); McLellan JS, et al. Science 340, 1113-1117 (2013); Swanson KA, et al. Proceedings of the National Academy of Sciences of the United States of America 108, 9619-9624 (2011)), as well as for the fusion proteins from related paramyxoviruses, providing insight into the mechanism of this complex fusion machine. Like other type I fusion proteins, the inactive precursor, RSV Fo, requires cleavage during intracellular maturation by a furin-like protease. RSV F contains two furin sites, which leads to three proteins: F2, p27 and Fl, with the latter containing a hydrophobie fusion peptide (FP) at its N-terminus. In order to refold from the pre-fusion to the post-fusion conformation, the refolding région 1 (RR1) between residue 137 and 216, that includes the FP and heptad repeat A (HRA) has to transform from an assembly of helices, loops and strands to a long continuous hélix. The FP, located at the N-terminal segment of RR1, is then able to extend away from the viral membrane and insert into the proximal membrane of the target cell. Next, the refolding région 2 (RR2), which forms the C-terminal stem in the prefusion F spike and includes the heptad repeat B (HRB), relocates to the other side of the RSV F head and binds the HRA coiled-coil trimer with the HRB domain to form the six-helix bundle. The formation of the RR1 coiled-coil and relocation of RR2 to complété the six-helix bundle are the most dramatic structural changes that occur during the refolding process.
A vaccine against RSV infection is currently not available, but is desired due to the high disease burden. The RSV fusion glycoprotein (RSV F) is an attractive vaccine antigen it is the principal target of neutralizing antibodies in human sera. Most neutralizing antibodies in human sera are directed against the pre-fusion conformation, but due to its instability the pre-fusion conformation has a propensity to prematurely refold into the post-fusion conformation, both in solution and on the surface of the virions. As indicated above, crystal structures hâve revealed a large conformational change between the pre-fusion and postfusion States. The magnitude of the rearrangement suggested that only a portion of antibodies directed to the post-fusion conformation of RSV-F will be able to cross react with the native conformation of the pre-fusion spike on the surface of the virus. Accordingly, efforts to produce a vaccine against RSV hâve focused on developing vaccines that contain pre-fusion forms of RSV F protein (see, e.g., WO20101149745, WO2010/1149743, WO2009/1079796, WO2012/158613). However, these efforts hâve not yet yielded stable pre-fusion RSV F proteins that could be used as candidates for testing in humans.
Therefore, a need remains for efficient vaccines against RSV, in particular vaccines comprising RSV F proteins in the pre-fusion conformation. The présent invention aims at providing such stable pre-fusion RSV F proteins for use in vaccinating against RSV in a safe and effïcacious manner.
Summary of the invention
The présent invention provides stable, recombinant, pre-fusion respiratory syncytial virus (RSV) fusion (F) proteins, i.e. recombinant RSV F proteins that are stabilized in the pre-fusion conformation, and fragments thereof. The RSV F proteins, or fragments thereof, comprise at least one epitope that is spécifie to the pre-fusion conformation F protein. In certain embodiments, the pre-fusion RSV F proteins are soluble proteins. In certain embodiments, the RSV F proteins are trimers. In certain embodiments the RSV F proteins are multimers of trimeric RSV F proteins. The invention also provides nucleic acid molécules encoding the pre-fusion RSV F proteins, or fragments thereof, as well as vectors comprising such nucleic acid molécules.
The invention also relates to compositions, preferably immunogenic compositions, comprising a RSV F protein, a nucleic acid molécule and/or a vector, and to the use thereof in inducing an immune response against RSV F protein, in particular to the use thereof as a vaccine. The invention also relates to methods for inducing an anti-respiratory syncytial virus (RSV) immune response in a subject, comprising administering to the subject an effective amount of a pre-fusion RSV F protein, a nucleic acid molécule encoding said RSV F protein, and/or a vector comprising said nucleic acid molécule. Preferably, the induced immune response is characterized by neutralizing antibodies to RSV and/or protective immunity against RSV. In particular aspects, the invention relates to a method for inducing antirespiratory syncytial virus (RSV) F antibodies in a subject, comprising administering to the subject an effective amount of an immunogenic composition comprising a pre-fusion RSV F protein, a nucleic acid molécule encoding said RSV F protein, and/or a vector comprising said nucleic acid molécule.
Brief description of the Figures
FIG.l: Purification of protein F with mutations N67I, S215P, T357K, N371 Y, and D486N. Superdex200 gel filtration chromatogram of the eluate from the ion-exchange column. The arrow indicates the collected peak.
FIG.2: A) SDS-PAGE analysis of the F N67I, S215P, T357K, N371 Y, D486N protein sample containing peak from the SEC chromatogram under reducing (R) and nonreducing (NR). The gels are stained with Coomassie Brilliant Blue.
FIG. 3: The protein concentration of purified RSV F protein F N67I, S215P, T357K, N371 Y, D486N was measured by Q Octet assay with CR9501 and CR9503 monoclonal antibodies. CR9501 only binds to RSV F in the pre-fusion conformation. CR9503 binds RSV F both in the pre-fusion conformation and the post-fusion conformation. Plotted as Mean±SE.
FIG. 4: Température stability of RSV F protein F N67I, S215P, T357K, N371Y, D486N. Melting température (Tm °C ) determined by differential scanning fluorimetry (DSF) assay with SyproOrange fluorescent dye. Introduction of the T357K and N371Y substitutions increased the Tm of PRPM by 3.5 degrees to 68,5 degrees.
Detailed description of the invention
The fusion protein (F) of the respiratory syncytial virus (RSV) is involved in fusion of the viral membrane with a host cell membrane, which is required for infection. RSV F mRNA is translated into a 574 amino acid precursor protein designated FO, which contains a signal peptide sequence at the N-terminus (e.g. amino acid residues 1-26 of SEQ ID NO: 13) which is removed by a signal peptidase in the endoplasmic réticulum. FO is cleaved at two sites (between amino acid residues 109/110 and 136/137) by cellular proteases (in particular furin, or furin-like)) removing a short glycosylated intervening sequence (also referred to a p27 région, comprising the amino acid residues 110 to 136, and generating two domains or subunits designated Fl and F2. The Fl domain (amino acid residues 137-574) contains a hydrophobie fusion peptide at its N-terminus and the C-terminus contains the transmembrane (TM) (amino acid residues 530-550) and cytoplasmic région (amino acid residues 551-574). The F2 domain (amino acid residues 27-109) is covalently linked to Fl by two disulfide bridges. The F1-F2 heterodimers are assembled as homotrimers in the virion.
A vaccine against RSV infection is currently not yet available. One potential approach to producing a vaccine is a subunit vaccine based on purifîed RSV F protein. However, for this approach it is désirable that the purifîed RSV F protein is in a conformation which resembles the conformation of the pre-fusion State of RSV F protein, which is stable over time and can be produced in sufficient quantities. In addition, for a soluble, subunit-based vaccine, the RSV F protein needs to be truncated by délétion of the transmembrane (TM) and the cytoplasmic région to create a soluble secreted F protein (sF). Because the TM région is responsible for membrane anchoring and stability, the anchorless soluble F protein is considerably more labile than the full-length protein and will readily refold into the postfusion end-state. In order to obtain soluble F protein in the stable pre-fusion conformation that shows high expression levels and high stability, the pre-fusion conformation thus needs to be stabilized. Because also the full length (membrane-bound) RSV F protein is metastable, the stabilization of the pre-fusion conformation is also désirable for the full length RSV F protein, e.g. for any life attenuated or vector based vaccine approach.
For the stabilization of soluble RSV F, that is cleaved into the Fl and F2 subunit, in the pre-fusion conformation, a fibritin - based trimerization domain was fused to the Cterminus of the soluble RSV-F C-terminal end (McLellan et al., Nature Struct. Biol. 17: 2248-250 (2010); McLellan et al., Science 340(6136):1113-7 (2013)). This fibritin domain or Foldon’ is derived ffom T4 fibritin and was described earlier as an artifïcial natural trimerization domain (Letarov et al., Biochemistry Moscow 64: 817-823 (1993); S-Guthe et al., J. Mol. Biol. 337: 905-915. (2004)). However, the trimerization domain does not resuit in stable pre-fusion RSV-F protein (Krarup et al., Nature Comm. 6:8143, (2015)). Moreover, these efforts hâve not yet resulted in candidates suitable for testing in humans.
Recently, we described combinations of several mutations that are capable of stabilizing the RSV F protein in the pre-fusion conformation (WO2014/174018 and WO2014/202570). Thus, stable pre-fusion RSV F proteins hâve been described comprising a mutation of the amino acid residue on position 67 and/or a mutation of the amino acid residue on position 215, preferably a mutation of amino acid residue N/T on position 67 into I and/or a mutation of amino acid residue S on position 215 into P. In addition, soluble pre-fusion RSV F proteins hâve been described comprising a truncated Fl domain, and comprising a mutation of the amino acid residue on position 67 and/or a mutation of the amino acid residue on position 215, preferably a mutation of amino acid residue N/T on position 67 into I and/or a mutation of amino acid residue S on position 215 into P, wherein the protein comprises a heterologous trimerization domain linked to said truncated Fl domain. Additional pre-fusion RSV F proteins hâve been described, wherein the proteins comprise at least one further mutation, such as a mutation of the amino acid residue D on position 486 into N.
According to the présent invention it has been found that the introduction of two other mutations, i.e. on positions 357 and 371 (numbering according to SEQ ID NO: 13) also stabilizes the protein in the pre-fusion conformation.
The présent invention thus provides recombinant pre-fusion F proteins comprising a mutation of the amino acid residue on position 215, in particular a mutation of the amino acid S on position 215 into P (S215P), in combination with a mutation of the amino acids on positions 357, in particular a mutation of the amino acid T on position 357 into K (T357K) and a mutation of the amino acid on position 371, in particular a mutation of the amino acid N on position 371 into Y (N371 Y).
The présent invention thus provides a unique combination of mutations to provide recombinant stable pre-fusion RSV F proteins, i.e. RSV F proteins that are stabilized in the pre-fusion conformation, or fragments thereof. The stable pre-fusion RSV F proteins of the invention, or fragments thereof, are in the pre-fusion conformation, i.e. they comprise (display) at least one epitope that is spécifie to the pre-fusion conformation F protein. An epitope that is spécifie to the pre-fusion conformation F protein is an epitope that is not presented in the post-fusion conformation. Without wishing to be bound by any particular theory, it is believed that the pre-fusion conformation of RSV F protein may contain epitopes that are the same as those on the RSV F protein expressed on natural RSV virions, and therefore may provide advantages for eliciting protective neutralizing antibodies.
In certain embodiments, the pre-fusion RSV F proteins of the invention, or fragments thereof, comprise at least one epitope that is recognized by a pre-fusion spécifie monoclonal antibody, comprising a heavy chain CDR1 région of SEQ ID NO: 1, a heavy chain CDR2 région of SEQ ID NO: 2, a heavy chain CDR3 région of SEQ ID NO: 3 and a light chain CDR1 région of SEQ ID NO: 4, a light chain CDR2 région of SEQ ID NO: 5, and a light chain CDR3 région of SEQ ID NO: 6 (hereafter referred to as CR9501) and/or a pre-fusion spécifie monoclonal antibody, comprising a heavy chain CDR1 région of SEQ ID NO: 7, a heavy chain CDR2 région of SEQ ID NO: 8, a heavy chain CDR3 région of SEQ ID NO: 9 and a light chain CDR1 région of SEQ ID NO: 10, a light chain CDR2 région of SEQ ID NO: 11, and a light chain CDR3 région of SEQ ID NO: 12 (referred to as CR9502). CR9501 and CR9502 comprise the heavy and light chain variable régions, and thus the binding specificities, of the antibodies 58C5 and 30D8, respectively, which hâve previously been shown to bind specifically to RSV F protein in its pre-fusion conformation and not to the post-fusion conformation (see WO2012/006596).
In certain embodiments, the recombinant pre-fusion RSV F proteins are trimeric.
As used throughout the présent application nucléotide sequences are provided from 5’ to 3’ direction, and amino acid sequences from N-terminus to C-terminus, as custom in the art.
As indicated above, fragments of the pre-fusion RSV F protein are also encompassed by the présent invention. The fragment may resuit from either or both of amino-terminal (e.g. by cleaving off the signal sequence) and carboxy-terminal délétions (e.g. by deleting the transmembrane région and/or cytoplasmic tail). The fragment may be chosen to comprise an immunologically active fragment of the F protein, i.e. a part that will give rise to an immune response in a subject. This can be easily determined using in silico, in vitro and/or in vivo methods, ail routine to the skilled person.
In certain embodiments, the encoded proteins or fragments thereof according to the invention comprise a signal sequence, also referred to as leader sequence or signal peptide, corresponding to amino acids 1-26 of SEQ ID NO: 13. Signal sequences typically are short (e.g. 5-30 amino acids long) amino acid sequences présent at the N-terminus of the majority of newly synthesized proteins that are destined towards the secretory pathway, and are typically cleaved by signal peptidase to generate a free signal peptide and a mature protein.
In certain embodiments, the proteins or fragments thereof according to the invention do not comprise a signal sequence.
In certain embodiments, the (fragments of the) pre-fusion RSV F proteins are soluble. In certain embodiments, the stable pre-fusion RSV F proteins or fragments thereof according to the invention comprise a truncated Fl domain, and comprise a heterologous trimerization domain linked to said truncated F1 domain. According to the invention, it was shown that by linking a heterologous trimerization domain to the C-terminal amino acid residue of a truncated Fl domain, combined with the stabilizing mutation(s), soluble RSV F proteins are provided that show high expression and that bind to pre-fusion-specific antibodies, indicating that the proteins are in the pre-fusion conformation. In addition, the RSV F proteins are stabilized in the pre-fusion conformation, i.e. even after processing of the proteins they still bind to the pre-fusion spécifie antibodies CR9501 and/or CR9502, indicating that the prefusion spécifie epitope is retained.
In certain embodiments, the RSV F proteins are multimers of trimeric RSV F proteins. Thus, in some embodiments, the RSV F proteins may comprise an assembly domain for higher order assemblies of trimers.
It is known that RSV exists as a single serotype having two antigenic subgroups: A and B. The amino acid sequences of the mature processed F proteins of the two groups are about 93% identical. As used throughout the présent application, the amino acid positions are given in référencé to the sequence of RSV F protein of subgroup A (SEQ ID NO: 13). As used in the présent invention, the wording “the amino acid at position “x” of the RSV F protein thus means the amino acid corresponding to the amino acid at position “x” in the RSV F protein of SEQ ID NO: 13. Note that, in the numbering System used throughout tbis application 1 refers to the N-terminal amino acid of an immature FO protein (SEQ ID NO: 13) When another RSV strain is used, the amino acid positions of the F protein are to be numbered with reference to the numbering of the F protein of SEQ ID NO: 13 by aligning the sequences of the other RSV strain with the F protein of SEQ ID NO: 13 with the insertion of gaps as needed. Sequence alignments can be done using methods well known in the art, e.g. by CLUSTALW, Bioedit or CLC Workbench.
In certain embodiments, the RSV strain is the RSV strain of SEQ ID NO: 20.
In certain embodiments, the RSV strain is an RSV B strain. In certain embodiments, the RSV strain is the RSV B strain of SEQ ID NO: 15.
An amino acid according to the invention can be any of the twenty naturally occurring (or ‘standard’ amino acids) or variants thereof, such as e.g. D-amino acids (the D-enantiomers of amino acids with a chiral center), or any variants that are not naturally found in proteins, such as e.g. norleucine. The standard amino acids can be divided into several groups based on their properties. Important factors are charge, hydrophilicity or hydrophobicity, size and functional groups. These properties are important for protein structure and protein-protein interactions. Some amino acids hâve spécial properties such as cysteine, that can form covalent disulfide bonds (or disulfide bridges) to other cysteine residues, proline that induces tums of the protein backbone, and glycine that is more flexible than other amino acids. Table 1 shows the abbreviations and properties of the standard amino acids.
It will be appreciated by a skilled person that the mutations can be made to the protein by routine molecular biology procedures. The mutations according to the invention preferably resuit in increased expression levels and/or increased stabilization of the pre-fusion RSV F proteins as compared RSV F proteins that do not comprise these mutation(s).
In certain embodiments, the pre-fusion RSV F proteins or fragments thereof comprise at least one further mutation selected from the group consisting of:
(a) a mutation of the amino acid residue on position 67; and (b) a mutation of the amino acid residue on position 486.
In certain embodiments, the at least one further mutation is selected from the group consisting of:
(a) a mutation of the amino acid residue N/T on position 67 into I; and (b) a mutation of the amino acid residue D on position 486 into N.
In certain embodiments, the pre-fusion RSV F proteins or fragments thereof comprise at least four mutations (as compared to a wild-type RV F protein, e.g. the comprising the amino acid sequence of SEQ ID NO : 13). In certain embodiments, the proteins or fragments thereof comprise at least five mutations.
In certain embodiments, the proteins or fragments thereof comprise at least six mutations.
In certain embodiments, the pre-fusion RSV F polypeptides thus comprise at least one further mutation selected from the group consisting of:
(a) a mutation of the amino acid residue on position 46;
(b) a mutation of the amino acid residue on position 83;
(c) a mutation of the amino acid residue on position 92;
(d) a mutation of the amino acid residue on position 184;
(e) a mutation of the amino acid residue on position 203;
(f) a mutation of the amino acid residue on position 207; and (g) a mutation of the amino acid residue on position 487.
In certain embodiments, the at least one further mutation is selected from the group consisting of:
(a) a mutation of the amino acid residue S on position 46 into G;
(b) a mutation of the amino acid residue L on position 83 into M:
(c) a mutation of the amino acid residue E on position 92 into D;
(d) a mutation of the amino acid residue G on position 184 into N;
(e) a mutation of the amino acid residue L on position 203 into I;
(f) a mutation of the amino acid residue V on position 207 into I: and (g) a mutation of the amino acid residue E on position 487 into Q, N or I.
In certain other embodiments, the heterologous trimerization domain comprises the amino acid sequence GYIPEAPRDGQAYVRKDGEWVLLSTFL (SEQ ID NO: 14).
As described above, in certain embodiments, the proteins of the invention or fragments thereof comprise a truncated Fl domain. As used herein a “truncated” Fl domain refers to a Fl domain that is not a full length Fl domain, i.e. wherein either N-terminally or C-terminally one or more amino acid residues hâve been deleted. According to the invention, at least the transmembrane domain and cytoplasmic tail hâve been deleted to permit expression as a soluble ectodomain.
In certain other embodiments, the trimerization domain is linked to amino acid residue 513 of the RSV Fl domain. In certain embodiments, the trimerization domain thus comprises SEQ ID NO: 14 and is linked to amino acid residue 513 of the RSV Fl domain.
In certain embodiments, the RSV F protein of the invention comprises the amino acid sequence of SEQ ID NO: 21.
In certain embodiments, the level of expression of the pre-fusion RSV F proteins of the invention is increased, as compared to a wild-type RSV F protein. In certain embodiments the level of expression is increased at least 5-fold, preferably up to 10-fold. In certain embodiments, the level of expression is increased more than 10-fold.
The pre-fusion RSV F proteins according to the invention are stable, i.e. do not readily change into the post-fusion conformation upon processing of the proteins, such as e.g. purification, freeze-thaw cycles, and/or storage etc.
In certain embodiments, the pre-fusion RSV F proteins according to the invention hâve an increased stability upon storage a 4°C as compared to a RSV F protein without the mutation(s). In certain embodiments, the proteins are stable upon storage at 4°C for at least 30 days, preferably at least 60 days, preferably at least 6 months, even more preferably at least 1 year. With “stable upon storage”, it is meant that the proteins still display the at least one epitope spécifie for the a pre-fusion spécifie antibody (e.g. CR9501) upon storage of the protein in solution (e.g. culture medium) at 4° C for at least 30 days. In certain embodiments, the proteins display the at least one pre-fusion spécifie epitope for at least 6 months, preferably for at least 1 year upon storage of the pre-fusion RSV F proteins at 4°C.
In certain embodiments, the pre-fusion RSV F proteins according to the invention hâve an increased stability when subjected to heat, as compared to RSV F proteins without said mutation(s). In certain embodiments, the pre-fusion REV F proteins are heat stable for at least 30 minutes at a température of 55° C, preferably at 58° C, more preferably at 60° C With heat stable it is meant that the proteins still display the at least one pe-fusion spécifie epitope after having been subjected for at least 30 minutes to an increased température (i.e. a température of 55°C or above), e.g. as determined using a method as described in the Examples (see Fig. 4).
In certain embodiments, the proteins display the at least one pre-fusion spécifie epitope after being subjected to 1 to 6 freeze-thaw cycles in an appropriate formulation buffer.
As used throughout the présent application nucléotide sequences are provided from 5’ to 3 ’ direction, and amino acid sequences from N-terminus to C-terminus, as custom in the art.
In certain embodiments, the encoded proteins according to the invention further comprise a leader sequence, also referred to as signal sequence or signal peptide, corresponding to amino acids 1-26 of SEQ ID NO: 13. This is a short (typically 5-30 amino acids long) peptide présent at the N-terminus of the majority of newly synthesized proteins that are destined towards the secretory pathway. In certain embodiments, the proteins according to the invention do not comprise a leader sequence.
In certain embodiments, the proteins comprise a HIS-Tag. A His-Tag or polyhistidine-tag is an amino acid motif in proteins that consists of at least five histidine (H) residues, often at the N- or C-terminus of the protein, which is generally used for purification purposes.
The présent invention further provides nucleic acid molécules encoding the RSV F proteins according to the invention.
In preferred embodiments, the nucleic acid molécules encoding the proteins according to the invention are codon-optimized for expression in mammalian cells, preferably human cells. Methods of codon-optimization are known and hâve been described previously (e.g. WO 96/09378). A sequence is considered codon-optimized if at least one non-preferred codon as compared to a wild type sequence is replaced by a codon that is more preferred. Herein, a non-preferred codon is a codon that is used less frequently in an organism than another codon coding for the same amino acid, and a codon that is more preferred is a codon that is used more frequently in an organism than a non-preferred codon. The frequency of codon usage for a spécifie organism can be found in codon frequency tables, such as in http://www.kazusa.or.jp/codon. Preferably more than one non-preferred codon, preferably most or ail non-preferred codons, are replaced by codons that are more preferred. Preferably the most frequently used codons in an organism are used in a codon-optimized sequence. Replacement by preferred codons generally leads to higher expression.
It will be understood by a skilled person that numerous different polynucleotides and nucleic acid molécules can encode the same protein as a resuit of the degeneracy of the genetic code. It is also understood that skilled persons may, using routine techniques, make nucléotide substitutions that do not affect the protein sequence encoded by the nucleic acid molécules to reflect the codon usage of any particular host organism in which the proteins are to be expressed. Therefore, unless otherwise specifîed, a nucléotide sequence encoding an amino acid sequence includes ail nucléotide sequences that are degenerate versions of each other and that encode the same amino acid sequence. Nucléotide sequences that encode proteins and RNA may or may not include introns.
Nucleic acid sequences can be cloned using routine molecular biology techniques, or generated de novo by DNA synthesis, which can be performed using routine procedures by service companies having business in the fïeld of DNA synthesis and/or molecular cloning (e.g. GeneArt, GenScripts, Invitrogen, Eurofins).
The invention also provides vectors comprising a nucleic acid molécule as described above. In certain embodiments, a nucleic acid molécule according to the invention thus is part of a vector. Such vectors can easily be manipulated by methods well known to the person skilled in the art, and can for instance be designed for being capable of réplication in prokaryotic and/or eukaryotic cells. In addition, many vectors can be used for transformation of eukaryotic cells and will integrate in whole or in part into the genome of such cells, resulting in stable host cells comprising the desired nucleic acid in their genome. The vector used can be any vector that is suitable for cloning DNA and that can be used for transcription of a nucleic acid of interest. Suitable vectors according to the invention are e.g. adenovectors, alphavirus, paramyxovirus, vaccinia virus, herpes virus, retroviral vectors etc. The person skilled in the art is capable of choosing suitable expression vectors, and inserting the nucleic acid sequences of the invention in a functional manner.
Host cells comprising the nucleic acid molécules encoding the pre-fusion RSV F proteins form also part of the invention. The pre-fusion RSV F proteins may be produced through recombinant DNA technology involving expression of the molécules in host cells, e.g. Chinese hamster ovary (CHO) cells, tumor cell fines, BHK cells, human cell fines such as
HEK293 cells, PER.C6 cells, or yeast, fungi, insect cells, and the like, or transgenic animais or plants. In certain embodiments, the cells are from a multicellular organism, in certain embodiments they are of vertebrate or invertebrate origin. In certain embodiments, the cells are mammalian cells. In certain embodiments, the cells are human cells. In general, the production of a recombinant proteins, such the pre-fusion RSV F proteins of the invention, in a host cell comprises the introduction of a heterologous nucleic acid molécule encoding the protein in expressible format into the host cell, culturing the cells under conditions conducive to expression of the nucleic acid molécule and allowing expression of the protein in said cell. The nucleic acid molécule encoding a protein in expressible format may be in the form of an expression cassette, and usually requires sequences capable of bringing about expression of the nucleic acid, such as enhancer(s), promoter, polyadenylation signal, and the like. The person skilled in the art is aware that varions promoters can be used to obtain expression of a gene in host cells. Promoters can be constitutive or regulated, and can be obtained from varions sources, including viruses, prokaryotic, or eukaryotic sources, or artificially designed.
Cell culture media are available from varions vendors, and a suitable medium can be routinely chosen for a host cell to express the protein of interest, here the pre-fusion RSV F proteins. The suitable medium may or may not contain sérum.
A heterologous nucleic acid molécule” (also referred to herein as ‘transgene’) is a nucleic acid molécule that is not naturally présent in the host cell. It is introduced into for instance a vector by standard molecular biology techniques. A transgene is generally operably linked to expression control sequences. This can for instance be done by placing the nucleic acid encoding the transgene(s) under the control of a promoter. Further regulatory sequences may be added. Many promoters can be used for expression of a transgene(s), and are known to the skilled person, e.g. these may comprise viral, mammalian, synthetic promoters, and the like. A non-limiting example of a suitable promoter for obtaining expression in eukaryotic cells is a CMV-promoter (US 5,385,839), e.g. the CMV immédiate early promoter, for instance comprising nt. -735 to +95 from the CMV immédiate early gene enhancer/promoter. A polyadenylation signal, for example the bovine growth hormone polyA signal (US 5,122,458), may be présent behind the transgene(s). Altematively, several widely used expression vectors are available in the art and from commercial sources, e.g. the pcDNA and pEF vector sériés of Invitrogen, pMSCV and pTK-Hyg from BD Sciences, pCMV-Script from Stratagene, etc, which can be used to recombinantly express the protein of interest, or to obtain suitable promoters and/or transcription terminator sequences, polyA sequences, and the like.
The cell culture can be any type of cell culture, including adhèrent cell culture, e.g. cells attached to the surface of a culture vessel or to microcarriers, as well as suspension culture. Most large-scale suspension cultures are operated as batch or fed-batch processes because they are the most straightforward to operate and scale up. Nowadays, continuous processes based on perfusion principles are becoming more common and are also suitable. Suitable culture media are also well known to the skilled person and can generally be obtained from commercial sources in large quantifies, or custom-made according to standard protocols. Culturing can be done for instance in dishes, roller bottles or in bioreactors, using batch, fed-batch, continuous Systems and the like. Suitable conditions for culturing cells are known (see e.g. Tissue Culture, Academie Press, Kruse and Paterson, editors (1973), and R.I. Freshney, Culture of animal cells: A manual of basic technique, fourth édition (Wiley-Liss Inc., 2000, ISBN 0-471-34889-9)).
The invention further provides compositions comprising a pre-fusion RSV F protein and/or a nucleic acid molécule, and/or a vector, as described above. The invention thus provides compositions comprising a pre-fusion RSV F protein that displays an epitope that is présent in a pre-fusion conformation of the RSV F protein but is absent in the post-fusion conformation. The invention also provides compositions comprising a nucleic acid molécule and/or a vector, encoding such pre-fusion RSV F protein. The invention further provides immunogenic compositions comprising a pre-fusion RSV F protein, and/or a nucleic acid molécule, and/or a vector, as described above. The invention also provides the use of a stabilized pre-fusion RSV F protein, a nucleic acid molécule, and/or a vector, according to the invention, for inducing an immune response against RSV F protein in a subject. Further provided are methods for inducing an immune response against RSV F protein in a subject, comprising administering to the subject a pre-fusion RSV F protein, and/or a nucleic acid molécule, and/or a vector, according to the invention. Also provided are pre-fusion RSV F proteins, nucleic acid molécules, and/or vectors, according to the invention for use in inducing an immune response against RSV F protein in a subject. Further provided is the use of the pre-fusion RSV F proteins, and/or nucleic acid molécules, and/or vectors according to the invention for the manufacture of a médicament for use in inducing an immune response against RSV F protein in a subject.
The pre-fusion RSV F proteins, nucleic acid molécules, or vectors of the invention may be used for prévention (prophylaxis) and/or treatment of RSV infections. In certain embodiments, the prévention and/or treatment may be targeted at patient groups that are susceptible RSV infection. Such patient groups include, but are not limited to e.g., the elderly (e.g. > 50 years old, > 60 years old, and preferably > 65 years old), the young (e.g. < 5 years old, < l year old), hospitalized patients and patients who hâve been treated with an antiviral compound but hâve shown an inadéquate antiviral response.
The pre-fusion RSV F proteins, nucleic acid molécules and/or vectors according to the invention may be used e.g. in stand-alone treatment and/or prophylaxis of a disease or condition caused by RSV, or in combination with other prophylactic and/or therapeutic treatments, such as (existing or future) vaccines, antiviral agents and/or monoclonal antibodies.
The invention further provides methods for preventing and/or treating RSV infection in a subject utilizing the pre-fusion RSV F proteins, nucleic acid molécules and/or vectors according to the invention. In a spécifie embodiment, a method for preventing and/or treating RSV infection in a subject comprises administering to a subject in need thereof an effective amount of a pre-fusion RSV F protein, nucleic acid molécule and/or a vector, as described above. A therapeutically effective amount refers to an amount of a protein, nucleic acid molécule or vector, that is effective for preventing, ameliorating and/or treating a disease or condition resulting from infection by RSV. Prévention encompasses inhibiting or reducing the spread of RSV or inhibiting or reducing the onset, development or progression of one or more of the symptoms associated with infection by RSV. Amelioration as used in herein may refer to the réduction of visible or perceptible disease symptoms, viremia, or any other measurable manifestation of influenza infection.
For administering to subjects, such as humans, the invention may employ pharmaceutical compositions comprising a pre-fusion RSV F protein, a nucleic acid molécule and/or a vector as described herein, and a pharmaceutically acceptable carrier or excipient. In the présent context, the term pharmaceutically acceptable means that the carrier or excipient, at the dosages and concentrations employed, will not cause any unwanted or harmful effects in the subjects to which they are administered. Such pharmaceutically acceptable carriers and excipients are well known in the art (see Remington's Pharmaceutical Sciences, 18th édition, A. R. Gennaro, Ed., Mack Publishing Company [1990]; Pharmaceutical Formulation Development of Peptides and Proteins, S. Frokjaer and L. Hovgaard, Eds., Taylor & Francis [2000]; and Handbook of Pharmaceutical Excipients, 3rd édition, A. Kibbe, Ed., Pharmaceutical Press [2000]). The RSV F proteins, or nucleic acid molécules, preferably are formulated and administered as a stérile solution although it may also be possible to utilize lyophilized préparations. Stérile solutions are prepared by stérile filtration or by other methods known per se in the art. The solutions are then lyophilized or filled into pharmaceutical dosage containers. The pH of the solution generally is in the range of pH 3.0 to 9.5, e.g. pH 5.0 to 7.5. The RSV F proteins typically are in a solution having a suitable pharmaceutically acceptable buffer, and the composition may also contain a sait. Optionally stabilizing agent may be présent, such as albumin. In certain embodiments, detergent is added. In certain embodiments, the RSV F proteins may be formulated into an injectable préparation.
In certain embodiments, a composition according to the invention further comprises one or more adjuvants. Adjuvants are known in the art to further increase the immune response to an applied antigenic déterminant. The terms “adjuvant” and immune stimulant are used interchangeably herein, and are defined as one or more substances that cause stimulation of the immune System. In this context, an adjuvant is used to enhance an immune response to the RSV F proteins of the invention. Examples of suitable adjuvants include aluminium salts such as aluminium hydroxide and/or aluminium phosphate; oil-emulsion compositions (or oil-in-water compositions), including squalene-water émulsions, such as MF59 (see e.g. WO 90/14837); saponin formulations, such as for example QS21 and Immunostimulating Complexes (ISCOMS) (see e.g. US 5,057,540; WO 90/03184, WO 96/11711, WO 2004/004762, WO 2005/002620); bacterial or microbial dérivatives, examples of which are monophosphoryl lipid A (MPL), 3-O-deacylated MPL (3dMPL), CpG-motif containing oligonucleotides, ADP-ribosylating bacterial toxins or mutants thereof, such as E. coli heat labile enterotoxin LT, choiera toxin CT, and the like; eukaryotic proteins (e.g. antibodies or fragments thereof (e.g. directed against the antigen itself or CD la, CD3, CD7, CD80) and ligands to receptors (e.g. CD40L, GMCSF, GCSF, etc), which stimulate immune response upon interaction with récipient cells. In certain embodiments the compositions of the invention comprise aluminium as an adjuvant, e.g. in the form of aluminium hydroxide, aluminium phosphate, aluminium potassium phosphate, or combinations thereof, in concentrations of 0.05 - 5 mg, e.g. from 0.075-1.0 mg, of aluminium content per dose.
The pre-fusion RSV F proteins may also be administered in combination with or conjugated to nanoparticles, such as e.g. polymers, liposomes, virosomes, virus-like particles. The pre-fusion F proteins may be combined with, encapsidated in or conjugated to the nanoparticles with or without adjuvant. Encapsulation within liposomes is described, e.g. in US 4,235,877. Conjugation to macromolecules is disclosed, for example in US 4,372,945 or US 4,474,757. In other embodiments, the RSV F proteins are assembled in higher order assemblies of multimers.
In other embodiments, the compositions do not comprise adjuvants.
In certain embodiments, the invention provides methods for making a vaccine against respiratory syncytial virus (RSV), comprising providing a composition according to the invention and formulating it into a pharmaceutically acceptable composition. The terni vaccine refers to an agent or composition containing an active component effective to induce a certain degree of immunity in a subject against a certain pathogen or disease, which will resuit in at least a decrease (up to complété absence) of the severity, duration or other manifestation of symptoms associated with infection by the pathogen or the disease. In the présent invention, the vaccine comprises an effective amount of a pre-fusion RSV F protein and/or a nucleic acid molécule encoding a pre-fusion RSV F protein, and/or a vector comprising said nucleic acid molécule, which results in an immune response against the F protein of RSV. This provides a method of preventing serions lower respiratory tract disease leading to hospitalization and the decrease in frequency of complications such as pneumonia and bronchiolitis due to RSV infection and réplication in a subject. The term “vaccine” according to the invention implies that it is a pharmaceutical composition, and thus typically includes a pharmaceutically acceptable diluent, carrier or excipient. It may or may not comprise further active ingrédients. In certain embodiments it may be a combination vaccine that further comprises other components that induce an immune response, e.g. against other proteins of RSV and/or against other infectious agents. The administration of further active components may for instance be done by separate administration or by administering combination products of the vaccines of the invention and the further active components.
Compositions may be administered to a subject, e.g. a human subject. The total dose of the RSV F proteins in a composition for a single administration can for instance be about 0.01 pg to about 10 mg, e.g. 1 pg - 1 mg, e.g. 10 pg - 100 pg. Determining the recommended dose will be carried out by expérimentation and is routine for those skilled in the art.
Administration of the compositions according to the invention can be performed using standard routes of administration. Non-limiting embodiments include parentéral administration, such as intradermal, intramuscular, subcutaneous, transcutaneous, or mucosal administration, e.g. intranasal, oral, and the like. In one embodiment a composition is administered by intramuscular injection. The skilled person knows the various possibilities to administer a composition, e.g. a vaccine in order to induce an immune response to the antigen(s) in the vaccine.
A subject as used herein preferably is a mammal, for instance a rodent, e.g. a mouse, a cotton rat, or a non-human-primate, or a human. Preferably, the subject is a human subject.
The proteins, nucleic acid molécules, vectors, and/or compositions may also be administered, either as prime, or as boost, in a homologous or heterologous prime-boost regimen. If a boosting vaccination is performed, typically, such a boosting vaccination will be administered to the same subject at a time between one week and one year, preferably between two weeks and four months, after administering the composition to the subject for the first time (which is in such cases referred to as ‘priming vaccination’). In certain embodiments, the administration comprises a prime and at least one booster administration.
In addition, the proteins of the invention may be used as diagnostic tool, for example to test the immune status of an individual by establishing whether there are antibodies in the sérum of such individual capable of binding to the protein of the invention. The invention thus also relates to an in vitro diagnostic method for detecting the presence of an RSV infection in a patient said method comprising the steps of a) contacting a biological sample obtained from said patient with a protein according to the invention; and b) detecting the presence of antibodyprotein complexes.
Stabilized pre-fusion RSV F proteins obtainable and/or obtained by such method also form part of the invention, as well as uses thereof as described above.
Examples
EXAMPLE 1 : Préparation of stable pre-fusion RSV F polypeptide of SEQID NO: 21
To increase the stability of RSV F in the pre-fusion conformation two additional amino acid substitutions were introduced in a pre-fusion RSV F variant that was described previously (WO2014/174018 and WO2014/202570). The constructs were synthesized and codon-optimized at Gene Art (Life Technologies, Carlsbad, CA). The constructs were cloned into pCDNA2004 or generated by standard methods widely known within the field involving site-directed mutagenesis and PCR and sequenced. The expression platform used was the 293Freestyle cells (Life Technologies). The cells were transiently transfected using 293Fectin (Life Technologies) according to the manufacturer’s instructions and cultured for 5 days at 37°C and 10% CO2. The culture supematant was harvested and spun for 5 minutes at 300 g to remove cells and cellular débris. The spun supematant was subsequently stérile filtered using a 0.22 um vacuum fïlter and stored at 4°C until use.
EXAMPLE 2: Purification ofpre-fusion RSV F protein
The recombinant polypeptide was purified by a 2-step purification protocol applying a cat-ion exchange column for the initial purification and subsequently a superdex200 column for the polishing step to remove residual contaminants. For the initial ion-exchange step the culture supematant was diluted with 2 volumes of 50 mM NaOAc pH 5.0 and passed over a 5 ml HiTrap Capto S column at 5 ml per minute. Subsequently the column was washed with 10 column volumes (CV) of 20 mM NaOAc, 50mM NaCl, 0.01% (v/v) tween20, pH 5 and eluted 2 CV of 20 mM NaOAc, IM NaCl, 0.01% (v/v) tween20, pH 5. The eluate was concentrated using a spin concentrator and the protein was further purified using a superdex200 column using 40mM Tris, 500mM NaCl, 0.01% (v/v) tween20, pH 7.4 as running buffer. In Figure 1 the chromatogram of the gel filtration column is shown. The dominant peak contained the pre-fusion RSV F protein. The fractions containing this peak were again pooled and the protein concentration was determined using OD280 and stored a 4°C until use. In Figure 2 a non-reduced and reduced SDS-PAGE analysis of the final protein préparation is shown and as can be seen the purity was >95%. The identity of the band was verified using western blotting and protein F spécifie antibodies (not shown).
Quantitative Octet (BioLayer Interferometry) was used for measuring protein concentration in the supematants. CR9501 (an antibody specifically recognizing pre-fusion RSV F protein) and CR9503 (recognizing post-fusion RSV F protein) were biotinylated by standard protocols and immobilized on Streptavidin biosensors (ForteBio, Portsmouth, UK). Afterwards, the coated biosensors were blocked in mock cell culture supematant. A quantitative experiment was performed as follows: température 3 0C, shaking speed 1000 rpm, time of the assay 300 sec.
The concentration of the protein was calculated using a standard curve. The standard curve was prepared for each coated antibody using the pre-fusion RSV F protein (Krarup et. al., 2015, supra) diluted in mock medium (Figure 3). The data analysis was done using the ForteBio Data Analysis 6.4 software (ForteBio).
EXAMPLE 3: Température stability of the RSV F protein
Température stability of the purified protein was determined by differential scanning fluorometry (DSF). The purified pre-fusion F protein was mixed with SYPRO orange fluorescent dye (Life Technologies S6650) in a 96-well optical qPCR plate. The optimal dye and protein concentration was determined experimentally (data not shown). Protein dilutions were performed in PBS, and a négative control sample containing the dye only was used as a reference subtraction. The measurement was performed in a qPCR instrument (Applied Biosystems ViiA 7) using the foliowing parameters: a température ramp from 25-95 °C with a rate of 0.015 °C per second. Data was collected continuously. The melting curves were plotted using GraphPad PRISM software (version 5.04). Melting températures were calculated at the 50% maximum of fluorescence using a non-linear EC50 shift équation. The melting température of the RSV F protein of SEQ ID NO: 21 was 68.5 degrees (Fig. 4). A référencé pre-fusion RSV F without substitutions at position 357 and 371 had a melting température of 65.0 which means the double mutation increased the melting température by 3.5 degrees.
Table 1. Standard amino acids, abbreviations and properties
Amino Acid 3-Lettei 1-Letter Side chain polarity Side chain charge (pH 7.4)
alanine Ala A non-polar Neutral
arginine Arg R polar Positive
asparagine Asn N polar Neutral
aspartic acid Asp D polar Négative
cysteine Cys C non-polar Neutral
glutamic acid Glu E polar Négative
glutamine Gin Q polar Neutral
glycine Gly G non-polar Neutral
histidine His H polar positive(10%) neutral(90%)
isoleucine Ile I non-polar Neutral
leucine Leu L non-polar Neutral
lysine Lys K polar Positive
méthionine Met M non-polar Neutral
phenylalanine Phe F non-polar Neutral
proline Pro P non-polar Neutral
serine Ser S polar Neutral
threonine Thr T polar Neutral
tryptophan Trp w non-polar Neutral
tyrosine Tyr Y polar Neutral
valine Val V non-polar Neutral
Table 2. Amino acid sequences of antibodies CR9501 and CR9502
Ab VH domain VH CDR1 VH CDR2 VH CDR3
CR9501 Amino acids 1- 125 ofSEQ ID NO: 16 GASINSDNYYWT (SEQ ID NO:1) HISYTGNTYYTPSLKS (SEQ ID NO:2) CGAYVLISNCGWFDS (SEQ ID NO:3)
CR9502 Amino acids 1- 121 ofSEQ ID NO:18 GFTFSGHTIA (SEQ ID NO:7) WVSTNNGNTEYAQKI QG (SEQ ID NO:8) EWLVMGGFAFDH (SEQ ID NO:9)
Ab VL domain VL CDR1 VL CDR2 VL CDR3
CR9501 Amino acids 1-107 ofSEQ ID NO: 17 QASQDISTYLN (SEQ ID NO: 4) GASNLET (SEQ ID NO:5) QQYQYLPYT (SEQ ID NO:6)
CR9502 Amino acids 1-110 ofSEQ ID NO: 19 GANNIGSQNVH (SEQ ID NO:10) DDRDRPS (SEQ ID NO:11) QVWDSSRDQAVI (SEQ ID NO:12)
Sequences
RSV F protein full length sequence subgroup A (SEQ ID NO: 13)
MELLILKANAITTILTAVTFCFASGQNITEEFYQSTCSAVSKGYLSALRTGWYTSVITIE LSNIKENKCNGTDAKVKLIKQELDKYKNAVTELQLLMQSTPATNNRARRELPRFMN YTLNNAKKTNVTLSKKRKRRFLGFLLGVGSAIASGVAVSKVLHLEGEVNKIKSALLS TNKAWSLSNGVSVLTSKVLDLKNYIDKQLLPIVNKQSCSISNIETVIEFQQKNNRLLE ITREFSVNAGVTTPVSTYMLTNSELLSLINDMPITNDQKKLMSNNVQIVRQQSYSIMSI IKEEVLAYVVQLPLYGVIDTPCWKLHTSPLCTTNTKEGSNICLTRTDRGWYCDNAGS VSFFPQAETCKVQSNRVFCDTMNSLTLPSEVNLCNVDIFNPKYDCKIMTSKTDVSSSV ITSLGAIVSCYGKTKCTASNKNRGIIKTFSNGCDYVSNKGVDTVSVGNTLYYVNKQE GKSLYVKGEPIINFYDPLVFPSDEFDASISQVNEKINQSLAFIRKSDELLHNVNAVKST TNIMITTIIIVIIVILLSLIAVGLLLYCKARSTPVTLSKDQLSGINNIAFSN
RSV F protein B1 full length sequence (SEQ ID NO: 15)
MELLIHRLSAIFLTLAINALYLTSSQNITEEFYQSTCSAVSRGYFSALRTGWYTSVITIE LSNIKETKCNGTDTKVKLIKQELDKYKNAVTELQLLMQNTPAANNRARREAPQYMN YTINTTKNLNVSISKKRKRRFLGFLLGVGSAIASGIAVSKVLHLEGEVNKIKNALLSTN KAVVSLSNGVSVLTSKVLDLKNYINNQLLPIVNQQSCRISNIETVIEFQQKNSRLLEIN REFSVNAGVTTPLSTYMLTNSELLSLINDMPITNDQKKLMSSNVQIVRQQSYSIMSIIK EEVLAYVVQLPIYGVIDTPCWKLHTSPLCTTNIKEGSNICLTRTDRGWYCDNAGSVSF FPQADTCKVQSNRVFCDTMNSLTLPSEVSLCNTDIFNSKYDCKIMTSKTDISSSVITSL GAIVSCYGKTKCTASNKNRGIIKTFSNGCDYVSNKGVDTVSVGNTLYYVNKLEGKN LYVKGEPIINYYDPLVFPSDEFDASISQVNEKINQSLAFIRRSDELLHNVNTGKSTTNI MITTIIIVIIVVLLSLIAIGLLLYCKAKNTPVTLSKDQLSGINNIAFSK
SEQ ID NO: 14 (fibritin)
GYIPEAPRDGQAYVRKDGEWVLLSTFL
RSV F protein CL57-v224 full length sequence (SEQ ID NO: 20)
MELPILKTNAITTILAAVTLCFASSQNITEEFYQSTCSAVSKGYLSALRTGWYTSVITIE LSNIKENKCNGTDAKVKLIKQELDKYKNAVTELQLLMQSTPAANNRARRELPRFMN YTLNNTKNNNVTLSKKRKRRFLGFLLGVGSAIASGIAVSKVLHLEGEVNKIKSALLST NKAWSLSNGVSVLTSKVLDLKNYIDKQLLPIVNKQSCSISNIETVIEFQQKNNRLLEI TREFSVNAGVTTPVSTYMLTNSELLSLINDMPITNDQKKLMSNNVQIVRQQSYSIMSII KEEVLAYVVQLPLYGVIDTPCWKLHTSPLCTTNTKEGSNICLTRTDRGWYCDNAGS VSFFPQAETCKVQSNRVFCDTMNSLTLPSEVNLCNIDIFNPKYDCKIMTSKTDVSSSVI TSLGAIVSCYGKTKCTASNKNRGIIKTFSNGCDYVSNKGVDTVSVGNTLYYVNKQEG KSLYVKGEPIINFYDPLVFPSDEFDASISQVNEKINQSLAFIRKSDELLHNVNVGKSTT NIMITTIIIVIIVILLLLIAVGLFLYCKARSTPVTLSKDQLSGINNIAFSN
RSV F, N67I, S215P, D486N, and 357K and 371Y (SEQ ID NO: 21)
MELLILKANAITTILTAVTFCFASGQNITEEFYQSTCSAVSKGYLSALRTGWYTSVITIE LSNIKEIKCNGTDAKVKLIKQELDKYKNAVTELQLLMQSTPATNNRARRELPRFMNY TLNNAKKTNVTLSKKRKRRFLGFLLGVGSAIASGVAVSKVLHLEGEVNKIKSALLST NKAVVSLSNGVSVLTSKVLDLKNYIDKQLLPIVNKQSCSIPNIETVIEFQQKNNRLLEI TREFSVNAGVTTPVSTYMLTNSELLSLINDMPITNDQKKLMSNNVQIVRQQSYSIMSII KEEVLAYVVQLPLYGVIDTPCWKLHTSPLCTTNTKEGSNICLTRTDRGWYCDNAGS VSFFPQAEKCKVQSNRVFCDTMYSLTLPSEVNLCNVDIFNPKYDCKIMTSKTDVSSS VITSLGAIVSCYGKTKCTASNKNRGIIKTFSNGCDYVSNKGVDTVSVGNTLYYVNKQ EGKSLYVKGEPIINFYDPLVFPSNEFDASISQVNEKINQSLAFIRKSDELLSAIGGYIPEA PRDGQAYVRKDGEWVLLSTFL
CR9501 heavy chain (SEQ ID NO: 16):
QVQLVQSGPGLVKPSQTLALTCNVSGASINSDNYYWTWIRQRPGGGLEWIGHISYTG NTYYTPSLKSRLSMSLETSQSQFSLRLTSVTAADSAVYFCAACGAYVLISNCGWFDS WGQGTQVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGAL TSGVHTFPAVLQSSGLYSLSSWTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSC
CR9501 light chain (SEQ ID NO: 17):
EIVMTQSPSSLSASIGDRVTITCQASQDISTYLNWYQQKPGQAPRLLIYGASNLETGVP SRFTGSGYGTDFSVTISSLQPEDIATYYCQQYQYLPYTFAPGTKVEIKRTVAAPSVFIF 5 PPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYS
LS STLTLSKAD YEKHKVYACE VTHQGLS SP VTKSFNRGEC
CR9502 heavy chain (SEQ ID NO: 18):
EVQLLQSGAELKKPGASVKISCKTSGFTFSGHTIAWVRQAPGQGLEWMGWVSTNNG 10 NTEYAQKIQGRVTMTMDTSTSTVYMELRSLTSDDTAVYFCAREWLVMGGFAFDHW GQGTLLTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTS GVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKRVEPKSC
CR9502 light chain (SEQ ID NO: 19):
QSVLTQASSVSVAPGQTARITCGANNIGSQNVHWYQQKPGQAPVLVVYDDRDRPSG IPDRFSGSNSGNTATLTISRVEAGDEADYYCQVWDSSRDQAVIFGGGTKLTVLGQPK AAPSVTLFPPSSEELQANKATLVCLISDFYPGAVTVAWKADSSPVKAGVETTTPSKQS NNKYAASSYLSLTPEQWKSHRSYSCQVTHEGSTVEKTIAPTECS

Claims (17)

  1. Claims
    1. A recombinant pre-fusion respiratory syncitial virus (RSV) Fusion (F) protein comprising a mutation of the amino acid residue on position 215, in combination with a mutation of the amino acids on position 357 and a mutation of the amino acid on position 371.
  2. 2. Pre-fusion RSV F protein according to claim 1, comprising a mutation of the amino acid S on position 215 into P (S215P), a mutation of the amino acid T on position 357 into K (T357K) and a mutation of the amino acid N on position 371 into Y (N371Y) .
  3. 3. Pre-fusion RSV F protein according to claim 1 or 2, further comprising at least one mutation selected from the group consisting of:
    (a) a mutation of the amino acid residue on position 67; and (b) a mutation of the amino acid residue on position 486.
  4. 4. Pre-fusion RSV F protein according to claim 3, wherein the at least one further mutation is selected from the group consisting of:
    (a) a mutation of the amino acid residue N/T on position 67 into I; and (b) a mutation of the amino acid residue D on position 486 into N.
  5. 5. Pre-fusion RSV F protein according to any one of the preceding claims, wherein the protein comprises at least one epitope that is spécifie to the pre-fusion conformation F protein, wherein the at least one epitope is recognized by a pre-fusion spécifie monoclonal antibody, comprising a heavy chain CDR1 région of SEQ ID NO: 1, a heavy chain CDR2 région of SEQ ID NO: 2, a heavy chain CDR3 région of SEQ ID NO: 3 and a light chain CDR1 région of SEQ ID NO: 4, a light chain CDR2 région of
    SEQ ID NO: 5, and a light chain CDR3 région of SEQ ID NO: 6 and/or a pre-fusion spécifie monoclonal antibody, comprising a heavy chain CDR1 région of SEQ ID NO: 7, a heavy chain CDR2 région of SEQ ID NO: 8, a heavy chain CDR3 région of SEQ ID NO: 9 and a light chain CDR1 région of SEQ ID NO: 10, a light chain CDR2 région of SEQ ID NO: 67, and a light chain CDR3 région of SEQ ID NO: 11.
  6. 6. Pre-fusion RSV F protein according to any one of the preceding daims, wherein the protein is trimeric.
  7. 7. Pre-fusion RSV F protein according to any one of the preceding daims, comprising a truncated Fl domain and a heterologous trimerization domain linked to said truncated Fl domain.
  8. 8. Pre-fusion RSV F protein according to daim 7, wherein the heterologous trimerization domain comprises the amino acid sequence GYIPEAPRDGQAYVRKDGEWVLLSTFL (SEQ ID NO: 14).
  9. 9. Pre-fusion RSV F protein according to daim 7 or 8, wherein the trimerization domain is linked to amino acid residue 513 of the RSV F protein.
  10. 10. Pre-fusion RSV F protein according to any one of the preceding daims, wherein the RSV F protein comprises the amino acid sequence of SEQ ID NO: 21.
  11. 11. Nucleic acid molécule encoding a pre-fusion RSV F protein according to any one of the preceding daims 1-10.
  12. 12. Nucleic acid molécule according to claim 11, wherein the nucleic acid molécule has been codon-optimized for expression in mammalian cells.
  13. 13. Vector comprising a nucleic acid molécule according to claim 11 or claim 12.
  14. 14. Composition comprising a pre-fusion RSV F protein according to any of the daims 110, a nucleic acid molécule according to claim 11 or claim 12 and/or a vector according to claim 13.
  15. 15. Pre-fusion RSV F protein according to any of the daims 1-10, a nucleic acid molécule according to claim 11 or claim 12, and/or a vector according to claim 13 for use in inducing an immune response against RSV F protein.
  16. 16. Pre-fusion RSV F protein according to any of the daims 1-10, a nucleic acid molécule according to claim 11 or claim 12, and/or a vector according to claim 13 for use as a vaccine.
  17. 17. Pre-fusion RSV F protein according to any of the daims 1-10, a nucleic acid molécule according to claim 11 or claim 12, and/or a vector according to claim 13 for use in the prophylaxis and/or treatment of RSV infection.
OA1201800460 2016-05-30 2017-05-29 Stabilized pre-fusion RSV F proteins. OA19273A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
EP16172008.1 2016-05-30

Publications (1)

Publication Number Publication Date
OA19273A true OA19273A (en) 2020-06-05

Family

ID=

Similar Documents

Publication Publication Date Title
US11034731B2 (en) Stabilized soluble pre-fusion RSV F polypeptides
AU2021232702B2 (en) Stabilized pre-fusion RSV F proteins
US11229694B2 (en) Vaccine against RSV
US20220017574A1 (en) Stabilized pre-fusion rsv f proteins
WO2017207477A1 (en) Stabilized pre-fusion rsv f proteins
OA19273A (en) Stabilized pre-fusion RSV F proteins.
OA17598A (en) Stabilized soluble pre-fusion RSV F polypeptides